本执行摘要回顾了PDQ总结中有关结直肠癌(CRC)遗传学的主题内容,并显示了以下描述各主题相关证据的详细章节的超链接。
提示遗传对CRC产生影响的因素包括:(1)聚集性的CRC和/或息肉家族史;(2)CRC患者中的多种原发性癌;(3)家族中存在其他癌症与引起结直肠癌遗传风险的已知综合征一致,例如子宫内膜癌;(4)确诊CRC时年龄较小。尽管有两种综合征为常染色体隐性遗传(MUTYH相关的息肉和NTHL1),但遗传性CRC最常见的遗传方式是常染色体显性遗传。
至少可采用3个经验证的计算机模型预估罹患癌症的患者携带与Lynch综合征(最常见的遗传性CRC综合征)相关的错配修复(MMR)基因致病性变异的可能性。其中包括MMRpro、MMRpredict和PREMM5(基因突变的PREdiction模型)预测模型。建议对PREMM5定量风险为2.5%或以上,或对MMRpro和MMRpredict定量风险为5%或以上的患者进行基因评估转诊和检测。
充分描述了两种遗传性CRC:(1)息肉病(包括由FAP基因致病性变异引起的家族性腺瘤性息肉病[FAP]和衰减型FAP(AFAP);以及由MUTYH基因致病性变异引起的MUTYH相关息肉病);(2)由DNA MMR基因的种系致病性变异引起的Lynch综合征(常称为遗传性非息肉病性结直肠癌)。其他CRC综合征及其相关基因包括寡聚症(POLE,POLD1)、NTHL1、幼年性息肉综合征、Cowden综合征(PTEN)和Peutz-Jeghers综合征(STK11)。其中一些综合征也与结肠外肿瘤和其他表现有关。锯齿状息肉病综合征表现为出现增生性息肉,可能具有家族性成分,但遗传基础尚未可知。其中一些综合症的自然病史描述仍在进行中。表现为家族聚集性的许多其它类型CRC和/或腺瘤,但与已确定的遗传性综合征无明显关系,统称为家族性CRC。另外,大多数在50岁之前确诊CRC且无癌症家族史的人员未出现与遗传性癌症综合征相关的致病性变异。
低外显率易感等位基因
通过分子诊断肿瘤检测评估MMR缺陷,对所有最新确诊CRC患者进行Lynch综合征评估,已成为许多中心的标准治疗。推荐采用一种普遍的筛查方法进行肿瘤检测,其中对所有CRC病例进行评估,不考虑确诊时年龄或是否符合Lynch综合征的现有临床标准。据报道,可采用一种更具成本效益的方法对所有≤70岁CRC患者以及符合修订版Bethesda指导原则的老年患者进行检测,以确认是否患有Lynch综合征。一般情况下,在免疫组织化学检测Lynch综合征相关的MMR蛋白表达水平或微卫星不稳定性(MSI)检测、BRAF检测和MLH1超甲基化分析后进行肿瘤评估。
对遗传性CRC综合征患者常规进行结肠镜检查,从而进行CRC筛查和监测,且该检查可改善生存结局。例如,每1-2年使用结肠镜检查对Lynch综合征患者进行监测,一项为期3年的研究表明,该方法可降低CRC发生率和死亡率。结肠外监测也是确证某些遗传性CRC综合征的重要方法,取决于与该综合征相关的其他癌症。例如,已证实对FAP患者的十二指肠进行常规内窥镜检查监测可延长生存期。
研究也已证明预防性手术(结肠切除术)可以延长FAP患者的生存期。何时实施这类降低患癌风险的手术,如何确定手术范围,通常取决于息肉的数量、体积、组织学和症状。与进行CRC分段切除术的患者相比,患有Lynch综合征并确诊为CRC的患者行扩大切除术可降低异时性CRC的发生率以及减少行其他结直肠肿瘤术的次数。进行手术前,必须考虑患者的年龄、合并症、肿瘤的临床分期、括约肌功能以及患者的意愿。
也有研究探讨了化学预防剂在FAP和Lynch综合征治疗中的有效性。在FAP患者中,塞来昔布和舒林酸可减小息肉体积并减少相应的数量。一项双盲、随机、对照试验,评估舒林酸+表皮生长因子受体抑制剂厄洛替尼对比安慰剂在十二指肠息肉、FAP或AFAP患者中的疗效,结果表明厄洛替尼可抑制FAP患者中的十二指肠息肉。一项正在进行的临床试验将确定单独使用较低剂量的厄洛替尼是否会显著减轻十二指肠息肉负担。一项大型随机临床试验显示,使用阿司匹林(600 mg,每日一次)可对Lynch综合征患者的癌症发生率产生预防性作用;一项正在进行的研究中正在检测较低剂量的使用效果。
已在MMR缺陷型肿瘤(包括与Lynch综合征相关的肿瘤)中评估了激活免疫系统的新疗法。 MMR缺陷型肿瘤中密集的免疫浸润和富含细胞因子的环境可能改善临床结局。 导致介导肿瘤诱导的免疫抑制的关键途径是程序性细胞死亡1(PD-1)介导的检查点途径。 两项使用抗PD-1免疫检查点抑制剂(帕博利珠单抗和纳武利尤单抗)的II期研究显示具有良好的结局,包括无进展生存期、影像反应率,以及MMR缺乏的转移性CRC和既往接受过细胞毒性化疗后出现进展的MSI的疾病控制率。帕博利珠单抗已在MMR缺乏和MSI的其他非结直肠癌中显示出相似的获益,但在微卫星稳定的肿瘤中却未显示类似的获益。
社会心理因素可影响有关遗传性癌症风险和风险管理策略的基因检测决策。在研究中,对Lynch综合征和FAP进行遗传咨询和检测纳入量的方法差异很大。与Lynch综合症家族的遗传咨询和基因检测相关的因素包括生育子女、受累亲戚的数量、感知患CRC的风险以及想到患CRC的频率。心理研究表明,在对携带者和非携带者的Lynch综合症进行基因检测后,尤其是从长期来看,其困扰程度很低。但是,其他研究表明,在对FAP进行基因检测后,可能增加困扰。结果公布后一年内,MMR致病性变异携带者的结肠和妇科癌症筛查率得以增加或维持,而非携带者筛查率下降。后者的结果可以预期,因为针对未受累患者的筛查建议是适用于普通人群的筛查建议。评估FAP患者生活质量变量的研究显示正常范围的结果。但是,这些研究表明,针对FAP的降低患癌风险的手术可能会对部分受累的人群的生活质量产生负面影响。 患者在与家人沟通交流遗传性CRC风险时,过程十分复杂;性别、年龄和相关程度是影响此信息交流的一些因素。目前在研的研究旨在充分理解并解决高危家庭的社会心理和行为问题。
This executive summary reviews the topics covered in the PDQ summary on the genetics of colorectal cancer (CRC), with hyperlinks to detailed sections below that describe the evidence on each topic.
Factors suggestive of a genetic contribution to CRC include the following: (1) a strong family history of CRC and/or polyps; (2) multiple primary cancers in a patient with CRC; (3) the existence of other cancers within the kindred consistent with known syndromes causing an inherited risk of CRC, such as endometrial cancer; and (4) early age at diagnosis of CRC. Hereditary CRC is most commonly inherited in an autosomal dominant pattern, although two syndromes are inherited in an autosomal recessive pattern (MUTYH-associated polyposis and NTHL1).
At least three validated computer models are available to estimate the probability that an individual affected with cancer carries a pathogenic variant in a mismatch repair (MMR) gene associated with Lynch syndrome, the most common inherited CRC syndrome. These include the MMRpro, MMRpredict, and PREMM5 (PREdiction Model for gene Mutations) prediction models. Individuals with a quantified risk of 2.5% or greater on PREMM5 or 5% or greater on MMRpro and MMRpredict are recommended for genetic evaluation referral and testing.
Hereditary CRC has two well-described forms: (1) polyposis (including familial adenomatous polyposis [FAP] and attenuated FAP (AFAP), which are caused by pathogenic variants in the gene; and MUTYH-associated polyposis, which is caused by pathogenic variants in the MUTYH gene); and (2) Lynch syndrome (often referred to as hereditary nonpolyposis colorectal cancer), which is caused by germline pathogenic variants in DNA MMR genes (, , , and ) and . Other CRC syndromes and their associated genes include oligopolyposis (POLE, POLD1), NTHL1, juvenile polyposis syndrome (, ), Cowden syndrome (PTEN), and Peutz-Jeghers syndrome (STK11). Many of these syndromes are also associated with extracolonic cancers and other manifestations. Serrated polyposis syndrome, which is characterized by the appearance of hyperplastic polyps, appears to have a familial component, but the genetic basis remains unknown. The natural history of some of these syndromes is still being described. Many other families exhibit aggregation of CRC and/or adenomas, but with no apparent association with an identifiable hereditary syndrome, and are known collectively as familial CRC. In addition, most individuals with CRC diagnosed before age 50 years and without a family history of cancer do not have a pathogenic variant associated with an inherited cancer syndrome.
Genome-wide searches are showing promise in identifying common, low-penetrance susceptibility alleles for many complex diseases, including CRCs, but the clinical utility of these findings remains uncertain.
It is becoming the standard of care at many centers that all individuals with newly diagnosed CRC are evaluated for Lynch syndrome through molecular diagnostic tumor testing assessing MMR deficiency. A universal screening approach to tumor testing is supported, in which all CRC cases are evaluated regardless of age at diagnosis or fulfillment of existing clinical criteria for Lynch syndrome. A more cost-effective approach has been reported whereby all patients aged 70 years or younger with CRC and older patients who meet the revised Bethesda guidelines are tested for Lynch syndrome. Tumor evaluation often begins with immunohistochemistry testing for the expression of the MMR proteins associated with Lynch syndrome or microsatellite instability (MSI) testing, BRAF testing, and MLH1 hypermethylation analyses.
Colonoscopy for CRC screening and surveillance is commonly performed in individuals with hereditary CRC syndromes and has been associated with improved survival outcomes. For example, surveillance of Lynch syndrome patients with colonoscopy every 1 to 2 years, and in one study up to 3 years, has been shown to reduce CRC incidence and mortality. Extracolonic surveillance is also a mainstay for some hereditary CRC syndromes depending on the other cancers associated with the syndrome. For example, regular endoscopic surveillance of the duodenum in FAP patients has been shown to improve survival.
Prophylactic surgery (colectomy) has also been shown to improve survival in patients with FAP. The timing and extent of risk-reducing surgery usually depends on the number of polyps, their size, histology, and symptomatology. For patients with Lynch syndrome and a diagnosis of CRC, extended resection is associated with fewer metachronous CRCs and additional surgical procedures for colorectal neoplasia than in patients who undergo segmental resection for CRC. The surgical decision must take into account the age of the patient, comorbidities, clinical stage of the tumor, sphincter function, and the patient’s wishes.
Chemopreventive agents have also been studied in the management of FAP and Lynch syndrome. In FAP patients, celecoxib and sulindac have been associated with a decrease in polyp size and number. A double-blind, randomized, controlled trial evaluating the efficacy of sulindac plus an epidermal growth factor receptor inhibitor, erlotinib, versus placebo in FAP or AFAP patients with duodenal polyps suggested that erlotinib has the potential to inhibit duodenal polyps in FAP patients. An ongoing trial will determine whether lower doses of erlotinib alone will significantly reduce duodenal polyp burden. Aspirin use (600 mg daily) was shown to have a preventive effect on cancer incidence in Lynch syndrome patients in a large randomized trial; lower doses are being examined in an ongoing study.
Novel therapies that stimulate the immune system have been evaluated in MMR-deficient tumors, including those related to Lynch syndrome. The dense immune infiltration and cytokine-rich environment in MMR-deficient tumors may improve clinical outcomes. A critical pathway responsible for mediating tumor-induced immune suppression is the programmed cell death-1 (PD-1)–mediated checkpoint pathway. Two phase 2 studies using anti–PD-1 immune checkpoint inhibitors (pembrolizumab and nivolumab) demonstrated favorable outcomes, including progression-free survival, radiographic response rates, and disease control rates in metastatic CRC with MMR deficiency and MSI that had progressed on prior cytotoxic chemotherapy. Pembrolizumab has shown similar benefit in other noncolorectal cancers with MMR deficiency and MSI, but not in tumors that are microsatellite stable.
Psychosocial factors influence decisions about genetic testing for inherited cancer risk and risk-management strategies. Uptake of genetic counseling and genetic testing for Lynch syndrome and FAP varies widely across studies. Factors that have been associated with genetic counseling and testing uptake in Lynch syndrome families include having children, the number of affected relatives, perceived risk of developing CRC, and frequency of thoughts about CRC. Psychological studies have shown low levels of distress, particularly in the long term, after genetic testing for Lynch syndrome in both carriers and noncarriers. However, other studies have demonstrated the possibility of increased distress following genetic testing for FAP. Colon and gynecologic cancer screening rates have been shown to increase or be maintained among carriers of MMR pathogenic variants within the year after disclosure of results, while screening rates decrease among noncarriers. The latter is expected as the screening recommendations for unaffected individuals are those that apply to the general population. Studies measuring quality-of-life variables in FAP patients show normal-range results; however, these studies suggest that risk-reducing surgery for FAP may have negative quality-of-life effects for at least some proportion of those affected. Patients' communication with their family members about an inherited risk of CRC 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.
结直肠癌(CRC)在男性和女性最常见肿瘤中均排第三位。
据估计,2020年美国新发病例数和死亡病例数分别为:
约75%的CRC患者散发性,无明显证据表明具有遗传特征。 其余10%-30%的患者有CRC家族史,提示有遗传因素、家族成员之间有共同暴露或共同的危险因素,或两者兼而有之。
在一些易患结肠癌的家庭中,高外显率基因的致病性变异已被确定为遗传性癌症风险的原因。据估计,这些仅占CRC病例总数的5%-6%。
此外,较低的外显率基因中的致病性变异可能会导致家族性结肠癌风险。 在这种情况下,基因-基因和基因-环境的相互作用可能导致CRC的发展。
(如需了解散发性CRC的更多信息,请参见PDQ总结:结直肠癌筛查、结直肠癌预防、结肠癌治疗和直肠癌治疗。)
结直肠肿瘤的表现范围很广,从良性肿瘤到浸润性癌,并且主要是上皮来源的肿瘤(即,腺瘤或腺癌)。
任何息肉转化为癌症都要经过从腺瘤到癌这个过程。 传统上非肿瘤性息肉包括增生性、幼年性、错构瘤性、炎症性和淋巴样性息肉。但是,在某些情况下,错构瘤和幼年性息肉会进展成癌。
然而,研究证实,幼年性息肉病综合征和Peutz-Jeghers综合征患者出现结肠癌的风险很高,尽管与这些综合征相关的非腺瘤性息肉曾一度被认为是非肿瘤性的
流行病学研究表明,个人结肠腺瘤病史可增加患结肠癌的风险。
此观察结果的两种补充解释如下所示:
超过95%的CRC为癌,其中约95%为腺癌。 众所周知,腺瘤性息肉是可能经历恶性转化的良性肿瘤。 将其分为三种组织学类型,其恶性潜能不断增加:管状、管状绒毛状、绒毛状, 通常认为腺癌是由腺瘤引起,
基于以下重要观察结果:
腺瘤的以下三个特征与转化为癌的可能性高度相关:
另外,切除腺瘤性息肉可降低CRC发生率。
尽管大多数腺瘤为息肉样,但扁平和凹陷性病灶的发生率高于以往人们公认的结果。 体积大、扁平和凹陷的病灶更可能为严重的不典型增生,尽管这仍有待明确证明。 可能需要专门的技术来识别、活检和清除此类病灶。
有关CRC家族史的最早研究是针对犹他州家庭。据报告,因CRC死亡患者的一级亲属(FDR)中死于CRC的比例(3.9%)高于性别匹配和年龄匹配的对照组(1.2%)。
此差异已经在众多研究中得到了证实,这些研究一致地发现,受累病例的FDR自身患CRC的风险增加了两至三倍。 尽管有各种研究设计(病例对照,队列研究)、抽样框架、样本量、数据验证方法、分析方法以及研究起源的国家,但风险程度保持一致。
已报道了家族性CRC风险的系统评价和荟萃分析。
在纳入分析的24项研究中,除一项研究外,所有研究的报告结果均显示,如果FDR受累,则CRC风险增加。如果FDR受累,则汇总研究中CRC的相对风险(RR)为2.25(95%置信区间[CI],2.00–2.53)。 在11项研究中的8项中,如果原发病例发生在结肠,则患癌风险略高于原发病例发生直肠。汇总分析显示,结肠癌和直肠癌患者亲属的RR分别为2.42(95%CI,2.20-1.65)和1.89(95%CI,1.62-2.21)。该分析未显示基于肿瘤位置(右侧与左侧)的结肠癌RR的差异。
受影响家庭成员的数量和确诊时的年龄与CRC风险相关。在报告一位以上FDR患有CRC的研究中,RR为3.76(95%CI,2.56-5.51)。与原发病例确诊年龄为45-59岁(RR,2.25;95% CI,1.85–2.72)以及60岁或60岁以上(RR为1.82;95%CI,1.47–2.25)相比,确诊年龄45岁以下(RR,3.87;95%CI,2.40–6.22)的家庭成员观察到的RR最高。在该荟萃分析中,分析了FDR中与腺瘤相关的CRC家族风险。汇总分析表明,在伴腺瘤的FDR患者中CRC的RR为1.99(95%CI,1.55-2.55)。
这一结果得到了证实。
其他研究报告称,腺瘤诊断时的年龄可能影响CRC风险,而腺瘤诊断时年龄越小,RR越高。
与任何荟萃分析一样,可能存在会影响分析结果的潜在偏倚,包括研究的不完全和非随机确定;发表文献偏倚;以及设计、目标人群和对照选择相关研究之间的异质性。该研究进一步证明,家族性CRC风险、CRC和腺瘤确诊时年龄以及受累家庭成员的多样性之间存在显著的关系。
家族史 | CRC的相对风险 | 79岁时结直肠癌的绝对风险 |
---|---|---|
无家族史 | 1 | 4a |
一例FDR患CRC | 2.3(95% CI,2.0–2.5) | 9b |
多例FDR患CRC | 4.3(95% CI,3.0-6.1) | 16b |
一例受累FDR在45岁前确诊为CRC | 3.9(95% CI,2.4-6.2) | 15b |
一例FDR患结直肠腺瘤 | 2.0(95% CI,1.6-2.6) | 8b |
CI =置信区间;FDR =一级亲属。 | ||
a来自监测、流行病学和最终结果数据库的数据 | ||
b使用患CRC的相对风险以及79岁时发生CRC的绝对风险 计算有受累亲属的个体发生CRC的绝对风险。 |
如果家族史中有两个或两个以上患CRC的亲属,遗传综合征的发生率大大提高。 评估的第一步是详细审查家族史,以确定受累的亲属的数量、彼此之间的关系、确诊CRC时的年龄、是否存在多个原发性CRC、以及是否患有与其他任何与遗传性CRC综合征一致的癌症(例如子宫内膜癌)。 (有关更多信息,请参阅该总结的“重大遗传综合征”章节。) 目前可使用计算机模型来估计发生CRC的概率。
这些模型有助于为处于患癌中等风险和高风险的患者提供遗传咨询。此外,至少3个经验证的模型可用于预测错配修复(MMR)基因中携带致病性变异的可能性。
在各种家庭风险情况下发生CRC病例的比例请见图1。
已经发现了几种与CRC风险相关的基因。这些基因在本总结的“结肠癌基因”章节中进行了详细说明。 已知几乎所有导致CRC易感性的致病性变异均以常染色体显性方式遗传。
已发现常染色体隐性遗传的一个实例,即MUTYH相关的息肉病(MAP)。(更多信息请参见本总结的“MUTYH相关的息肉病[MAP]”章节。) 因此,表明癌症易感性的常染色体显性遗传的家族特征是高风险和癌症易感性致病变体可能存在的重要指标。其中包括:
遗传性CRC的两个最常见病因是由APC基因的种系致病性变异引起的FAP(包括AFAP)
以及由DNA MMR基因的种系致病性变异引起的Lynch综合征(前称遗传性非息肉病结直肠癌[HNPCC])。
(图2描绘了经典的Lynch综合征家庭,重点强调上述遗传性CRC的一些指标。)许多其他家系出现聚集性CRC和/或腺瘤,但与已确定的遗传性综合征无明显关系,统称为家族性CRC。
美国医学遗传学学院和国家遗传咨询师协会已制定了指南,以帮助确定适合转诊至癌症遗传咨询服务的患者。
在确定此类人员后,将考虑针对患者选择治疗策略。 (如需了解有关个体综合症的具体干预措施的信息,请参见该总结的“重大遗传综合征”章节。)
目前,在确定对CRC的遗传易感性时,不建议将致病性变异试验作为一般人群的筛查手段。 由于CRC相关基因中致病性变异的罕见性以及当前检测策略的灵敏度有限,一般人群检测可能产生误导且无成本效益。
多个组织代表各医学专业团体和学会,提供了有关FAP和Lynch综合征监测的详细建议。这些组织包括:
专家推荐的证据是基于说明或指南。在许多情况下,这些指南反映了专家的意见,这些意见基于的研究很少是随机前瞻性试验。
在使用家族史评估临床实践中的个体风险以及确定适合癌症研究的家族时,必须考虑家族史数据的准确性和完整性。报告的家族史可能是数据错误,或个人可能不知道有患癌亲属。
结肠镜检查的使用率增加可降低家族史中CRC发生率,并提高癌前结肠息肉的检出率。与了解癌症家族史的人员相比,患者了解息肉家族史(即亲属中息肉类型和息肉总数)的可能性偏低。 此外,家庭规模小及提前死亡可能掩盖家族史的信息量。 同样,由于不完全外显,某些人员可能对CRC具有遗传易感性,但不会发展成癌症,在家族树中表现为隔代遗传。
已证明患者自报的结肠癌家族史的准确性较好,但并非最佳。应该通过获取病历核实患者的报告(如可能),尤其是对于可能与确定Lynch综合征风险有关并且某些患者报告的可靠性较差的生殖道肿瘤 。 (如需了解更多信息,请参见PDQ“癌症遗传学风险评估和咨询”总结中家族史准确性章节。)
可采用多种方法评估最新确诊为CRC的患者,这些患者可能被怀疑也可能不被怀疑患有癌症遗传综合征。临床医生可能根据家族史和体格检查怀疑潜在遗传性倾向,并且可以通过基因检测证实这些怀疑。美国医学遗传学和基因组学学会已发布了用于评估疑似结肠癌易感性综合征患者的指南。
该指南旨在确定其临床特征值得转诊进行遗传学咨询的人员。如果一个人有多个息肉(> 20),根据组织学,特定的基因指导检测可能是有用的诊断工具。同样,如果患者的临床表现疑似Lynch综合征,可以针对该综合征进行种系基因检测。但是,当临床情况不明确时,诊断将更具挑战性。当前,针对Lynch综合征的肿瘤筛查是普遍认可的方法。然而,越来越多地,表现体细胞突变的基因组套被用于各种临床决策。
在许多情况下,优先风险评估测试(基于多种因素来模拟风险,例如癌症发病年龄和家族肿瘤谱)可以作为适当的替代方法。此类风险模型的应用确实可以预测使用多基因(组合)检测的有效性。但是,其确切作用还有待确定。
我们对CRC分子发病机理的最初了解大部分来自罕见的遗传性CRC综合征,并揭示了CRC在分子和临床上的异质性。众所周知,大多数CRC均由腺瘤转变而成。从正常上皮到腺瘤再到癌的转变与获得性分子事件有关。
目前,根据相似的分子遗传学特征,CRC可分为三类,提示不同的肿瘤发生途径:染色体不稳定(CIN)、微卫星不稳定(MSI)和CpG岛甲基化表型(CIMP)。对结直肠肿瘤发生的分子遗传途径的理解仍在不断发展,并且每个新的理解水平都是在先前知识水平的背景下发生的。此外,这些途径来自结直肠息肉和肿瘤的重要临床和组织学异质性。因此,以下介绍充分说明了我们目前对结直肠肿瘤发生的理解以及时间顺序演变。
大多数CRC通过CIN途径发展。CIN癌症的关键变化包括染色体数目的广泛变化(非整倍性)和在染色体部分分子水平上经常可检测到缺失(杂合性缺失),例如5q、18q和17p;KRAS致癌基因的致病性变异。与这些染色体缺失有关的重要基因是APC(5q)、DCC / MADH2 / MADH4(18q)和TP53(17p)。
这些染色体缺失表明在分子和染色体水平下遗传不稳定。
结直肠肿瘤进展途径中最早发生且最常见的事件之一是APC基因的缺失或致病性变异失活。首次证明在FAP中APC的致病性变异-失活对结直肠癌的产生是至关重要的,而FAP是一种遗传性CRC综合征,其中受累的患者携带种系APC突变,从而导致其功能丧失以及结直肠息肉和癌症的发病率急剧增加。DNA损伤-修复基因的获得性或遗传性致病性变异,例如碱基切除修复、核苷酸切除修复、双链修复和MMR,也在结直肠上皮细胞易感致病性变异中发挥作用。
此后不久,就发现了一部分CRC(10%–15%),缺乏染色体不稳定性的证据,但在微卫星重复序列中表现出畸变,
这是Lynch综合征患者的肿瘤特征。
随后发现,MLH1启动子的高甲基化是导致MSI散发性CRC的原因。 在Lynch综合征患者中发现了DNA MMR基因的种系变异,其CRC时常显示MSI。 因此,提出了微卫星不稳定性路径(MSI,有时称为MIN)。
MSI癌症的关键特征是它们具有完整的染色体互补,并且由于DNA MMR系统的缺陷,更容易获得重要且通常独特的癌症相关基因的致病变异。这些类型的癌症可以通过在整个基因组中正常发生的DNA重复单元改变而在分子水平上检测到,这种重复称为DNA微卫星。
与微卫星稳定的肿瘤相比,微卫星不稳定的肿瘤中腺瘤向癌进展的速率较快。
其基础是近期结肠镜检查正常的患者中反复出现间隔癌的报道。在锯齿状途径中(见下文)可以观察到对这一点的进一步支持,同时也观察到间隔癌发生率较高。
在表现出MSI、肿瘤内T淋巴细胞浸润/克罗恩样反应等特征的肿瘤中可以观察到特征性的组织学变化,例如粘蛋白产生增加,从而区分通过这一途径形成的结直肠肿瘤
从遗传性结直肠癌综合征的研究中获得的信息,为发现无种系异常人群中介导肿瘤发生和发展的分子事件提供了重要线索。结直肠肿瘤进展途径(MSI和CIN)中最早发生的事件之一是APC基因产物功能丧失。
从20世纪80年代开始,研究开始报告患有增生性息肉综合征(HPS)(现称为锯齿状息肉综合征(SPS))的患者出现CRC的风险增加。
仅有少量的SPS似乎是家族性的,但迄今为止,在这些家族中尚未发现常见的种系变异。对SPS患者和对照中发现的增生性息肉(HPs)的比较显示,SPS息肉在组织学上是不同的,并且与先前描述的锯齿状腺瘤,具有HPs特征的息肉和腺瘤性息肉(AP)相似。
从而观察到,这些无柄锯齿状腺瘤(SSA)往往发生在右结肠,其中通常较大且无柄,并表现出隐窝基底细胞的增殖、扩张和锯齿增加、内分泌细胞减少以及缺乏发育不良。
从组织学特征上,可以将锯齿状息肉分为以下亚型:传统的锯齿状腺瘤(TSA)、混合锯齿状息肉(MP),以及最近的无蒂锯齿状腺瘤/无柄锯齿状息肉(SSA / SSP)。
TSA的特征为形态突出、异位隐窝形成(提示骨形态发生蛋白信号传导不足)以及绒毛状和不典型增生的组织病理学。
TSA不仅是不典型增生的SSA,并且无证据表明SSA是TSA的前体。 MP的特征与HP、SSA和TSA重叠。
在结肠镜检查中,锯齿状大息肉与晚期结直肠肿瘤的发生密切相关,而左侧HP则不然。 SSA一词已引起临床医生的关注,因为这些特征通常缺乏核异型性,这是腺瘤的传统标志,但由于其他架构特性而被称为腺瘤。分子特征表示癌症风险增加,这可以支持SSA的分类。
虽然Lynch综合征患者的AP可能表现出MSI,但散发性腺瘤罕见。但是,具有不典型增生的锯齿状息肉会表现出MSI,且MLH1启动子发生超甲基化。 锯齿状大息肉(> 1cm)的癌变风险比常规增生性息肉更大,当发展为癌症时,其特征性表现为MSI。
在一项切除后伴恶性病灶的有关锯齿状息肉的回顾性研究中,发现所有息肉均起源于右结肠,且均为SSA。
恶性肿瘤病灶为MSI,表现出MLH1免疫反应缺失,表明SSA与散发性MSI结肠癌之间存在一定关系。
散发性CRC中的MSI是由于MLH1启动子的超甲基化导致其表达被抑制。随着其他抑癌基因的启动子区域通过超甲基化被“沉默”,结直肠癌的癌症基因组研究随之展开。结果显示大约50%的CRC中被评估基因的超甲基化模式是一致的。
大量未经选择的CRC患者的研究表明,少数CRC(20%–30%)表现出CIMP,定义为MINT1、MINT2、MINT31、CDKN2A(p16)和MLH1中两个或多个CpG岛超甲基化。
CIMP一词创造的目的是对这些具有共同临床特征的癌症进行分类。 早期区分CIMP阳性和CIMP阴性CRC的尝试均未成功。
然而,随后的研究使用了CRC中重甲基化基因的无偏倚层序聚类分析和基于人群的研究设计,成功地确定了支持CIMP途径的独特临床和分子特征。
与微卫星稳定的CRC(24.4%;P < 0.0001)相比,CIMP高的CRC表达MSI的可能性更高(82.1%;P < 0.0001)。
一项研究显示,与以下因素相关的微卫星稳定且CIMP高(上述> 2个CIMP标志物)的结直肠肿瘤显著多于CIMP低(上述<2个CIMP标志物)的结直肠肿瘤:BRAF V600E变异、KRAS2变异、近端部位、AJCC分期较高、患者年龄较大、分化差以及黏液腺癌。
与以下因素相关的微卫星不稳定且CIMP高的结直肠肿瘤显著多于微卫星不稳定且CIMP低的肿瘤:BRAF V600E致病性变异、近端部位、患者年龄较大以及缺乏KRAS2致病性变异。
无论MSI如何,在CIMP高结直肠肿瘤中,BRAF V600E致病性变异的存在显著增加。
因此,不同于先前的研究,一旦排除不稳定的结直肠肿瘤,CIMP的生物学意义就会被质疑,
而本研究证实在微卫星稳定和微卫星不稳定的结直肠肿瘤中,CIMP确实与一些临床病理特征有关。
息肉的研究显示HPS患者中CIMP阳性息肉,最常见的是右侧SSA。
最近,发现BRAF致病性变异热点(V600E)在MSI结肠癌和锯齿状息肉中很常见。
Lynch综合征患者的CRC中不存在BRAF致病性变异,在散发性腺瘤性结肠息肉中罕见,但是存在于绝大多数锯齿状息肉中,尤其是SSA。
CIMP阳性通常见于微泡增生性息肉(MVHP),提示MVHP进展为SSA,然后进展为结肠癌。
CIMP CRC的特征以及在腺瘤-癌序列中MSI发生较晚的证据导致对先前结直肠肿瘤发生模型的修正,该模型包括两个途径: MSI(MIN)和CIN。 MSI和CIMP途径之间有很多重叠。CIMP途径的核心是携带BRAF致病性变异的锯齿状息肉。 CIN途径的特征是AP前体,其中绝大多数都携带APC致病性变异,这些变异发生在该途径的早期。
Colorectal cancer (CRC) is the third most commonly diagnosed cancer in both men and women.
Estimated new cases and deaths from CRC in 2020 in the United States:
About 75% of patients with CRC have sporadic disease with no apparent evidence of having inherited the disorder. The remaining 10% to 30% of patients have a family history of CRC that suggests a hereditary contribution, common exposures or shared risk factors among family members, or a combination of both.
Pathogenic variants in high-penetrance genes have been identified as the cause of inherited cancer risk in some colon cancer–prone families; these are estimated to account for only 5% to 6% of CRC cases overall.
In addition, pathogenic variants in lower penetrance genes may contribute to familial colon cancer risk. In such cases, gene-gene and gene-environment interactions may contribute to the development of CRC.
(Refer to the PDQ summaries on Colorectal Cancer Screening; Colorectal Cancer Prevention; Colon Cancer Treatment; and Rectal Cancer Treatment for more information about sporadic CRC.)
Colorectal tumors present with a broad spectrum of neoplasms, ranging from benign growths to invasive cancer, and are predominantly epithelial-derived tumors (i.e., adenomas or adenocarcinomas).
Transformation of any polyp into cancer goes through the adenoma-carcinoma sequence. Polyps that have traditionally been considered nonneoplastic include those of the hyperplastic, juvenile, hamartomatous, inflammatory, and lymphoid types. However, in certain circumstances, hamartomatous and juvenile polyps can progress into cancer.
Research, however, does suggest a substantial risk of colon cancer in individuals with juvenile polyposis syndrome and Peutz-Jeghers syndrome, although the nonadenomatous polyps associated with these syndromes have historically been viewed as nonneoplastic.
Epidemiologic studies have shown that a personal history of colon adenomas places one at an increased risk of developing colon cancer.
Two complementary interpretations of this observation are as follows:
More than 95% of CRCs are carcinomas, and about 95% of these are adenocarcinomas. It is well recognized that adenomatous polyps are benign tumors that may undergo malignant transformation. They have been classified into three histologic types, with increasing malignant potential: tubular, tubulovillous, and villous. Adenocarcinomas are generally considered to arise from adenomas,
based upon the following important observations:
The following three characteristics of adenomas are highly correlated with the potential to transform into cancer:
In addition, removal of adenomatous polyps is associated with reduced CRC incidence.
While most adenomas are polypoid, flat and depressed lesions may be more prevalent than previously recognized. Large, flat, and depressed lesions may be more likely to be severely dysplastic, although this remains to be clearly proven. Specialized techniques may be needed to identify, biopsy, and remove such lesions.
Some of the earliest studies of family history of CRC were those of Utah families that reported a higher percentage of deaths from CRC (3.9%) among the first-degree relatives (FDRs) of patients who had died from CRC than among sex-matched and age-matched controls (1.2%).
This difference has since been replicated in numerous studies that have consistently found that FDRs of affected cases are themselves at a twofold to threefold increased risk of CRC. Despite the various study designs (case-control, cohort), sampling frames, sample sizes, methods of data verification, analytic methods, and countries where the studies originated, the magnitude of risk is consistent.
A systematic review and meta-analysis of familial CRC risk has been reported.
Of 24 studies included in the analysis, all but one reported an increased risk of CRC if there was an affected FDR. The relative risk (RR) for CRC in the pooled study was 2.25 (95% confidence interval [CI], 2.00–2.53) if there was an affected FDR. In 8 of 11 studies, if the index cancer arose in the colon, the risk was slightly higher than if it arose in the rectum. The pooled analysis revealed an RR in relatives of colon and rectal cancer patients of 2.42 (95% CI, 2.20–2.65) and 1.89 (95% CI, 1.62–2.21), respectively. The analysis did not reveal a difference in RR for colon cancer based on location of the tumor (right side vs. left side).
The number of affected family members and age at cancer diagnosis correlated with the CRC risk. In studies reporting more than one FDR with CRC, the RR was 3.76 (95% CI, 2.56–5.51). The highest RR was observed when the index case was diagnosed in individuals younger than 45 years (RR, 3.87; 95% CI, 2.40–6.22) compared with family members of index cases diagnosed at ages 45 to 59 years (RR, 2.25; 95% CI, 1.85–2.72), and to family members of index cases diagnosed at age 60 years or older (RR, 1.82; 95% CI, 1.47–2.25). In this meta-analysis, the familial risk of CRC associated with adenoma in an FDR was analyzed. The pooled analysis demonstrated an RR for CRC of 1.99 (95% CI, 1.55–2.55) in individuals who had an FDR with an adenoma.
This finding has been corroborated.
Other studies have reported that age at diagnosis of the adenoma influences the CRC risk, with younger age at adenoma diagnosis associated with higher RR.
As with any meta-analysis, there could be potential biases that might affect the results of the analysis, including incomplete and nonrandom ascertainment of studies included; publication bias; and heterogeneity between studies relative to design, target populations, and control selection. This study is reinforcement that there are significant associations between familial CRC risk, age at diagnosis of both CRC and adenomas, and multiplicity of affected family members.
Family History | Relative Risk of CRC | Absolute Risk (%) of CRC by Age 79 y |
---|---|---|
No family history | 1 | 4a |
One FDR with CRC | 2.3 (95% CI, 2.0–2.5) | 9b |
More than one FDR with CRC | 4.3 (95% CI, 3.0–6.1) | 16b |
One affected FDR diagnosed with CRC before age 45 y | 3.9 (95% CI, 2.4–6.2) | 15b |
One FDR with colorectal adenoma | 2.0 (95% CI, 1.6–2.6) | 8b |
CI = confidence interval; FDR = first-degree relative. | ||
aData from the Surveillance, Epidemiology, and End Results database. | ||
bThe absolute risks of CRC for individuals with affected relatives was calculated using the relative risks for CRC and the absolute risk of CRC by age 79 yearsa. |
When the family history includes two or more relatives with CRC, the possibility of a genetic syndrome is increased substantially. The first step in this evaluation is a detailed review of the family history to determine the number of relatives affected, their relationship to each other, the age at which the CRC was diagnosed, the presence of multiple primary CRCs, and the presence of any other cancers (e.g., endometrial) consistent with an inherited CRC syndrome. (Refer to the Major Genetic Syndromes section of this summary for more information.) Computer models are now available to estimate the probability of developing CRC.
These models can be helpful in providing genetic counseling to individuals at average risk and high risk of developing cancer. In addition, at least three validated models are also available for predicting the probability of carrying a pathogenic variant in a mismatch repair (MMR) gene.
Figure 1 shows the proportion of CRC cases that arise in various family risk settings.
Several genes associated with CRC risk have been identified; these are described in detail in the Colon Cancer Genes section of this summary. Almost all pathogenic variants known to cause a predisposition to CRC are inherited in an autosomal dominant fashion.
One example of autosomal recessive inheritance, MUTYH-associated polyposis (MAP), has been identified. (Refer to the MUTYH-Associated Polyposis [MAP] section of this summary for more information.) Thus, the family characteristics that suggest autosomal dominant inheritance of cancer predisposition are important indicators of high risk and of the possible presence of a cancer-predisposing pathogenic variant. These include the following:
The two most common causes of hereditary CRC are FAP (including AFAP), due to germline pathogenic variants in the APC gene,
and Lynch syndrome (previously called hereditary nonpolyposis colorectal cancer [HNPCC]), which is caused by germline pathogenic variants in DNA MMR genes.
(Figure 2 depicts a classic family with Lynch syndrome, highlighting some of the indicators of hereditary CRC that are described above.) Many other families exhibit aggregation of CRC and/or adenomas, but with no apparent association with an identifiable hereditary syndrome, and are known collectively as familial CRC.
Guidelines have been developed by the American College of Medical Genetics and the National Society of Genetic Counselors to aid in the identification of patients appropriate for referral to a cancer genetic counseling service.
When such persons are identified, options tailored to the patient situation are considered. (Refer to the Major Genetic Syndromes section of this summary for information on specific interventions for individual syndromes.)
At this time, the use of pathogenic variant testing to identify genetic susceptibility to CRC is not recommended as a screening measure in the general population. The rarity of pathogenic variants in CRC-associated genes and the limited sensitivity of current testing strategies render general population testing potentially misleading and not cost-effective.
Rather detailed recommendations for surveillance in FAP and Lynch syndrome have been provided by several organizations representing various medical specialties and societies. These organizations include the following:
The evidence bases for recommendations are generally included within the statements or guidelines. In many instances, these guidelines reflect expert opinion resting on studies that are rarely randomized prospective trials.
The accuracy and completeness of family history data must be taken into account in using family history to assess individual risk in clinical practice, and in identifying families appropriate for cancer research. A reported family history may be erroneous, or a person may be unaware of relatives with cancer.
Increased use of colonoscopy may result in fewer CRCs and more precancerous colon polyps in a family history. Individuals are much less likely to know about their family history of polyps (i.e., type of polyps and total number of polyps in their relatives) than they are to know about their family history of cancer. In addition, small family sizes and premature deaths may limit how informative a family history may be. Also, due to incomplete penetrance, some persons may carry a genetic predisposition to CRC but do not develop cancer, giving the impression of skipped generations in a family tree.
Accuracy of patient-reported family history of colon cancer has been shown to be good, but it is not optimal. Patient report should be verified by obtaining medical records whenever possible, especially for reproductive tract cancers that may be relevant in identifying risk of Lynch syndrome and less reliably reported by some patients. (Refer to the Accuracy of the family history section in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for more information.)
Several approaches are available to evaluate a patient with newly diagnosed CRC who may or may not be suspected of having a cancer genetics syndrome. The clinician may suspect a potential inherited disposition based on the family history and physical exam, and genetic tests are available to confirm these suspicions. The American College of Medical Genetics and Genomics has published guidelines for evaluating patients with suspected colon cancer susceptibility syndromes.
The guidelines aim to identify individuals whose clinical features warrant referral for genetics consultation. If an individual has multiple polyps (>20), depending on the histology, specific gene-directed testing can be a useful diagnostic tool. Similarly, if a patient’s clinical presentation is suspicious for Lynch syndrome, germline genetic testing can be directed towards this syndrome. However, diagnosis is more challenging when the clinical picture is less clear. Currently, tumor screening for Lynch syndrome is the most commonly accepted approach. However, increasingly, panels characterizing somatic mutations in tumors are being utilized for a variety of clinical decisions.
A priori risk-assessment testing (which models risk based on a variety of factors, such as age at cancer onset and the spectrum of tumors in the family) may be an appropriate alternative in many cases. Application of such risk models does anticipate the use of multigene (panel) testing; however, their exact role remains to be established.
Much of our initial understanding of the molecular pathogenesis of CRC derived from rare hereditary CRC syndromes and revealed heterogeneity of CRC both molecularly and clinically. It is well accepted that most CRCs develop from adenomas. The transition from normal epithelium to adenoma to carcinoma is associated with acquired molecular events.
Presently, CRC can be separated into three categories based on similar molecular genetic features, suggesting divergent pathways of tumorigenesis: chromosomal instability (CIN), microsatellite instability (MSI), and CpG island methylator phenotype (CIMP). The understanding of the molecular genetic pathways of colorectal tumorigenesis is still evolving, and each new level of understanding has occurred in the context of the preceding level of knowledge. In addition, these pathways emerged from important clinical and histological heterogeneity of colorectal polyps and cancers. Thus, the introduction below captures the chronological evolution of our current understanding of colorectal tumorigenesis.
The majority of CRCs develop through the CIN pathway. Key changes in CIN cancers include widespread alterations in chromosome number (aneuploidy) and frequent detectable losses at the molecular level of portions of chromosomes (loss of heterozygosity), such as 5q, 18q, and 17p; and pathogenic variants of the KRAS oncogene. The important genes involved in these chromosome losses are APC (5q), DCC/MADH2/MADH4 (18q), and TP53 (17p).
These chromosomal losses are indicative of genetic instability at the molecular and chromosomal levels.
Among the earliest and most common events in the colorectal tumor progression pathway is loss or pathogenic variant–inactivation of the APC gene. Pathogenic variant–inactivation of APC was first shown to be important to CRC in FAP, a hereditary CRC syndrome in which affected individuals harbor germline APC alterations, resulting in its loss of function and a dramatically increased incidence of colorectal polyps and cancers. Acquired or inherited pathogenic variants of DNA damage-repair genes, for example, base excision repair, nucleotide excision repair, double stranded repair, and MMR, also play a role in predisposing colorectal epithelial cells to pathogenic variants.
Soon thereafter, a subset (10%–15%) of CRCs was identified that lacked evidence of chromosomal instability but exhibited aberrations in microsatellite repeat sequences,
a characteristic of tumors in patients with Lynch syndrome.
It was later found that hypermethylation of the MLH1 promoter is responsible for sporadic CRCs with MSI. Germline variants in DNA MMR genes were discovered in Lynch syndrome patients, whose CRCs frequently displayed MSI. Thus, the microsatellite instability pathway (MSI, sometimes referred to as MIN) was proposed.
The key characteristics of MSI cancers are that they have a largely intact chromosome complement and, as a result of defects in the DNA MMR system, more readily acquire pathogenic variants in important and often unique cancer-associated genes. These types of cancers are detectable at the molecular level by alterations in repeating units of DNA that occur normally throughout the genome, known as DNA microsatellites.
The rate of adenoma-to-carcinoma progression appears to be faster in microsatellite-unstable tumors than in microsatellite-stable tumors.
The foundation for this is the repeated reports of interval cancers in patients with recent, normal colonoscopy. Further support for this is seen in the serrated pathway (see below), in which high rates of interval cancer have also been observed.
Characteristic histologic changes, such as increased mucin production, can be seen in tumors that demonstrate MSI, intratumoral T lymphocyte infiltration/Crohn-like reaction, etc., distinguishing the colorectal tumors in this pathway.
The knowledge derived from the study of inherited CRC syndromes has provided important clues regarding the molecular events that mediate tumor initiation and tumor progression in people without germline abnormalities. Among the earliest events in the colorectal tumor progression pathway (both MSI and CIN) is loss of function of the APC gene product.
Beginning in the 1980s, studies began reporting an increased risk of CRC in patients with hyperplastic polyposis syndrome (HPS), now referred to as serrated polyposis syndrome (SPS).
Only a minority of SPS appear to be familial, but no common germline variant has been identified in these families to date. A comparison of the hyperplastic polyps (HPs) found in SPS patients and controls revealed that SPS polyps are histologically distinct and are similar to previously described serrated adenomas, polyps with features of HPs and adenomatous polyps (APs).
This led to observations that these sessile serrated adenomas (SSA) tend to occur in the right colon, where they are frequently large and sessile, and exhibit increased proliferation, dilation and serration of the crypt bases, decreased endocrine cells, and lack of dysplasia.
Further histological characterization of serrated polyps led to subtypes: traditional serrated adenomas (TSA), mixed serrated polyps (MP), and more recently, sessile serrated adenoma/sessile serrated polyp (SSA/SSP).
TSAs are characterized by a protuberant morphology, ectopic crypt formation (suggestive of deficient bone morphogenetic protein signaling), and villiform and dysplastic histopathology.
TSAs are not simply SSAs with dysplasia, and evidence that SSAs are precursors of TSAs is lacking. MPs have overlapping features of HPs, SSAs, and TSAs.
In colonoscopy screening studies, large serrated polyps were strongly and independently associated with the development of advanced colorectal neoplasms, while left-sided HPs were not. The term SSA has been a concern to clinicians as these characteristically lack nuclear atypia, the traditional hallmark of adenomas, but rather are termed adenomas due to other architectural features. The classification of SSA is supported by the knowledge that the molecular characteristics denote an increased cancer risk.
While APs in Lynch syndrome patients can exhibit MSI, sporadic adenomas rarely do. However, serrated polyps with dysplasia can exhibit MSI with hypermethylation of the MLH1 promoter. Large (>1 cm) serrated polyps carry greater cancer risk than do conventional hyperplastic polyps and, when developing into cancers, characteristically exhibit MSI.
In a review of resected serrated polyps with a malignant focus, all of the polyps originated in the right colon and were SSAs.
The malignant foci were MSI and demonstrated loss of MLH1 immunoreactivity, suggesting an association between SSAs and sporadic MSI colon cancers.
The MSI seen in sporadic CRCs is due to hypermethylation of the promoter of MLH1, which abrogates its expression. As promoter regions of other tumor suppressor genes were “silenced” through hypermethylation, cancer genome studies of CRC ensued. These showed a consistent pattern of hypermethylation in the evaluated genes in approximately 50% of CRCs.
Studies of larger numbers of unselected CRC patients show that a minority of CRCs (20%–30%) demonstrate CIMP, defined as hypermethylation of two or more of the CpG islands in MINT1, MINT2, MINT31, CDKN2A (p16), and MLH1.
The term CIMP was coined to classify these cancers, which shared clinical features. Early attempts to differentiate CIMP-positive and CIMP-negative CRCs were unsuccessful.
However, subsequent studies using unbiased hierarchical cluster analysis of heavily methylated genes in CRCs and a population-based study design successfully identified unique clinical and molecular characteristics supporting a CIMP pathway.
CIMP-high CRCs were much more likely (82.1%; P < .0001) to express MSI than were microsatellite-stable CRCs (24.4%; P < .0001).
In one study, microsatellite-stable, CIMP-high (>2 CIMP markers mentioned above) colorectal tumors were significantly more associated with BRAF V600E variants, KRAS2 variants, proximal site, higher American Joint Committee on Cancer stage, older patient age, poor differentiation, and mucinous histology than were CIMP-low (<2 CIMP markers mentioned above) colorectal tumors.
Microsatellite-unstable, CIMP-high colorectal tumors were significantly more associated with BRAF V600E pathogenic variants, proximal site, older patient age, and absence of KRAS2 pathogenic variants than were microsatellite unstable, CIMP-low tumors.
There was a significantly greater presence of BRAF V600E pathogenic variants in CIMP-high colorectal tumors regardless of MSI.
Thus, unlike a previous study that questioned the biological significance of CIMP once unstable colorectal tumors were excluded,
this study demonstrated several clinicopathologic variables were indeed associated with CIMP in microsatellite-stable and microsatellite-unstable colorectal tumors.
Studies of polyps revealed CIMP-positive polyps in HPS patients and most frequently in right-sided SSAs.
More recently, a hotspot BRAF pathogenic variant (V600E) was found to be common in MSI colon cancers and serrated polyps.
A BRAF pathogenic variant is absent in CRCs from Lynch syndrome patients and is rare in sporadic adenomatous colorectal polyps, but it is present in the vast majority of serrated polyps, especially SSAs.
CIMP positivity is commonly found in microvesicular hyperplastic polyps (MVHP), suggesting progression of MVHP to SSA and then to colon cancer.
The characterization of CIMP CRCs and evidence that MSI occurs later in the adenoma-carcinoma sequence leads to modification of the previous colorectal tumorigenesis model, which was comprised of two pathways: MSI (MIN) and CIN. There is much overlap between the MSI and CIMP pathways. At the heart of the CIMP pathway are serrated polyps harboring BRAF pathogenic variants. The CIN pathway is characterized by AP precursors of which the vast majority harbor APC pathogenic variants that occur early in the pathway.
主基因是指对致病是必要和充分的,以该基因的重要致病性变异(例如无义、错义、移码)作为起因机制。通常认为主基因与单基因疾病有关,而主基因引起的疾病通常相对罕见。主基因中的大多数致病性变异导致患病风险偏高,而且环境因素往往难以识别。
在过去,大多数主要结肠癌易感基因都是通过使用高风险家族的连锁分析来确定的;因此,由于研究设计的缘故,该定义符合上述标准。
在过去十年中,主要的CRC相关基因的功能已被合理地充分说明
抑癌基因是引发遗传性癌症综合征的最重要的基因类别,并且是引发家族性腺瘤性息肉病(FAP)、Lynch综合征和幼年性息肉病综合征(JPS)的基因类别。与CRC重大风险的相关基因及其相应疾病总结请见表2。
基因 | 综合征 | 遗传模式 | 主要癌症 |
---|---|---|---|
APC | FAP、AFAP | 显性 | 结直肠、小肠、胃等 |
TP53(p53) | Li-Fraumeni | 显性 | 多发性(包括结直肠) |
STK11(LKB1) | 显性 | 多发性(包括结直肠、小肠、胰腺) | |
PTEN | 显性 | 多发性(包括结直肠) | |
BMPR1A、SMAD4(MADH/DPC4) | 显性 | 胃和结直肠 | |
MLH1, MSH2, MSH6, PMS2, EPCAM | 显性 | 多发性(包括结直肠、子宫内膜等) | |
MUTYH(MYH) | 隐性 | 结直肠 | |
POLD1, POLE | PPAP | 显性 | 结直肠、子宫内膜 |
FAP =家族性腺瘤性息肉病;JPS =幼年性息肉综合征;PJS = Peutz-Jeghers综合征;PPAP =聚合酶校对相关息肉病。 |
直到20世纪90年代,遗传性息肉病综合征的诊断均基于临床表现和家族史。目前已确定了与这些综合征有关的一些基因,多项研究已尝试预估这些人群的自发致病性变异率(新发致病变异率)。值得注意的是,认为FAP、JPS、Peutz-Jeghers综合征、Cowden综合征和Bannayan-Riley-Ruvalcaba综合征的自发致病性变异率较高,范围为25%-30%,
而与Lynch综合征相关的MMR基因的新发致病变异率估计值较低,范围为0.9%-5%。
这些Lynch综合征自发致病性变异率的估计值可能与各人群中非亲子关系率的估计值重叠(0.6%-3.3%),
与其他息肉病综合征相对较高的致病性变异率相比,Lynch综合征的新发致病变异率可能相当低。
众所周知,结肠癌的家族性聚集也发生在特征明确的结肠癌家族综合症之外。
根据流行病学研究,受累患者的一级亲属患结肠癌的风险可使个体终身患结肠癌的风险增加2-4.3倍。
不同家族史类别的CRC的相对风险(RR)和绝对风险预估值请见表1。 此外,患结肠腺瘤的患者的一级亲属中患结肠癌的终生风险也增加。
风险程度取决于先证者诊断时的年龄、先证者与高危病例的相关程度以及受累的亲属数量。 当前认为,许多中等和低风险病例受到单低外显基因或低外显基因组合变异的影响。考虑到确定这种风险增加的病因对公共健康的影响,对相关基因的深入研究正在进行中。
预计每个基因位点对CRC风险的影响相对较小,并且不会产生Lynch综合征或FAP所见的众多家族聚集。但是,与其他常见的遗传基因位点和/或环境因素结合分析时,此类变异可能显著改变CRC风险。这些类型的遗传变异通常称为多态性。大多数多态位点对疾病风险或人类特征(良性多态性)无影响,而那些与疾病风险差异或人类特征(无论微妙)相关的位点有时被称为疾病相关多态性或功能相关多态性。 此类变异涉及DNA单核苷酸变化时,称为单核苷酸多态性(SNP)。
一些全基因组相关研究(GWAS)已经对相对较多的未经选择的CRC患者进行了研究,这些患者已经被评估了全基因组中候选基因和匿名基因的多态性模式。
目的是确定等位基因,尽管不是致病性变异,但可能导致CRC风险增加(或可能降低)。确定未知的异常CRC等位基因时,允许在遗传基础上进一步对高危患者进行分层。这种风险分层可能增强CRC筛查。在成千上万的CRC病例和对照病例中使用全基因组扫描,发现了多个常见的低风险CRC SNP,请见美国国家人类基因组研究所GWAS目录。有关GWAS的详细讨论,请参见PDQ总结:癌症遗传学概述。
人们越来越有兴趣利用SNP扩大种系风险评估,从单基因高/中等外显率形式的CRC易感性到多基因形式的肠癌风险评估,这可能对一般人群有更广泛的适用性。为此,多项研究检查了多基因风险评分(PRS)在处于CRC平均风险水平的患者的个性化CRC风险评估中的作用。
一项基于日本人群人口登记的研究调查了341例CRC男性和329例对照男性,发现了先前与CRC敏感性相关的36种不同SNP(按GWAS列出)。研究者最终确定了该人群中的六个SNP与CRC风险有关,并构建了PRS,该PRS在评估10年CRC绝对风险方面具有合理的区分能力(曲线下面积[AUC],0.63)。研究者发现,PRS的性能略微优于先前验证的结合年龄、体重指数以及吸烟和饮酒的非遗传风险预测评分(AUC,0.60),并发现包括SNP数据和非遗传因素的组合模型在评估10年绝对CRC风险方面具有优效的区分能力(AUC,0.66)。
同样,另一项研究检查了PRS(由48个先前CRC风险有关的SNP组成,按GWAS列出)在接受筛选结肠镜检查的1043例50-79岁德国患者中的作用。
研究者证明,PRS有效地区分了晚期肿瘤(癌或晚期腺瘤)与非晚期腺瘤和正常结肠镜检查结果的风险。研究估计,PRS最高三分位数的受试者与PRS最低三分位数的17.5岁以上受试者患结直肠癌的风险相同,表明该PRS数据可能有助于充分估计患者的风险,从而可以就先前处于CRC中等风险水平的患者在开始结肠镜检查时的年龄提供个性化建议。值得注意的是,另一项针对2363例CRC患者和2198例对照患者的病例对照研究表明,53个SNP PRS和CRC家族史均与CRC风险增加相关,但这些关联彼此独立。
研究者得出结论:PRS可能因此大大增加了基于家族史的CRC风险分层,并且与CRC风险相关的经GWAS鉴定的SNP可能并非大多数家族性CRC聚集的潜在因素。
尽管获得理想的数据,但是,必须强调的是此类PRS目前在常规临床环境中未使用,并且目前不具有临床作用。基于上述令人鼓舞的数据,有必要正式实施研究,以检查此类PRS的使用是否可以指导CRC风险评估和常规临床护理筛查。
值得一提的是APC I1307K多态性,因为它通常在接受多基因(组合)检测的Ashkenazi犹太血统的患者中检测出。
并且与CRC风险增加有关;但是,并不会引起结肠息肉病。I1307K多态性几乎只发生在Ashkenazi犹太人后裔中,导致结肠腺瘤和腺癌的风险比普通人群高出两倍。
I1307K多态性是由APC基因中第3920位核苷酸从T到A的转变所致,并且由于其导致了A8微卫星编码序列,可能形成一个超突变区。
尽管目前已有用于评估APC I1307K多态性的临床检测方法,但相关的CRC风险还不足以支持其常规使用。 根据当前可用的数据,尚不清楚I1307K携带者是否应指导有关开始筛查的年龄、筛查的频率或筛查策略选择的决策。
Major genes are defined as those that are necessary and sufficient for disease causation, with important pathogenic variants (e.g., nonsense, missense, frameshift) of the gene as causal mechanisms. Major genes are typically considered those that are involved in single-gene disorders, and the diseases caused by major genes are often relatively rare. Most pathogenic variants in major genes lead to a very high risk of disease, and environmental contributions are often difficult to recognize.
Historically, most major colon cancer susceptibility genes have been identified by linkage analysis using high-risk families; thus, these criteria were fulfilled by definition, as a consequence of the study design.
The functions of the major colorectal (CRC) cancer genes have been reasonably well characterized over the past decade.
Tumor suppressor genes constitute the most important class of genes responsible for hereditary cancer syndromes and represent the class of genes responsible for familial adenomatous polyposis (FAP), Lynch syndrome, and juvenile polyposis syndrome (JPS), among others. Table 2 summarizes the genes that confer a substantial risk of CRC, with their corresponding diseases.
Gene | Syndrome | Hereditary Pattern | Predominant Cancers |
---|---|---|---|
APC | FAP, AFAP | Dominant | Colorectal, small bowel, gastric, etc. |
TP53 (p53) | Li-Fraumeni | Dominant | Multiple (including colorectal) |
STK11 (LKB1) | Dominant | Multiple (including colorectal, small bowel, pancreas) | |
PTEN | Dominant | Multiple (including colorectal) | |
BMPR1A, SMAD4 (MADH/DPC4) | Dominant | Gastric and colorectal | |
MLH1, MSH2, MSH6, PMS2, EPCAM | Dominant | Multiple (including colorectal, endometrial, and others) | |
MUTYH (MYH) | Recessive | Colorectal | |
POLD1, POLE | PPAP | Dominant | Colorectal, endometrial |
FAP = familial adenomatous polyposis; JPS = juvenile polyposis syndrome; PJS = Peutz-Jeghers syndrome; PPAP = polymerase proofreading–associated polyposis. |
Until the 1990s, the diagnosis of genetically inherited polyposis 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, FAP, JPS, Peutz-Jeghers syndrome, Cowden syndrome, and Bannayan-Riley-Ruvalcaba syndrome are all thought to have high rates of spontaneous pathogenic variants, in the 25% to 30% range,
while 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.
These estimates of spontaneous pathogenic variant rates in Lynch syndrome seem to overlap with the estimates of nonpaternity rates in various populations (0.6% to 3.3%),
making the de novo pathogenic variant rate for Lynch syndrome seem quite low in contrast to the relatively high rates in the other polyposis syndromes.
It is widely acknowledged that the familial clustering of colon cancer also occurs outside of the setting of well-characterized colon cancer family syndromes.
Based on epidemiological studies, the risk of colon cancer in a first-degree relative of an affected individual can increase an individual’s lifetime risk of colon cancer 2-fold to 4.3-fold.
The relative risk (RR) and absolute risk of CRC for different family history categories is estimated in Table 1. In addition, the lifetime risk of colon cancer also increases in first-degree relatives of individuals with colon adenomas.
The magnitude of risk depends on the age at diagnosis of the index case, the degree of relatedness of the index case to the at-risk case, and the number of affected relatives. It is currently believed that many of the moderate- and low-risk cases are influenced by alterations in single low-penetrance genes or combinations of low-penetrance genes. Given the public health impact of identifying the etiology of this increased risk, an intense search for the responsible genes is under way.
Each locus would be expected to have a relatively small effect on CRC risk and would not produce the dramatic familial aggregation seen in Lynch syndrome or FAP. However, in combination with other common genetic loci and/or environmental factors, variants of this kind might significantly alter CRC risk. These types of genetic variations are often referred to as polymorphisms. Most loci that are polymorphic have no influence on disease risk or human traits (benign polymorphisms), while those that are associated with a difference in risk of disease or a human trait (however subtle) are sometimes termed disease-associated polymorphisms or functionally relevant polymorphisms. When such variation involves changes in single nucleotides of DNA they are referred to as single nucleotide polymorphisms (SNPs).
Several genome-wide association studies (GWAS) have been conducted with relatively large, unselected series of patients with CRC, who have been evaluated for patterns of polymorphisms in candidate and anonymous genes throughout the genome.
The goal is to identify alleles that, while not pathogenic variants, may confer an increase (or a potential decrease) in CRC risk. Identification of yet unknown aberrant CRC alleles would permit further stratification of at-risk individuals on a genetic basis. Such risk stratification would potentially enhance CRC screening. The use of genome-wide scans in thousands of CRC cases and controls has led to the discovery of multiple common low-risk CRC SNPs, which can be found in the National Human Genome Research Institute GWAS catalog. Refer to the PDQ summary on Cancer Genetics Overview for a thorough discussion of GWAS.
There is increasing interest in using SNPs to expand germline risk assessment from monogenic high-/moderate-penetrance forms of CRC predisposition to polygenic forms of CRC risk assessment that may have broader applicability to the general population. To that end, multiple studies have examined the utility of polygenic risk scores (PRSs) to personalize CRC risk assessment in individuals otherwise considered to be at average risk for CRC.
One study examined 36 different SNPs previously linked to CRC susceptibility by GWAS in 341 men with CRC and 329 controls from a population-based registry of Japanese individuals. Investigators ultimately identified six of these SNPs to be associated with CRC risk in this population and constructed a PRS, which had reasonable discriminatory capacity (area under the curve [AUC], 0.63) for assessing a 10-year absolute risk of CRC. The investigators found that the performance of the PRS was marginally superior to a previously validated nongenetic risk prediction score (AUC, 0.60) incorporating age, body mass index, and tobacco and alcohol use, and found that a combined model including both SNP data and these nongenetic factors had superior discriminatory capacity for assessing a 10-year absolute CRC risk (AUC, 0.66).
Likewise, another study examined the use of a PRS consisting of 48 SNPs previously linked to CRC risk by GWAS in 1,043 German individuals aged 50 to 79 years undergoing screening colonoscopy.
Investigators demonstrated that the PRS effectively discriminated between risk for advanced neoplasms (carcinoma or advanced adenomas) versus nonadvanced adenomas and normal colonoscopic findings. The study estimated that participants with the highest tertile of PRS have the same risk of advanced colorectal neoplasm as participants 17.5 years older from the lowest tertile of PRS, suggesting that such PRS data may help estimate individuals’ risk sufficiently well to allow for personalized recommendations regarding age at initiation of colonoscopic screening in individuals previously considered at average risk for CRC. Interestingly, another case-control study of 2,363 patients with CRC and 2,198 controls demonstrated that a 53 SNP PRS and family history of CRC were both associated with increased CRC risk, but that these associations appeared to be independent of one another.
Investigators concluded that PRS may thus substantially augment family history–based CRC risk stratification, and that GWAS-identified SNPs associated with CRC risk may not be the factor underlying most familial CRC clustering.
Despite such promising data, however, it is important to emphasize that such PRSs are not currently used in routine clinical settings and are not currently considered to be clinically actionable. Formal implementation studies examining the use of such PRSs to guide CRC risk assessment and screening in routine clinical care are warranted, on the basis of these encouraging data.
The APC I1307K polymorphism deserves special mention, given that it is commonly identified in individuals of Ashkenazi Jewish ancestry undergoing multigene (panel) testing
and is associated with an increased risk of CRC; however, it does not cause colonic polyposis. The I1307K polymorphism occurs almost exclusively in people of Ashkenazi Jewish descent and results in a twofold increased risk of colonic adenomas and adenocarcinomas compared with the general population.
The I1307K polymorphism results from a transition from T to A at nucleotide 3920 in the APC gene and appears to create a region of hypermutability by virtue of the fact that this results in an A8 microsatellite coding sequence.
Although clinical assays to assess for the APC I1307K polymorphism are currently available, the associated CRC risk is not high enough to support their routine use. On the basis of currently available data, it is not yet known whether the I1307K carrier status should guide decisions regarding the age to initiate screening, the frequency of screening, or the choice of screening strategy.
结肠癌易感综合症的名称通常反映出与该综合症(例如Gardner综合症、Turcot综合症、Muir-Torre综合症、Lynch综合症、Peutz-Jeghers 综合征[PJS]、Bannayan-Riley-Ruvalcaba综合征和Cowden综合征)相关的医师或患者及其家人,最初在20世纪80年代和90年代通过临床结果进行了描述。这些综合征与结直肠腺癌的终生风险增加有关。认为大多综合征具有常染色体显性遗传模式。腺瘤性结肠息肉是前四个综合症的特征,而错构瘤是后三个的特征。
随着人类基因组计划的发展以及1990年对5q号染色体上的腺瘤性息肉病(APC)基因的鉴定,这些家族性综合征之间的重叠和差异显而易见。 Gardner综合征和家族性腺瘤性息肉病(FAP)为同义词,两者均由APC基因的致病性变异所致。衰减型FAP(AFAP)被认为是一种由于APC基因3'或5'端致病性变异而导致的腺瘤和肠外表现较少的综合征。MUTYH相关息肉病(MAP)被认为是一种具有常染色体隐性遗传的单独腺瘤性息肉综合征。一旦确定了致病性变异,就可以更好地评估致病性变异携带者患结直肠癌(CRC)的绝对风险(见表3)
综合征 | 致病性变异携带者患CRC的绝对风险 |
---|---|
90%(45岁) | |
69%(80岁) | |
10%-56%(75岁),具体取决于累及的基因 | |
35%-53% | |
39%(70岁) | |
17%-68%(60岁) | |
FAP =家族性腺瘤性息肉病;JPS =幼年性息肉综合征;PJS = Peutz-Jeghers综合征。 | |
a此处引用的癌症风险估算值早于监测和预防性手术广泛使用的估算值。 | |
这些发现表明,基因检测和风险管理成为可能。基因检测是指使用多种技术在已知的癌症易感基因中寻找变异体。全面基因检测包括对基因的整个编码区进行测序、内含子-外显子边界(剪接位点)以及重排、缺失或其他拷贝数变化评估(使用多重连接依赖性探针扩增[MLPA]或Southern印迹法等技术)。尽管积累了丰富的经验,有助于区分致病性变异与良性变异和多态性,但基因检测识别出的不确定显著性变异体并不能用于预测(VUS)
直至1900年,多份报告表明,伴有大量息肉(随后被归类为腺瘤)的患者患CRC的风险非常高,而且家庭中的遗传方式为常染色体显性遗传。 在20世纪,腺瘤向肿瘤的发展过程被证实,并且FAP被认为是肿瘤发展的典型模式。
典型的FAP特征是在10岁后,结肠和直肠中会形成无数(数百至数千个)腺瘤性息肉(请见图3)。
也存在具有衰减型表型的典型FAP的亚型。AFAP是一种异质性临床实体,其特征是结肠和直肠的腺瘤性息肉少于典型FAP。(更多信息请参见本总结的“衰减型家族性腺瘤性息肉病[AFAP]”章节。)
FAP是遗传性结肠癌综合症中定义最明确和易理解的一种疾病。
其为常染色体显性遗传疾病,报道的活产子发病率为1/7,000-1/22,000。
有人认为FAP患病率存在种族差异
但是,一项大型研究并未发现在超过6169例具有CRC和息肉的个人和/或家族史的患者中,种族变异存在显著差异,其中这些患者在一家大型参考实验室接受了基因检测。
大多数FAP病例是由5q21号染色体上APC基因的致病性变异所致。 (有关APC基因和基因检测的更多信息,请参见该总结的FAP遗传学章节。)
除了FAP患者患结肠腺瘤的风险较高外,还描述了多种结肠外表现,包括上消化道(GI)腺瘤和腺癌;胃腺息肉;非上皮良性肿瘤(骨瘤、表皮囊肿、牙齿异常);硬纤维瘤;先天性视网膜色素上皮肥大症(CHRPE);和恶性肿瘤(甲状腺和脑瘤、肝母细胞瘤)。有关FAP中这些结肠外表现的风险,请参见表4。
恶性肿瘤 | 相对风险 | 终生绝对风险(%) |
---|---|---|
硬纤维瘤 | 852.0 | 15.0 |
十二指肠肿瘤和癌症 | 330.8 | 5.0–12.0 |
甲状腺癌 | 7.6 | 2.0 |
脑肿瘤 | 7.0 | 2.0 |
壶腹癌 | 123.7 | 1.7 |
胰腺癌 | 4.5 | 1.7 |
肝母细胞瘤 | 847.0 | 1.6 |
胃癌 | 未确定 | 0.6a |
改编自 Giardiello et al., Jagelman et al.,Sturt et al.,Lynch et al.,Bülow et al.,Burt et al.,and Galiatsatos et al. | ||
a利兹堡息肉病小组。 |
FAP也称为家族性结肠息肉病或腺瘤性息肉病(APC)。 Gardner综合征以前是以结肠息肉病、骨瘤和软组织肿瘤为表现的FAP患者的诊断。 但是,Gardner综合征在基因层面被证明是FAP的一个变种,因此术语Gardner综合征在临床实践中基本已过时。
携带APC基因遗传致病性变异的患者极有可能发展为结肠腺瘤。风险估计超过90%。
结肠中腺瘤发作时年龄不同,出现结直肠腺瘤的中位年龄为16岁。
10岁时,仅15%的APC种系变体携带者出现腺瘤。20岁时,这一概率上升到75%;30岁时,出现FAP的概率达90%。
AFAP是一个例外,在AFAP中,受影响的个体通常会有较少的结肠息肉,这些息肉主要分布在右半结肠,并且以后会发展为CRC。(更多信息请参见本总结的“衰减型家族性腺瘤性息肉病[AFAP]”章节。)在未进行任何干预的情况下,大多数FAP患者将在40岁出现CRC。
因此,对APC致病性变异携带者和高危人群进行监测和干预,通常包括从青春期开始的每年一次结肠镜检查,以早期检测结肠息肉且有助于计划何时进行结肠切除术。
(有关更多信息,请参阅该总结的“FAP干预”章节。)
CHRPE是视网膜上扁平的黑色素病变,FAP患者的发生率约为75%
而一般人群的发生率为1.2%。
在FAP患者中,病变通常出现在出生时或儿童早期,并且常常是多发性或双侧的。
一项针对17例确诊FAP的患者和13名高危家庭成员的研究显示,CHRPE病变与FAP中结肠息肉相关的检测灵敏度为76%,特异性为92%,阳性预测值为93%,阴性预测值为75%;因此,筛查CHRPE高危人群可能是发现FAP的合理方法。
硬纤维瘤为增生性、局部浸润性、无转移性,同时也是胶原基质中的纤维瘤。 尽管不会转移,但可以非常迅速地生长并威胁生命。
硬纤维瘤可能是散发,属于典型FAP的一部分,或以遗传方式出现而无FAP结肠征象。
即使硬纤维瘤与典型的结肠腺瘤性息肉病无关,也与遗传性APC致病性变异有关。
大多数研究发现,FAP患者中有10%出现硬纤维瘤,报告率为8%-38%。 发生率随确定方法和APC基因的致病性变异部位而异。
密码子1445和1578之间发生的APC致病性变异可增加FAP患者中硬纤维瘤的发生率。
在密码子1924的致病性变异患者中,已描述了发作较晚和肠息肉病表型较轻的硬纤维瘤(遗传性硬纤维瘤)。
已出现的硬纤维瘤危险因素量表用于辨别可能发展为硬纤维瘤的患者。
硬纤维瘤危险因素量表是基于性别、是否存在结肠外表现、硬纤维瘤家族史和基因型(如果可行)。通过使用该量表,可以将FAP患者分为发展为硬纤维瘤的低、中和高风险组。作者得出结论,硬纤维瘤危险因素量表可用于计划安排手术。来自欧洲的一项大型、多登记、回顾性研究支持了包括该量表在内的危险因素验证结果。
硬纤维瘤的自然病史易变。 一些作者提出了一种硬纤维瘤形成模型,即成纤维细胞功能异常会导致肠系膜样斑块状胶质前体病变,在某些情况下可能在术前发生,并在手术创伤后发展为肠系膜纤维瘤病,最终导致硬纤维瘤。
据估计,10%硬纤维瘤会分解,50%可以长时间保持稳定,30%出现波动,10%迅速生长。
硬纤维瘤通常在手术或生理创伤后发生,并且与内分泌和遗传因素相关。 FAP中约80%的腹腔内硬纤维瘤在手术创伤后发生。
FAP中的硬纤维瘤通常存在于腹腔内,可能出现较早,并可能导致肠梗阻或缺血和/或输尿管梗阻。
硬纤维瘤是FAP患者中仅次于CRC的第二常见死亡原因
有人提出了一个分期系统,以便于按疾病严重程度对腹腔内硬纤维瘤进行分层。
拟定的腹腔内硬纤维瘤分期系统如下:无症状非生长性硬纤维瘤为I期;最大直径不超过10 cm的有症状的非生长性硬纤维瘤为II期;有症状的11 cm-20 cm硬纤维瘤或无症状的缓慢生长的硬纤维瘤为III期;大于20 cm或生长迅速或危及生命的并发症的硬纤维瘤为IV期。
这些数据表明,基因检测可能对FAP和/或多发性硬纤维瘤患者的临床治疗有意义。 APC基因型易形成硬纤维瘤的患者(例如,在APC基因的3'末端或1445密码子)在进行任何手术(包括降低风险的结肠切除术和腹腔镜检查等外科手术)后患硬纤维瘤的风险偏高。
与FAP相关的最常见胃息肉为胃底腺息肉(FGP)。 FGP通常为弥漫性,不适合通过内窥镜切除。 据估计,FAP患者中FGP的发生率高达60%,而一般人群中FGP的发生率为0.8%-1.9%。
这些息肉由扭曲的基底腺组成,基底腺内衬有微囊,囊内衬有基底型上皮细胞或小叶粘液细胞。
增生的表面上皮,顾名思义,是非肿瘤性的。因此,FGPs并不被认为是癌前病变。但是,由FGP引起的胃癌病例报告导致对该问题进行重新审查。
一项FAP检查显示,25%患者在FGP的表面上皮中的局灶性非典型增生明显,而散发性FGP则为1%。
一项FAP患者接受食管胃十二指肠镜检查的前瞻性研究显示,在88%患者中检测到FGP。在上述患者中,有38%检出低度不典型增生,有3%检出高度非典型增生。该研究的作者建议,如果发现高度不典型增生的息肉,则应考虑在3-6个月内行息肉切除术并再次进行内窥镜检查。
使鉴别诊断问题复杂化的是,FGP在使用质子泵抑制剂(PPIs)的非FAP患者中的发现率越来越高。
在这种情况下,FGP通常表现出PPI效应,包括壁细胞分泌颗粒的充血,从而导致单个细胞向腺腔内不规则膨出。 有经验的医生会发现,存在不典型增生和特征性PPI效应同时缺失明确提示存在潜在的FAP。 尽管出现一些重叠,但FAP中FGP的数量往往比使用PPI的患者中观察到的多。
FAP患者也会出现胃腺瘤。 据报道,西方患者胃腺瘤的发生率为2%-12%,而在日本则为39%-50%。
这些腺瘤可发展为癌。据报道,韩国和日本FAP患者患胃癌的风险是这些国家一般人群的三倍至四倍,而在西方人群中未观察到这一现象。
亚洲FAP患者胃腺瘤患病率高于西方FAP患者,一种可能的解释是幽门螺杆菌感染的总体患病率较高。
最近,在西方FAP数据库中观察到胃腺癌的发病率上升。
在患有胃腺癌和胃近端息肉病(GAPPS)的家庭中发现了APC启动子(1B)改变,这些家庭表现为有许多(主要是胃底腺体)局限于胃体和胃底的胃息肉,伴有局灶性不典型增生或胃腺癌,但无结直肠或十二指肠息肉的迹象。 这些变异与多个GAPPS家族中的胃表型分离。 尽管胃息肉病表型的外显率很高,从无症状的成人到有严重症状的胃息肉病青少年,以及42-77岁接受内镜检查的未受累的携带者,其表型可能各不相同。但是,尚不清楚胃癌的外显率。 在有胃底腺息肉和结肠息肉的FAP家庭中很少发生启动子1B APC变异。
在未经选择的上消化道内镜检查患者中,十二指肠腺瘤的发生率仅为0.4%,
在FAP患者中十二指肠腺瘤发生率为80%-100%。大多数位于十二指肠的第一部分和第二部分,尤其是在壶腹周围区域。
FAP患者的十二指肠腺癌终身发病率为4%-12%。
一项针对来自北欧的368例FAP受试者的十二指肠腺瘤的前瞻性多中心监测研究显示,65%受试者在基线评估时出现腺瘤(平均年龄为38岁),70岁时累计患病率达90%。与早期关于惰性病程的看法相反,在平均8年的监测期间,腺瘤的体积和不典型增生程度增加,尽管在前瞻性监测中仅4.5%患癌。
这是一项大型研究,但是,由于使用了前视镜而不是侧视镜以及大量的研究人员参与了这项研究,因此受到了限制。胶囊内镜也可以评估FAP患者的肠息肉。
一项计算机断层扫描(CT)十二指肠造影术研究发现,可以精确测量较大体积腺瘤,但无法准确计算较小的扁平腺瘤。
对FAP患者的回顾性研究表明,腺瘤-癌序列与壶腹周围腺癌存在时间关系,平均年龄为39岁时确诊腺瘤,平均年龄为47岁时确诊为高度不典型增生,平均年龄为54岁时确诊为腺癌。
对601例FAP患者的决策分析表明,从30岁开始定期监测的获益可将预期寿命提高7个月。
尽管十二指肠中的息肉可能很难治疗,但小型研究表明,内窥镜检查可成功治疗息肉,但潜在的并发症可能主要来自胰腺炎、出血和十二指肠穿孔。
患有特别严重的十二指肠息肉病(有时称为致密性息肉病)或组织学晚期十二指肠腺瘤的FAP患者发展为十二指肠腺癌的风险最高。
由于十二指肠腺癌的风险与息肉的数量和体积以及息肉的不典型增生的严重程度相关,因此开发了一种结合这些特征的分层系统,以试图确定发展为十二指肠腺癌的风险最高的FAP患者。
根据该系统(称为Spigelman分类系统(请参见表5)),最晚期患者中有36%发展为癌。
评分 | 息肉数 | 息肉体积(mm) | 组织学 | 不典型增生 |
---|---|---|---|---|
1 | 1-4 | 1-4 | 管状 | 轻度 |
2 | 5-20 | 5-10 | 管状绒毛状 | 中度 |
3 | >20 | >10 | 绒毛 | 重度 |
I期,1-4分;II期,5-6分;III期,7–8分;IV期,9–12分。 |
FAP患者中出现的其他结肠外肿瘤包括甲状腺乳头状癌、肾上腺肿瘤、肝母细胞瘤和脑瘤。
据报道,甲状腺乳头状癌(筛状桑树型)会影响1%-2%的FAP患者。
但是一项 研究
针对6名FAP妇女的乳头状甲状腺癌的研究未能证明6种肿瘤的密码子545和1061至1678中杂合性(LOH)丢失或野生型等位基因的致病性变异。 此外,这些患者中有五分之四具有可检测的体细胞RET / PTC嵌合基因。该致病性变异通常局限于散发性甲状腺乳头状癌,提示除了APC致病性变异以外还涉及其他遗传因素。 还需要进一步研究来证明其他遗传因素(例如RET / PTC嵌合基因)是否独立引起或协同APC变异,从而引发FAP患者的甲状腺乳头状癌。
在FAP患者中有肾上腺肿瘤的报道,一项研究表明FAP患者肾上腺皮质癌(ACC)的APC位点有LOH。
一项针对162例接受腹部CT的FAP患者以评估腹部内硬纤维瘤的研究显示,发现15例(11名女性)患有肾上腺肿瘤。
其中,2例患者的症状是由皮质醇分泌过多所致。 其中3例患者接受了后续手术,发现患有ACC、双侧结节性增生或肾上腺皮质腺瘤。 该队列中非预期肾上腺肿瘤的患病率为7.4%,而非FAP患者的患病率为0.6%-3.4%(P < 0.001)。
在该研究中,未对切除的肿瘤进行分子遗传学分析。 后续研究表明,26%(23/90)的FAP患者、18%(2/11)的AFAP患者和24%(5/21)的MAP患者出现肾上腺病变。该研究中的大多数病变均遵循良性缓慢进展过程。未报告ACC病例。
肝母细胞瘤是一种罕见、快速进展且通常具有致命性的儿童恶性肿瘤,如果局限于肝脏,则可以通过根治性手术切除进行治愈。已有报道,在出现APC致病性变异的儿童中出现了多例肝母细胞瘤病例
一些研究还证明了这些肿瘤中APC的LOH。
在患有肝母细胞瘤的FAP患者中尚未发现特定的基因型-表型相关性。
(如需了解更多信息,请参见PDQ儿童肝癌治疗总结中的肝母细胞瘤章节。)
伴CRC和脑瘤的一系列病症
FAP脑肿瘤患者中也有
5q21染色体上的APC基因编码2,843个氨基酸蛋白,对细胞粘附和信号转导至关重要。APC蛋白的主要功能是调节细胞内β连环蛋白的浓度,而β连环蛋白是一种Wnt信号转导途径的主要介质。 APC是一种抑癌基因,APC的丢失是染色体不稳定性结直肠肿瘤途径中最早发生的事件之一。 FAP和AFAP可以通过检测外周血白细胞DNA中APC基因的种系致病性变异进行遗传诊断。 已报道了300多种与疾病相关的APC基因致病性变异。
这些变化大多数为插入、缺失和无意义的变异,这些变异导致基因的转录物中移码和/或提前终止密码子。 最常见的APC致病性变异(占FAP患者的10%)是1309号密码子中AAAAG缺失。无其他致病性变异占主导地位。 减少(而非消除)APC蛋白产生的变异也可能导致FAP。
出现在169号密码子和1249号密码子之间的大多数APC致病性变异导致典型的FAP表型。
人们对基因中致病性变异部位与临床表型之间关系的关注度逐渐提高:
外显率
表现出典型FAP表型的患者可以进行APC检测。但是,在许多具有息肉病的个人或家族史的先证者中,考虑到息肉病的遗传异质性和相关综合症之间的表型重叠,多基因组合检测是一种适当的选择。
尤其是结直肠腺瘤性息肉少于100个的患者,可能对诊断带来挑战。鉴别诊断包括AFAP、MAP、聚合酶校正相关息肉病(PPAP)和双等位基因错配修复缺陷(BMMRD)。
可以通过检测种系APC致病性变异进行AFAP诊断。 (更多信息请参见本总结的“衰减型家族性腺瘤性息肉病[AFAP]”章节。)MAP是由MUTYH基因中的双等位基因种系致病性变异引起,以常染色体隐性方式遗传。
PPAP是由POLE和POLD1中的杂合致病性变异引起。
BMMRD是个体在MMR基因(MLH1、MSH2、MSH6、PMS2或EPCAM)的两个等位基因位点均发生遗传致病性变异的一种情况。
(更多信息请参见本总结的“MUTYH相关的息肉病[MAP]、寡聚性息肉和双等位基因错配修复缺陷[BMMRD]”章节。)
例如,一项大型的横断面研究显示,发现具有1,000个以上腺瘤的患者中APC的致病性变异率为80%(95%置信区间[CI],71%–87%),具有100-999个腺瘤的患者中APC的致病性变异率为56%(95% CI,54%–59%),具有20-99个腺瘤的患者中APC的致病性变异率为10%(95% CI,9%–11%),具有10-19个腺瘤的患者中APC的致病性变异率为5%(95% CI,4%–7%)。
在同一项研究中,双等位基因MUTYH致病性变异的发生率与衰减型表型(20–99个腺瘤)患者的APC相似,但是在少数(2%)典型息肉病患者中也观察到了MUTYH致病性变异。
大多数商业实验室不仅进行全基因测序,还对APC和其他基因进行缺失/复制分析。但是,重要的是需与每个实验室一起验证检测方法。缺失分析对于患有FAP的个体尤其重要,因为8%-12%受累患者在APC基因中有一个完整的外显子缺失或启动子1B缺失,而测序无法检测到。
如前所述,对于出现息肉病的患者,通常需要预定包括多个息肉病基因的多基因组合检测,这可以通过同时评估所有基因简化检测程序并降低检测成本。(如需了解更多信息,请参见PDQ“癌症遗传学风险评估和咨询”总结中多基因[组合]检测章节。)
在已发现APC基因的致病性变异的家庭中,对高危亲属的预测性检测可以明确鉴定或排除变异。该检测对于确定高危亲属是否需要进行积极筛查非常重要,并且能确定是否需要这些筛查或者确定是否终止此类筛查(即,对于家族致病性变异检测呈阴性的亲属)。
大多数FAP患者有患病的父亲或母亲,并且在家庭中可以观察到常染色体显性遗传的模式。因此,级联遗传咨询和检测可随后扩展到高危家庭成员。 但是,据估计25%的FAP患者的APC出现新发致病性变异,这意味着该变异可能并非遗传自父亲或母亲。
如果在父亲或母亲的白细胞DNA中未发现该变异,则种系嵌合可解释这一发现。因此,应始终为患者的兄弟姐妹进行APC检测,但是先证者的姨妈、叔叔和堂兄则不是必须检测。
FAP的临床特征的早期出现以及随后从青春期开始进行监测的建议引起了有关未成年人基因检测的特殊考虑。
通常不建议对未成年人进行遗传性癌症综合症的基因检测,除非其结果有望为儿童时期的医疗管理提供参考。因此,FAP提供了一个实例,其中可能的临床获益证明对具有已知致病性变异家庭中的未成年人进行基因检测的合理性,尤其是对于预计50%的高危儿童,若被发现未携带致病性变异体,则可以免于监测。 此外,对婴儿进行FAP检测,从而对肝母细胞瘤进行监测,直到5岁为止。 否则,如果未检测高危未成年人,则在10-15岁时开始进行结肠镜检查或乙状结肠软镜检查。
本总结的“遗传性结肠癌综合征”章节中的心理社会问题中解决了此类检测的心理影响。
由于家庭或自身已知的APC致病性变异,有FAP风险的个体可通过乙状结肠软镜或结肠镜评估息肉病的发病。一旦发现FAP家庭成员出现息肉,预防CRC的唯一有效方法即为结肠切除术。研究已证明预防性手术可以延长FAP患者的生存期。
如果可行,应将患者及其家庭成员纳入登记表中,因为回顾性分析表明,登记和监测可降低CRC的发生率和死亡率。
在病程中很早确诊典型FAP的患者中,外科医生、内镜医师和家人可能选择推迟手术数年,以实现社会价值。此外,在经仔细选择的AFAP患者(息肉负担最小且年龄高的患者)中,选择仅在息肉负担增加或出现不典型增生的情况下再进行手术,从而推迟结肠切除术的决定可能具有一定的合理性。
芬兰一项全国性的基于人口的回顾性研究评估了对FAP家庭成员的监测是否能降低总体死亡率并提高生存率,结果表明入选筛查计划的先证者的家庭成员在确诊FAP后存活期长达20年,与一般人群相当。
该研究包括154个家庭,其中从1963年至2015年至少一名家庭成员临床确诊FAP。有194名先证者和225名家庭成员(83名通过基因检测确诊,142名通过内窥镜检查确诊),中位随访时间为11.8年。 在该研究中,使用相对生存估计值计算了FAP家族成员的生存分析结果。
根据该估计值,比较了FAP先证者和家庭成员的生存期与每个日历年中性别和年龄相同的患者在无FAP的情况下预期的生存期。先证者的相对生存率在随访10年后为67%(95%CI,60%–75%),在20年后为66%(95%CI,58%–76%)。对于家庭成员,在10年时的相对生存率为98%(95%CI,95%–101%),在20年时为94%(95%CI,88%–100%)。 经过25年的随访,家庭成员的相对存活率低于一般人群,为87%(95%CI,79%–96%)。先证者的相对生存率明显低于家庭成员(P < 0.001)。 在死亡率方面,先证者的标准化死亡率比值在0-5年和5-10年随访期均增加,而对于家庭成员,仍保持稳定,直到20年的随访期。考虑到大多数可能有症状,并且很可能在诊断时患有CRC,因此在先证者随访开始时,这种差异更加明显。作者指出,如果成功治疗CRC而未复发,则先证者的生存率接近其家庭成员的生存率。
内窥镜检查监测通常较早开始(10-15岁)。
(如需了解有关早期监测对社会和情感的影响的更多信息,请参见本总结的“遗传性结肠癌综合征的社会心理问题”章节。)在过去,乙状结肠镜检查可能已成为大多数患者早期腺瘤鉴别的合理方法。然而,鉴于以下方面,结肠镜检查是首选方法:(a)全结肠镜检查器械的改进;(b)镇静;(c)对AFAP的认识,这种疾病通常最常出现在右半结肠;(d)延后手术的趋势不断增长,长达数年。
对其他已知的家庭致病性变异检测为阴性的患者无需进行面向FAP的内窥镜检查;NCCN建议进行平均风险人群筛查。对于受累的人员中未发现家族变异的家庭,有必要进行临床监测。 已知携带APC致病性变体但尚未表现出息肉的人,结肠监测并未停止,因为腺瘤有时直到生命的第四和第五十年才表现出来。 (如需了解有关上述方法的更多信息,请参见PDQ结直肠癌筛选期总结。)
在APC致病性变异阳性的患者中,近100%出现结肠腺瘤;在息肉出现以及息肉数量过多或组织学进展至无法使用内镜切除进行安全监测后,降低风险的手术是预防CRC标准治疗的组成部分
FAP患者及其医生应进行个性化讨论,以决定何时进行手术。 将术后发生硬纤维瘤的风险以及女性的生育能力纳入讨论是有益的降低风险的手术的时间通常取决于息肉的数量、体积、组织学和症状。
一旦大量息肉形成,结肠镜检查就不再有助于确定结肠切除的时机,因为息肉数量太多,以至不可能对所有息肉进行活检或切除。 此时,病人最好咨询一位有经验的外科医生,他有多种选择,包括全结肠切除术和全结肠切除一期重建。
保留直肠手术,辅以乙状结肠镜下监测直肠,是一种合理的替代全结肠切除术的方法,适用于那些直肠息肉分布稀疏、了解相应后果、对直肠残端癌知情同意的患者(尽管进行定期监测)。
手术选择包括重建性结肠直肠切除术辅以回肠储袋-肛管吻合术(IPAA),全结肠切除术辅以回肠直肠吻合术(IRA)或全结肠直肠切除术辅以回肠造口术(TPC)。对不能保留括约肌的低位直肠癌患者或因技术问题而不能进行IPAA的患者进行TPC。 TPC后没有发生直肠癌的风险,因为去除了处于危险中的整个粘膜。这些手术可以使用微创技术进行。
无论是否进行结肠切除术和IRA或全结肠切除术一期重建,大多数专家建议对直肠或回肠袋进行定期和终生监测,以切除或消融任何息肉。 早期未经选择的研究显示,IRA后20年全结肠切除术后患直肠癌的风险高达25%。
据报告,IRA患者选择越好,该风险越低。
据报道,IRA后直肠癌风险增加的因素包括整个结肠的息肉数量、直肠的息肉数量、IRA时是否存在结肠癌、直肠残端的长度、IRA后随访时间和基因型。
以IRA作为FAP的主要手术的腹部结肠切除术并不排除后来因不受控制的直肠息肉和/或直肠癌而转为IPAA的可能性。在丹麦息肉病登记研究中,24例患者的继发性IPAA(既往IRA后)的并发症率和功能结果与59例接受原发性IPAA的患者相似。
在大多数情况下,手术时直肠中的临床息肉负荷决定了手术干预的类型,即采用恢复性直肠结肠切除术加IPAA还是IRA。具有轻微表型(<1,000个结肠腺瘤)且直肠息肉少于20个的患者在进行预防性手术时可能适合IRA。
然而,在某些情况下,息肉负担不能明确,在这种情况下,研究者探索了基因型在预测直肠后续结果方面的作用。
据报道,IRA后增加直肠癌风险和最终完成直肠切除术的致病性变异包括外显子15密码子1250、外显子15密码子1309和1328以及外显子15密码子1250和1464之间的变异。
在接受IPAA治疗的患者中,继续对回肠贮袋进行年度监测非常重要,因为据报道,在15岁时,该囊中发展成腺瘤的累积风险高达75%。
尽管癌变罕见,但在FAP患者接受直肠结肠切除术后,报道显示在回肠贮袋和肛管移行区发生癌变。
对复原性全结肠切除术和IPAA术后生活质量的荟萃分析表明,在瘘管形成、贮袋炎、大便次数和渗漏方面,FAP患者的确比炎症性肠病患者稍好。
塞来昔布是一种特异性环氧合酶2(COX-2)抑制剂,同时也是非特异性COX-2抑制剂,例如舒林酸(一种非甾体类抗炎药[NSAID]),该药物可使FAP患者的息肉体积减小及数量减少,提示化学预防剂在治疗该疾病中的作用。
尽管塞来昔布已获得美国食品药品监督管理局(FDA)批准,但其许可证已由生产商自愿撤销。目前,尚无FDA批准的FAP化学预防药物。然而,诸如塞来昔布和舒林酸的药物已被广泛使用,以致化学预防性临床试验通常利用这些药物之一作为对照组。一项随机临床试验显示,与塞来昔布单药相比,塞来昔布和二氟甲基鸟氨酸联合给药可轻微改善息肉负荷。
一项评价塞来昔布在儿童人群(年龄10-14岁)中的疗效的小型、随机、安慰剂对照、剂量递增临床试验显示,给药时间在3个月内时,塞来昔布在所有剂量水平上都是安全的
该研究发现腺瘤性息肉负担减轻具有剂量依赖性。在16 mg/kg/日剂量下,相当于批准的成人每日两次400毫克的剂量,息肉负担减少与塞来昔布在成人中的有效性平行。
一项有关结肠次全切除术后的FAP患者的小型研究显示,游离脂肪酸形式的ω-3多不饱和脂肪酸二十碳五烯酸可减少直肠息肉的数量和体积。
虽然在随机试验中没有直接比较,但其疗效似乎与之前用塞来昔布观察到的结果相似。
目前尚不清楚如何将COX-2抑制剂纳入尚未接受降低风险手术的FAP患者治疗中。 一项对41例尚未表现出息肉病的APC致病性变异的儿童和年轻成人携带者进行的双盲安慰剂对照临床试验表明,舒林酸可能并非FAP的主要有效治疗药物。 治疗4年后,舒林酸和安慰剂组在息肉发生率、数量或体积方面不存在统计学显著差异。
一项随机、前瞻性、双盲、安慰剂对照的临床试验显示,在6个月疗程治疗后,塞来昔布可减少但不会消除32例FAP患者的十二指肠息肉数量,与COX-2抑制剂对结肠息肉的作用一致。 重要的是,仅在基线检查时息肉累及>5%十二指肠且口服剂量为400 mg(每日两次)的患者中观察到统计学显著作用。
先前在舒林酸治疗6个月的24例FAP患者中实施的一项随机研究显示,十二指肠息肉的减少无明显趋势。
与使用COX-2抑制剂治疗结肠息肉有关的问题在治疗十二指肠息肉时也同样存在(例如,仅部分消除息肉,COX-2抑制剂继发的并发症以及停药后疗效丧失)。
由于十二指肠乳头(胆汁进入肠道)周围常见腺瘤性息肉聚集,并且临床前数据表明熊去氧胆酸盐抑制携带Apc种系变异的小鼠的肠腺瘤,
因此进行了两项使用熊去氧胆酸的临床试验。
两项研究均显示,熊去氧胆酸对十二指肠息肉无显著的化学预防作用矛盾的是,在一项研究中,熊去氧胆酸联合塞来昔布可以增加FAP患者的息肉密度。
由于有报道显示服用罗非昔布和塞来昔布的患者中的心脏相关事件增加,
目前尚不清楚该类药物对于FAP患者和一般人群是否可以长期安全使用。 同样,由于这些试验的短期(6个月)性质,目前尚未获得长期服用COX-2抑制剂的FAP患者心脏事件的临床信息。
一项队列研究表明,以舒林酸治疗FAP可使结肠和直肠腺瘤消退。该试验报告的结局是息肉的数量和体积,这是主要关注的临床结局(CRC发生率)的替代指标。
评价小分子表皮生长因子受体(EGFR)抑制剂和低剂量舒林酸在Apcmin / +小鼠中的有效性的临床前研究显示肠腺瘤的发生率降低87%,
表明EGFR抑制剂可能抑制FAP患者的十二指肠息肉。 一项为期6个月的双盲、随机、安慰剂对照临床试验比较了150 mg舒林酸(每日两次)和75 mg厄洛替尼(每日一次)与安慰剂在FAP或AFAP十二指肠息肉患者中的疗效。
将92例FAP或AFAP患者随机分配,接受研究药物或安慰剂治疗,并接受治疗前和治疗后上消化道内镜检查,以确定十二指肠近端10 cm段息肉的总直径和息肉数量的变化。 该试验因达到主要终点而提前终止。 意向性治疗分析显示舒林酸/厄洛替尼组的十二指肠息肉负担(直径总和)中位数减少了8.5 mm,而安慰剂组增加了8 mm(P < 0.001)。 与安慰剂组相比,治疗组的1级和2级不良事件发生率明显更高:治疗组中60.9%患者出现了痤疮样皮疹,32.6%患者出现了口腔粘膜炎;在安慰剂组中,19.6%患者出现了痤疮样皮疹,10.9%患者出现了口腔粘膜炎。 基于先前舒林酸和塞来昔布对FAP患者十二指肠息肉的适度作用
以及基因EGFR抑制作用对Apcmin / +小鼠肠腺瘤形成的显著作用,
厄洛替尼可能是该试验成功的原因。 一项正在进行的临床试验正在确定单独使用较低剂量的厄洛替尼是否足以显著减轻FAP和AFAP患者的十二指肠息肉负担。
携带APC种系致病性变异的患者罹患其他类型恶性肿瘤(包括硬纤维瘤、胃肿瘤、十二指肠癌、小肠癌、肝母细胞瘤、甲状腺癌和脑瘤)的风险增高。上述结肠外肿瘤的治疗请见下文。
FAP中的硬纤维瘤治疗可能具有挑战性,并使预防工作复杂化。目前尚无针对硬纤维瘤的公认标准治疗方法。多种药物治疗通常无法成功治疗硬纤维瘤。治疗方案包括抗雌激素药、非甾体抗炎药、化疗和放疗等。研究评估了雷洛昔芬单药、他莫昔芬或雷洛昔芬联合舒林酸和吡非尼酮单药的疗效。
一项雷洛昔芬(120 mg,每日一次)的前瞻性研究纳入了13例腹腔内硬纤维瘤和/或对其他药物治疗反应不良且胶质组织中有雌激素-α受体表达的患者。
6例患者在接受雷洛昔芬治疗前接受过他莫昔芬或舒林酸治疗,而7例患者既往未接受过治疗。开始治疗后7个月至35个月,所有13例腹腔内硬纤维瘤患者均达到部分缓解或完全缓解,并且大多数硬纤维瘤的体积在治疗后4.7个月(±1.8 mo)时缩小。有硬纤维瘤斑块和病变明显的患者发生反应。研究局限性包括样本量小和临床疗效评价,这在所有患者中均不一致。关于已接受雷洛昔芬治疗且不表达雌激素-α受体的硬纤维瘤患者以及从该潜在治疗中获益的患者的预后,存在几个问题。
第二项在13例接受他莫昔芬120 mg/日或雷洛昔芬120 mg/日与舒林酸300 mg/日联合治疗的FAP相关硬纤维瘤患者中实施的临床研究显示,10例患者病情稳定(n = 6)或达到部分缓解或完全缓解(n = 4)超过6个月,3例患者病情稳定超过30个月。
上述结果表明这些药物联合治疗可有效减缓硬纤维瘤的生长。然而,硬纤维瘤的自然史易变,既有自发消退,也有生长速度的变化。
第三项研究报告了14例接受吡非尼酮治疗2年的FAP相关硬纤维瘤患者的综合结果。
本研究部分患者病情消退,部分患者出现疾病进展,部分患者病情稳定。
据报告,采用甲磺酸伊马替尼治疗FAP患者的硬纤维瘤取得一定成功。
在硬纤维瘤患者中,伊马替尼治疗失败后尼洛替尼可稳定硬纤维瘤生长。
酪氨酸激酶抑制剂索拉非尼治疗硬纤维瘤的获益在一项III期随机试验中得到证实,该试验在87例不可切除的进展性或症状性硬纤维瘤患者中比较了索拉非尼(400 mg,每日一次)与安慰剂的疗效。
对于安慰剂组中发生疾病进展的患者,允许交叉至索拉非尼组。49例接受索拉非尼治疗的患者中16例(33%)出现客观缓解,而35例接受安慰剂治疗的患者中7例(20%)出现客观缓解。此外,索拉非尼组的2年无进展生存率(PFS)(81%)显著高于安慰剂组(36%);进展或死亡的风险比为0.13(95%CI,0.05-0.31;P < 0.001)。最常见的不良事件包括1级或2级皮疹(73%)、疲乏(67%)、高血压(55%)和腹泻(51%)。 尽管毒性特征相对较小,但约20%的患者因毒性反应而中止索拉非尼治疗,强调了适当延迟和中断给药对治疗不良事件的重要性。
由于并发症率和复发率较高,一般情况下,治疗腹腔内硬纤维瘤时不推荐手术切除。回顾一家医院的经验表明,腹腔内硬纤维瘤的手术结局可能比先前认为的更佳。
研究对象的选择是评价手术结果的关键。
腹壁硬纤维瘤可行手术切除,但复发率较高。
目前尚不清楚胃腺瘤的治疗方法。 仅回顾性病例分析结果可供参考,结果表明FAP患者中胃腺癌的发生率相对较低。
最近,在西方FAP数据库中观察到胃腺癌的发病率上升,
表明FAP中胃肿瘤发生后治疗方案应该有所变化。有研究小组建议采用内镜下息肉切除术治疗胃腺瘤。
通常根据腺瘤的体积和不典型增生的程度,对胃内腺瘤进行个体化治疗。
FAP患者通常在25-30岁进行基线上消化道内镜检查,包括侧视十二指肠镜检查。
随后的内镜检查间隔时间根据既往内镜检查结果而不同,通常基于Spigelman分期。 推荐的间隔时间是基于专家意见,尽管0期至II期疾病相对宽松的间隔时间部分基于荷兰/斯堪的纳维亚十二指肠监测试验生成的自然史数据(关于按Spigelman分期的筛查频率的可用建议,请参见表6)。
Spigelman分类的主要优势在于本领域技术人员长期了解和使用,这使得研究结果间比较的合理标准化成为可能。
但是,Spigelman分类使用局限如下所示:
Spigelman分期 | NCCN(2019) | ESMO(2013) |
---|---|---|
0(无息肉) | 内镜检查(每4年一次) | 未指定 |
I | 内镜检查(每2-3年一次) | 内镜检查(每5年一次) |
II | 内镜检查(每1-3年一次) | 内镜检查(每3年一次) |
III | 内镜检查(每6-12个月一次) | 内镜检查(每1-2年一次) |
IV | 手术评估 | 内镜检查(每6-12个月一次) |
完全粘膜切除术或十二指肠切除术,或如果累及十二指肠乳头则进行Whipple手术 | ||
每3-6个月进行一次专家内镜监测 | 手术选择包括十二指肠切开术联合息肉切除术、保留胰腺的十二指肠切除术和胰十二指肠切除术(Whipple手术) | |
ESMO = 欧洲肿瘤医学学会;NCCN = 美国国家综合癌症网络。 | ||
如需了解有关针对Spigelman IV期疾病使用手术切除的更多信息,请见下文。 |
北欧国家和荷兰FAP患者的长期十二指肠腺瘤监测结果显示,FAP患者存在显著的十二指肠癌风险。
根据方案,从1990年至2000年每两年进行一次前视内镜检查。 随后,根据国际指南对患者进行监测随访。研究的304例患者中,261例(86%)接受一次以上内镜检查。 中位随访时间为14年(范围:9-17年)。 十二指肠腺瘤病的终生风险为88%。44%患者的Spigelman分期随时间恶化,而12%改善,34%保持不变。 20例患者(7%)在中位年龄56岁(范围:44-82岁)时发生十二指肠癌。75岁时癌症累积发生率为18%(95%CI,8%-28%)。有症状性癌症患者的生存率低于监测内镜检查时确诊的患者。
许多因素(包括息肉病的严重程度、合并症、患者偏好和是否有经验丰富的医生)决定了是否选择手术或内镜治疗进行息肉治疗。 内镜下切除或消融大的或病理分期较晚的腺瘤可安全有效地降低短期发生十二指肠腺癌的风险;
然而,内镜下切除腺瘤的患者仍有发生十二指肠复发性腺瘤的重大风险。
降低腺癌风险的最确切的手术是壶腹和十二指肠的手术切除,尽管这些手术导致的并发症发生率和死亡率高于内镜治疗。 有学者应用十二指肠切开术和十二指肠息肉局部切除术或粘膜切除术,但进行这些手术后息肉复发率高。
一项在47例接受根治性手术并处于Spigelman III期或IV期的FAP患者中实施的研究显示,平均随访时间为44个月时局部复发率为9%。该局部复发率显著低于同一研究中任何局部内镜或手术治疗的复发率。
胰十二指肠切除术和保留胰腺的十二指肠切除术被认为可显著降低患壶腹周围腺癌的风险,是可选用的术式。
如果考虑此类手术选择,保留幽门对该组患者尤其有益,因为大多数患者将接受IRA结肠次全切除术或IPAA结肠全切除术。如一项北欧研究,
以及其他研究所示,
大多数十二指肠腺瘤患者不会发生癌变,可通过内窥镜检查进行随访。然而,进展期腺瘤(Spigelman III期或IV期疾病)患者通常需要接受内镜或手术治疗息肉。FAP患者十二指肠腺瘤的化学预防研究正在进行中,可能在未来提供一种替代策略。(更多信息请参见本总结的化学预防章节。)
较大和(或)较平坦的十二指肠腺瘤的内镜检查取决于壶腹是否受累。 粘膜下注射生理盐水(加或不加肾上腺素和/或染料,如靛胭脂)后的内镜下粘膜切除术(EMR)可用于切除非壶腹病灶。 需要进一步治疗壶腹病灶,包括内镜超声检查评价胆汁或胰管受累的迹象。 胰管支架置入术常用于预防狭窄和胰腺炎。 支架需要在1至4周的时间间隔内通过内镜移除。 由于壶腹系于导管口,注射时通常不能均匀提起,因此通常不使用注射方法 进行EMR或壶腹切除术时,需要丰富的经验和判断力,仔细考虑未治疗病变的自然史,并知晓尽管进行了积极的内镜干预,腺瘤复发率仍然很高。
文献一致支持采用十二指肠切除术治疗Spigelman IV期疾病。对于Spigelman II期和III期疾病,内镜治疗的作用总是集中于存在的一个或两个最严重的病变。
手术切除的顾虑与手术相关短期并发症率、死亡率及远期并发症有关。尽管可能夸大了这些担忧,
但对手术干预的恐惧可能导致激进和有点不明智的内镜干预。在某些情况下,通过内镜检查不能完全或安全地完成壶腹和(或)其他十二指肠腺瘤的内镜切除术,在无短肠综合征风险的情况下也不能完成十二指肠切除术,或者因为肠系膜纤维化根本不能完成该手术。 在这种情况下,可以进行外科经十二指肠壶腹切除术/息肉切除术。 然而,这可能导致局部复发风险偏高,这与内窥镜治疗相似。
尽管缺乏1级证据,但在以下方面达成了共识:建议从青少年后期开始每年进行甲状腺检查,以筛查FAP患者的甲状腺乳头状癌。同一专家建议临床医生可以考虑在该筛查中常规增加每年一次的甲状腺超声检查。
尽管缺乏1级证据,但专家在以下方面达成了共识:建议有FAP倾向的儿童在5岁前每3-6个月进行一次肝脏触诊、腹部超声检查和血清甲胎蛋白测定。
5岁以后不必继续筛查。
髓母细胞瘤是一种高度恶性肿瘤,通常在诊断前6个月或更短时间内才出现症状;每年对无症状患者的监测可能不足。 因此,不提倡通过定期CT或磁共振成像进行监测。对于尚未患有息肉病,但有提示脑肿瘤的体征或症状的FAP家族成员,应进行神经影像学评价,因为半数以上的FAP患者在息肉病诊断前存在脑肿瘤。当有一个在FAP家庭成员已经出现脑瘤,则对该家族成员仔细评估也很重要,因其存在家族聚集性。在这类FAP相关脑瘤家庭中,40%有2名累及成员。
在1990年,首次在一个腺瘤数量不等的大型家族中进行了AFAP临床描述。该家族中腺瘤的平均数量为30个,尽管数量从几个到几百个不等。
建议AFAP患者的治疗包括结肠镜检查,而非可屈性乙状结肠镜检查,因为腺瘤可能主要位于右半结肠。
认为AFAP的腺瘤在20岁中期到20岁晚期形成。
与典型FAP相似,AFAP患者发生CRC的风险更高;然而,诊断时平均年龄比典型FAP大56 岁。
累及家族成员发生CRC,但同步息肉极少。
AFAP也出现与典型FAP相似的结肠外表现。这些表现包括上消化道息肉(FGP、十二指肠腺瘤和十二指肠腺癌)、骨瘤、表皮样囊肿和硬纤维瘤。
由于引起AFAP的APC致病性变异部位特定,这些患者通常不会出现CHRPE病变。
AFAP与APC致病性变异的特定亚型相关。已确定3组引起AFAP的部位特定APC致病性变异:
在无类似累及亲属家族史的情况下,鉴别诊断可能包括AFAP(包括MAP)、Lynch综合征、BMMRD、DNA聚合酶校对亚基(POLD1或POLE)的种系变异,或其他未分类的散发或遗传问题。细致询问家族史可能会发现AFAP或Lynch综合征。
APC检测是评价疑似AFAP患者的重要组成部分。
如果在可疑的AFAP患者中种系APC致病性变异检测呈阴性,则可能需要对MUTYH、POLE和POLD1致病性变异进行基因检测。
在适合的年龄进行结肠镜检查,遇到腺瘤计数异常高或无法计数的患者,鉴别诊断是一个挑战。
在AFAP中降低风险的结肠切除术的作用和时间存在争议。
来自不同专业协会有关AFAP监测的临床实践指南总结请见表7。
组织 | 疾病 | 筛查方法 | 筛查频率 | 开始筛查的年龄 | 注释 |
---|---|---|---|---|---|
Europe Mallorca Group(2008) | AFAP | 结肠镜检查 | 每2年一次;如果检测到腺瘤,则每1年一次 | 18-20年 | |
NCCN(2019) | 有腺瘤负担的AFAP个人病史a | 结肠镜检查 | 每1-2年一次 | 如果患者接受结肠切除术联合IRA,则每6-12个月进行一次内镜检查,具体取决于息肉负担。 | |
≥21岁患者可考虑行结肠切除术和IRA。 | |||||
NCCN(2019) | 内镜检查无法处理的有腺瘤负担的AFAP个人病史 | 不适用 | 不适用 | 不适用 | 首选结肠切除术联合IRA。如果出现严重的直肠息肉病,可以考虑行直肠结肠切除术联合IPAA。 |
NCCN(2019) | 未累及的高风险家庭成员;已知家庭致病性变异;APC致病性变异状态未知或阳性 | 结肠镜检查 | 如果APC为阳性,则每1-2年一次;如果未知,则每2–3年一次 | 青少年后期 | 如果未检测APC致病性变异状态,请考虑进行基因检测。 |
IPAA =回肠储袋-肛管吻合术;IRA =回肠直肠吻合术;NCCN =美国国家综合癌症网络。 | |||||
a 少于20个直径<1 cm且无晚期组织学的腺瘤,因此可同时采用结肠镜检查与息肉切除术有效地切除息肉。 |
MAP是由Mut Y同源基因的致病性变异引起的常染色体隐性遗传的息肉病综合征。Mut Y同源基因(称为MUTYH)最初被称为MYH,但由于肌球蛋白重链基因已经具有该名称,因此随后进行了更正。MUTYH位于染色体1p34.3-32.1上。
MUTYH编码的蛋白是碱基切除修复糖基化酶,可修复最常见的氧化性损伤之一。已报道了超过一百种MUTYH的独特序列变体(Leiden开放变异数据库)。假定MUTYH致病性变异为具有种族分化的先证者致病性变异。在北欧血统的白人人群中,两个主要变异,即Y179C和G396D(前称Y165C和G382D),占MAP患者双等位基因致病性变异的70%;这些患者中有90%至少携带一种致病性变异。
已经发现的其他诱发性变异包括P405L(前称P391L)(荷兰)、
E480X(印度)、
Y104X(巴基斯坦)、
1395delGGA(意大利)、
1186-1187insGG(葡萄牙)、
和p.A359V(日本和韩国)。
2002年,MUTYH基因由于存在于患有多发性结肠腺瘤和CRC但无APC致病性变异的三个兄弟姐妹中,而首次被证实与息肉病相关。
MAP具有广泛的临床谱。大多数情况下,它有类似于AFAP的临床表现,但也有与典型FAP和Lynch综合征表型相似的相关病例的报道。
与具有APC致病性变异的患者相比,MAP患者的年龄越大,则腺瘤更少
但患CRC的风险仍然较高(35%–75%)。
在2012年,一项针对7225例结直肠腺瘤负担的研究报告显示,10-19个腺瘤的双等位基因MUTYH致病性变异发生率为4%(95%CI,3%–5%),20-99个腺瘤中为7%(95%CI,6%–8%),100-999个腺瘤中为7%(95%CI,6%–8%)。
这种广泛的临床表现源于MUTYH纯合或复合杂合基因导致疾病的能力。根据来自多个FAP登记的研究,大约7%-19%具有FAP表型且没有可检测到的APC种系致病性变异的患者携带MUTYH基因的双等位基因变体。
在MAP患者中可观察到腺瘤、锯齿状腺瘤和增生性息肉。
CRC往往发生于右半结肠,且同时出现,可能比散发性CRC预后更佳。
MAP的临床治疗指南为:从18-30岁开始,每年1次至每3年1次进行结肠镜监测,
从25岁-30岁开始进行上消化道内镜监测。
(有关MAP患者结肠监测的可用临床实践指南的更多信息,请参见表8。)推荐的上消化道内镜监测间隔时间可参考基于Spigelman标准的受累负担。
MUTYH相关的息肉病患者可能需要进行全结肠切除术联合回肠直肠吻合术或结肠次全切除术,具体取决于总体息肉负荷。
尽管MAP是迄今为止唯一已知的双等位基因(隐性)腺瘤癌症倾向综合征,但也有一些双等位基因病例表现为儿童肿瘤,其中涉及MMR基因。(如需了解更多信息,请参见本总结Lynch 综合征章节的双等位基因错配修复缺陷章节。)
来自不同专业协会关于双等位基因MAP结肠监测的临床实践指南总结请见表8。
组织 | 状态 | 筛查方法 | 筛查频率 | 开始筛查的年龄 | 注释 |
---|---|---|---|---|---|
Europe Mallorca Group(2008) | MUTYH致病性变异携带者 | 结肠镜检查 | 每2年一次 | 18-20年 | |
Nieuwenhuis et al.(2012) | MUTYH致病性变异携带者 | 结肠镜检查 | 每1-2年一次 | ||
NCCN(2019) | MAP病史,小腺瘤负担a | 结肠镜检查 | 每1-2年一次 | 如果患者接受结肠切除术联合IRA,则每6-12个月进行一次内镜检查,具体取决于息肉负担。 | |
≥21岁患者可考虑行结肠切除术和IRA。 | |||||
NCCN(2019) | MAP病史伴内镜下不能处理的腺瘤负荷 | 不适用 | 不适用 | 不适用 | 首选结肠切除术联合IRA。如果出现稠密的直肠息肉病,可以考虑行直肠结肠切除术联合IPAA。如果患者接受结肠切除术联合IRA,则每6-12个月进行一次直肠内镜检查评估,具体取决于息肉负担。 |
NCCN(2019) | 未累及的高风险家庭成员;已知家庭致病性变异;MUTYH致病性变异状态未知或阳性(双等位基因) | 结肠镜检查 | 每1-2年一次 | 25-30年 | 如果单个MUTYH致病性变异呈阳性,则从40岁开始,或者FDR诊断为CRC年龄前10年开始,每5年进行一次结肠镜检查(如适用)。如出现以下情况,需要证据告知筛查建议:存在MUTYH变异和患CRC的SDR;以及没有CRC家族史的未受CRC影响的单等位MUTYH携带者 |
CRC = 结直肠癌;FDR = 一级亲属;IPAA = 回肠储袋-肛管吻合术;IRA = 回肠直肠吻合术;NCCN = 美国国家综合癌症网络;SDR = 二级亲属。 | |||||
a 少于20个直径<1 cm且无晚期组织学的腺瘤,因此可同时采用结肠镜检查与息肉切除术有效地切除息肉。 |
在MAP患者中报告了许多结肠外癌症,包括胃癌、小肠癌、子宫内膜癌、肝癌、卵巢癌、膀胱癌、甲状腺癌和皮肤癌(黑色素瘤、鳞状上皮癌和基底细胞癌)。
此外,在少数MAP患者中报告了非癌性结肠外表现,包括脂肪瘤、视网膜色素上皮先天性肥大、骨瘤和硬纤维瘤。
女性MAP患者患乳腺癌的风险增加。
MAP中的这些结肠外表现发生频率低于FAP、AFAP或Lynch综合征中的发生频率。
与FAP相似,MAP患者常发生十二指肠腺瘤,并有发展为十二指肠癌的风险。 相较于FAP,从最近辨别MAP的情况来看,MAP中十二指肠息肉的发生率和十二指肠癌的风险尚不明确。小样本研究表明,MAP中十二指肠息肉的发生率约为30%,远远低于FAP。 在一项基于登记的研究中,十二指肠息肉的发生率为17%;但是,本研究中仅50%患者接受了上消化道内窥镜检查,表明可能低估了十二指肠息肉的发生率。十二指肠癌的终生风险估计为4%。
来自英国和荷兰的一项登记研究探索了一组接受定期十二指肠监测的MAP患者中十二指肠息肉和十二指肠癌的发生率。
在92例患者中,31例(34%)有十二指肠息肉证据。十二指肠腺瘤检测时的中位年龄为50岁,65%患者在基线内镜检查时确诊为十二指肠腺瘤。84%患者在首次检测到息肉时为Spiegelman I期或II期息肉病,无患者出现IV期息肉病,且未检出高度不典型增生。在随后监测中,仅2例患者分别在3.6年和7.0年后进展为Spiegelman IV期息肉病。 此外,可能保留了壶腹,仅2例患者有小息肉,壶腹无不典型增生。在这些登记研究中入组上消化道监测项目的患者中未检测到癌症。 在首次上消化道内镜检查时,2例MAP患者分别在83岁和63岁时确诊为壶腹癌和十二指肠癌。因此,与FAP相比,MAP中十二指肠息肉的发生率较低,且在较晚的年龄出现。 基于上述结果,作者建议在35岁时开始对MAP进行上消化道内镜筛查。
由于MAP为常染色体隐性遗传,受累患者的兄弟姐妹携带双等位基因MUTYH致病性变异的概率达25%,应对其进行基因检测。同样,可对受累患者的伴侣进行检测,以便评估其子女的患病风险。
尚未确定单等位基因MUTYH致病性变异的临床表型在发病率和相关临床表型方面的特征,其在息肉病和结直肠癌易感性中的作用仍不明确。大约1%-2%的一般人群携带MUTYH的致病性变异。
2011年的一项荟萃分析发现,单等位基因MUTYH致病性变异携带者患CRC的风险轻度增加(比值比[OR],1.15;95%CI,0.98-1.36);然而,考虑到单等位基因致病性变异携带者的罕见性,该携带者在所有CRC病例中仅占微不足道的比例。
一项关于携带MUTYH致病性变异的264例CRC病例的9,504名亲属中2,332个杂合子的大型研究显示,无论家族史如何,男性在70岁时患CRC的风险为7.2%,女性为5.6%。 在50岁前确诊为CRC的FDR患者中,男性在70岁时的风险为12.5%,女性为10%。
在解读本研究时应谨慎,因为本研究的绝大多数携带者状态是估算出来的,而非基于基因型。作者认为,与一般人群相比,患CRC的FDR中MUTYH杂合子的风险足够高,需要更密集监测(但与50岁前确诊CRC的FDR相同)。
MMR基因可能与MUTYH相互作用,增加患CRC的风险。已报告MUTYH和MSH6之间的相关性。两种蛋白在碱基切除修复过程中相互作用。一项研究报道,与非CRC人群相比,患CRC的单等位基因MUTYH致病性变异携带者中MSH6致病性变异显著增加(11.5% vs. 0%;P = 0.037)。
然而,德国的一项研究未能重复上述结果。
此外,一项更大规模的研究发现,与单独携带MMR致病性变异的患者相比,携带MUTYH变体的MMR致病性变异携带者的患癌风险未增加。
寡息肉病是一个流行术语,用于描述息肉计数或负担的临床表现,在筛查过程中,平均风险患者的息肉计数或负担大于预期结果,但不符合FAP诊断要求。 因此,对少数患者,寡(希腊语)对不同的观察者可能有不同的意义。 虽然在该问题上缺乏共识,美国国家综合癌症网络(NCCN)CRC筛查委员会建议,当终生总共存在10-99个腺瘤时,应考虑AFAP的诊断。因此,术语寡息肉病将用于描述息肉计数(一般为腺瘤)足够大的情况,有或无任何伴随家族史,以提高内镜医师对遗传易感性的可能性。
即使可以在内窥镜下清除结肠息肉,但如果复发腺瘤的风险增加,通常也需处理这类病例。
由致病性种系APC变异所致的AFAP可能是寡息肉病的最常见原因,其中已确定了一种特定的致病种系突变癌。 一些伴有寡息肉病的AFAP病例最终将发展为100多个腺瘤,尽管年龄较晚,通常以右半结肠微腺瘤为主,左半结肠息肉较少、较大。 有阳性家族史和APC致病性变异的病例显然是FAP的变异病例,术语AFAP提示了这一点.
然而,无直接家族史和腺瘤负荷较小的患者可能未携带APC致病性变异。 息肉计数越低,携带APC致病性变异的概率越低。 目前,其中一些病例已知携带双等位基因MUTYH致病性变异或与寡息肉病相关的其他基因变异。
已在寡息肉病、CRC和子宫内膜癌家族中描述了相关DNA聚合酶基因POLE和POLD1的致病性变异,该疾病称为聚合酶校对相关息肉病(PPAP)。
在15例60岁前有10个以上腺瘤的患者中,使用先进的全基因组测序方法。 一些患者的近亲至少有5个腺瘤,也可以进行全基因组测序。 所有受试患者均患有CRC或一级亲属(FDR)患CRC。 APC、MUTYH、MMR基因致病性变异检测均为阴性。在被评估的家族中没有发现共同的变异。然而,在一个家系中,连锁已经建立了共享区域,其中发现一个共享变异(POLE p.Leu424Val;c.1270C>G),在蛋白质结构和功能上预计有严重重排。 在验证阶段,对近4000例富集存在多发性腺瘤的受累病例进行了该变异的检测,并与近7000例对照病例进行了比较。 在这项工作中,发现另外12例不相关病例存在L424V变异,对照组均不存在变异。 在受累家族中,多发性腺瘤风险的遗传可能为常染色体显性遗传。
采用类似的方法对共享变异进行全基因组检测,并通过连锁分析进一步“过滤”,从而确定了POLD1基因中的变异(p.Ser478Asn;c.1433G > A)。 在最初评价的两个家族中确定了该S478N变异,表明具有共同祖先的证据。 验证活动显示1例息肉患者存在变异,但对照组无变异。 体细胞突变模式与POLE变异相似。 可见数例早发型子宫内膜癌。 POLE L424V变异导致腺瘤和癌形成的潜在机制使复制相关聚合酶校对保真度降低。 相反可能导致与碱基置换相关的变异。随后的一项研究证实,POLE致病性变异是寡息肉病和早发性CRC的罕见原因。
本研究中所有患者的APC、MUTYH和MMR基因种系致病性变异均呈阴性。在485例结直肠息肉病和早发性CRC的索引病例中,3例发现了POLE变异体L424V。肿瘤显示微卫星不稳定性(MSI),3例索引病例中的2例缺乏一种或多种MMR蛋白。 在这2例病例中检测到MMR基因的体细胞突变,可能是继发于POLE缺陷的超突变结果。癌症基因组图谱研究网络对276个CRC进行了广泛的测序分析,结果发现POLE基因中存在的体细胞突变与超突变表型相关,突变负担远远大于携带MSI的CRC中的突变负担。因此,聚合酶变异可能在肿瘤中产生了超突变基因型。
一项对来自48个家庭的51例多发性结肠腺瘤患者进行全外显子测序的研究发现,在碱基切除修复基因NTHL1中,来自3个不相关家庭的7例受累患者存在纯合子种系无义致病性变异。
这些患者患有CRC、多发性腺瘤(8-50),都不是增生性或锯齿状的,其中3名受累女性中患有子宫内膜癌或子宫内膜复杂型增生症。 另外2例患者发生十二指肠腺瘤和十二指肠癌。所有家系均符合常染色体隐性遗传。 在检查了来自不同受累患者的三种癌症和五种腺瘤后,均未显示MSI。这些肿瘤确实显示胞嘧啶向胸腺嘧啶转变的富集。需要额外研究进一步确定表型。 随后对863个CRC家庭和1600个非CRC家庭的研究证实了双等位基因NTHL1致病性变异与遗传性CRC风险之间的相关性。
目前,对于携带单个单等位基因致病性种系NTHL1变异的患者,尚无已知增加的癌症风险。
遗传性混合性息肉病的组织学特征常包括腺瘤性和增生性息肉,与少数德系犹太人家族中GREM1致病性变异有关。该综合征的息肉数量具有高度变异性,但通常在与寡息肉病一致的范围内。(如需了解更多信息,请参见本总结的遗传性混合性息肉病综合征[HMPS]章节。)
NTHL1、POLE、POLD1和GREM1致病性变异体检测正在与APC和MUTYH一起被纳入商业用途的CRC易感性多基因(组合)检测中,以便可以为寡聚症患者提前订购息肉病组合检测。对于在NTHL1(仅双等位基因携带者)、POLE或POLD1中发现致病性种系变异的患者,最佳监测方法的数据极少,尽管推测CRC的风险与Lynch综合征中观察到的相当,一些指南支持类似的早期和频繁的结肠镜筛查。
可根据简单的内镜和组织学数据区分由其他息肉病组织学引起的寡息肉病与腺瘤性息肉病。 例如,患有幼年性息肉病综合征(JPS)、PJS或PTEN错构瘤肿瘤综合征(Cowden综合征)的患者均可表现出寡息肉病,通常包括错构瘤性息肉,以及其他更常见的息肉组织学病灶(例如腺瘤)。
锯齿状息肉病同样具有高度变异性。 世界卫生组织(WHO)锯齿状息肉病标准(乙状结肠近端≥5个锯齿状息肉伴2个息肉≥1 cm,或如果存在锯齿状息肉病的亲属,乙状结肠近端任何数量的息肉,或≥20个结肠任何部位的锯齿状息肉)从未得到验证。在罕见情况下,锯齿状息肉病家庭被确定为携带致病性种系RNF43变异,但大多数锯齿状息肉病病例与潜在的遗传疾病无关。
因此,越来越多的此类患者被转诊接受遗传咨询并考虑基因检测。 在至少具有锯齿状息肉病某些特征的患者中发现了MUTYH双等位基因致病性变异的偶发病例,Lynch综合征中可见锯齿状息肉。 然而,锯齿状息肉病患者的种系评价结果通常不显眼。
两项极小样本研究描述了先前因儿童期恶性肿瘤接受化疗和放疗治疗的患者中,具有不同息肉组织学(例如腺瘤、锯齿状、炎性和错构瘤性息肉)的寡息肉病。
这种现象称为治疗相关性息肉病(TAP),可能是由既往抗肿瘤治疗引起的获得性、非家族性表型,在既往接受过化疗和/或放疗的患者中发现非家族性寡息肉病时,可进行鉴别诊断。最近的另一项研究发现,在101例既往接受过化疗和/或放疗的霍奇金淋巴瘤生存者队列中,6%患者符合WHO锯齿状息肉综合征(SPS)标准,表明霍奇金淋巴瘤生存者可能是TAP表现中特别重要的人群。
Lynch综合征是最常见的遗传性CRC综合征,约占所有最新确诊CRC病例的3%。Lynch综合征是一种常染色体显性遗传病,由MMR基因MLH1(mutL同源物1)、MSH2(mutS同源物2)、MSH6(mutS同源物6)和PMS2(减数分裂后分离2)以及基因EPCAM(上皮细胞粘附分子,前称TACSTD1)的致病性变异引起,其中EPCAM缺失引起MSH2的表观遗传沉默。Lynch综合征也与易患多种结肠外癌有关,包括皮脂腺腺瘤及子宫内膜和卵巢癌、胃癌、小肠癌、输尿管和肾盂的移行细胞癌、肝胆系统癌、胰腺癌和脑瘤。Lynch综合征相关癌症表现出MSI;因此,除了家族史外,肿瘤检测是Lynch综合征诊断的重要组分。现在推荐对所有CRC进行普遍肿瘤检测,作为一种策略,以筛查Lynch综合征和识别那些随后可能从种系基因检测中获益的患者。强化癌症筛查和监测策略(包括频繁的结肠镜检查,以及降低风险的手术)是Lynch综合征患者的主要治疗手段。
在1913年至1993年期间,有很多的结直肠癌呈现明显的家族聚集性发病。随着一系列此类报告的积累,一些特征性的临床特征出现了:CRC发病年龄早;同时性(和异时性)结直肠肿瘤风险高;右半结肠癌常见;临床结局较好;多个相关的结肠外部位包括子宫内膜、卵巢、胃肠道的其他部位、尿路上皮、脑和皮肤(皮脂腺肿瘤)。 癌症家族综合征和遗传性非息肉病性结直肠癌(HNPCC)等术语用于描述该疾病。
Lynch综合征一词取代了HNPCC,适用于遗传学上实与DNA MMR基因的种系致病性变异相关的病例。此外,HNPCC具有误导性,因为许多患者患有息肉,并且许多患者出现肿瘤(不包括CRC)。
随着对具有CRC遗传倾向的家族的认识增加,对致病性病因进行研究后制定了1990年阿姆斯特丹标准。
阿姆斯特丹标准最初用于识别高风险家庭,并满足以下所有条件:三例或三例以上两代或两代以上CRC,至少一例在50岁前确诊,无FAP证据。
在1987年,由于5q小片段染色体缺失,检测到了FAP与该基因组区域之间的遗传连锁,
其中APC基因最终于1991年被克隆。
这促使在怀疑患有Lynch综合征的家族中寻找类似的遗传连锁,其中这些家族有多例常染色体显性遗传CRC,并且癌症发病较早。APC基因是在符合阿姆斯特丹标准的家族中评价的多个基因(以及DCC和MCC)之一,但在Lynch家族中未发现基因连锁。 在1993年,进行了扩展的全基因组搜索,从而在大家族中发现了候选的2号染色体易感位点。 对MSH2(首个Lynch综合征相关基因)进行测序后,从CRC肿瘤的体细胞突变模式可以明显看出,MMR基因家族可能受累。随后,其他MMR基因与Lynch综合征相关,包括MLH1、MSH6和PMS2。Lynch综合征现指由其中一个DNA MMR基因的种系变异引起的遗传病,将其与其他家族性CRC集群进行区分。
在2009年,在无MSH2种系致病性变异的情况下,将EPCAM的种系缺失确定为MSH2失活的另一大原因。EPCAM变异导致MSH2启动子超甲基化。因此,EPCAM并非DNA MMR基因,也与Lynch综合征有关,目前对高危患者的EPCAM以及上述DNA MMR基因进行常规检测。
根据家族史标准和年轻人CRC的个人史,具有CRC优势和可能遗传倾向的家族最初被归类为Lynch综合征。随着分子肿瘤诊断检测的出现和与Lynch综合征相关的种系突变的发现,由于临床标准的作用不大而失去了人们的青睐。然而,临床标准的使用或Lynch综合征预测模型提供的风险评估可能适用于无个人癌症史但有提示Lynch综合征家族史的患者,或有CRC但无可用肿瘤组织进行分子诊断检测的患者。(如需了解更多信息,请参见本总结“用于筛查Lynch综合征的普遍肿瘤检测和预测MMR基因致病性变异概率的临床风险评估模型”章节。)
确定Lynch综合征家族的首个标准是1990年在阿姆斯特丹召开的国际协作组会议上制定的标准,也称为阿姆斯特丹标准。
这些研究标准仅限于家族性CRC诊断。在1999年,对阿姆斯特丹标准进行了修订,纳入了一些结肠外癌症,主要是子宫内膜癌。
这些标准为识别Lynch综合征家族提供了一种通用方法,但并不具有全面性;在近一半符合阿姆斯特丹标准的家族中未检测到致病性变异。
随后将这些标准用于非研究目的,以确定微卫星和种系检测的潜在候选者。然而,阿姆斯特丹标准未能确定相当大比例的Lynch综合征家族;符合阿姆斯特丹标准I但无MSI证据且DNA MMR基因无致病性种系变异的家族称为家族性结直肠癌X型(FCCX)。(更多信息请参见本总结的FCCX章节。)
由于与Lynch综合征肿瘤相关的MSI的标志性特征,以及与低灵敏度相关的阿姆斯特丹标准的局限性,1997年引入了Bethesda指南。Bethesda指南结合了临床、组织病理学和家族癌症史特征,确定了需要MSI肿瘤筛查的CRC病例。Bethesda指南(在2004年进行了后续修订)是针对那些需要考虑评估MMR缺陷的CRC肿瘤患者而制定的,目的是提高临床标准的敏感性,这些临床标准用于确定适合突变DNA分析的个体。
(如需了解有关MSI和IHC检测的更多信息,请参见本总结“Lynch综合征的基因和分子检测”章节。)
*考虑进行MSI检测的肿瘤必须符合一个标准。
**Lynch综合征相关肿瘤包括结直肠、子宫内膜、胃、卵巢、胰腺、输尿管和肾盂、胆道和脑肿瘤;Muir-Torre综合征中的皮脂腺腺瘤和角化棘皮瘤;以及小肠癌。
尽管Bethesda指南识别Lynch综合征携带者的比例高于阿姆斯特丹标准,但仍会忽略大约30%的Lynch综合征家族。
此外,Bethesda指南在临床实践中并没有被一致地用于确定应该进行MSI检测的CRC患者的亚群;医护人员认为该指南繁琐且难以记忆,因此使得患者会失去基因检测的机会。
随着替代方法的出现,包括对所有最新确诊的MSI CRC病例进行普遍检测(不考虑诊断时的年龄或癌症家族史),Lynch综合征的临床标准已经过时。 虽然Bethesda指南是针对癌症患者,但其在未受癌症累及的个体中的表现可能仍然有用。鉴于对未累及的Lynch综合征患者进行评估的方法有限,家族史和临床标准的使用可能适合于识别需要进一步基因评估和检测的患者。
由于医护人员未有效地使用临床标准选择CRC患者进行Lynch综合征的基因检测和评估,2006年开发并引入了基于计算机的临床预测模型作为替代方法,从而对Lynch综合征进行系统的遗传风险评估。 风险模型包括PREMM(基因突变预测模型)模型、MMRpredict和MMRpro。
三个模型(PREMM [1,2,6]、MMRpredict和MMRpro)量化了个体携带MLH1、MSH2和MSH6中MMR基因变异的概率。 随后将扩展使用(1,2,6)模型预测致病性PMS2和EPCAM变异,并且该模型是唯一能预测与Lynch综合征(PREMM5)相关的所有5个基因的模型。
虽然这些模型创建的目的相同,但开发方式和用于预测风险的变量不同。此外,对其进行验证的人群显示了每个模型可能影响准确度的特定特征。
决定在风险评定过程中使用哪种模型取决于应用该模型的临床环境和正在评价的患者人群。 MMRpro的预测考虑了家庭规模和未受累亲属、将分子肿瘤数据纳入风险分析的可能性以及在种系检测后预测致病性变异携带状态的选择。MMRpro在常规实践中广泛使用的主要限制是需要输入整个家系的数据(包括无癌症的个体),这相对耗时。最佳用途可能是在专门的高风险诊所或研究机构中作为遗传咨询工具,因为其访问权限有限。PREMM的主要优势包括易于使用、可作为在线工具使用并已得到广泛验证,包括在GI诊所的自我管理中。
包括基于二级亲属的个人和家庭癌症史的风险预测,以确定一系列结肠外肿瘤。然而,该模型未考虑家族规模,可能高估了家系中致病性变异的概率,其中该家系包括多个未受CRC或子宫内膜癌累及的老年家族成员。考虑到可以使用PREMM模型的便利性(在验证研究中,认为其耗时少于MMRpro),
不同的医护人员可以使用,其主要目的是确定应转诊进行基因评价的患者,并且可能在预检测决策过程中最有用。最后,由于风险估计不太准确,MMRpredict使用可能整体受限
当用于评估Lynch综合征相关癌症家族和CRC累及的老年患者时;可使用年轻发病的CRC病例(< 55岁时确诊的患者)数据开发该模型,不包括结肠外恶性肿瘤。此外,该模型不纳入肿瘤检测结果或提供基于基因测序结果的事后风险估计。
总体而言,有充分证据表明,与现有的Lynch综合征诊断和评价临床指南相比,每种模型均具有优越的性能特征,包括灵敏度、特异性以及阳性和阴性预测值,从而支持其使用。由于临床环境的多样性,医护人员有机会评估患者的Lynch综合征,预测模型提供了一个潜在可行且有用的策略,以系统地识别处于风险的患者,无论他们是否受到CRC的影响
总之,由于CRC中存在肿瘤MSI,以及明确的个人和家族癌症史,因此可对Lynch综合征进行种系基因检测,大多数临床实践指南均提供了此类方法。这些指南将遗传咨询和检测策略与临床筛查和治疗措施相结合。提供者和患者均可使用这些指南来更好地理解可用的选择和关键决策。(如需了解Lynch综合征诊断和结肠监测实践指南的更多信息,请参见表13。)
肿瘤和种系的遗传学在Lynch综合征的发生和诊断中均发挥重要作用。Lynch综合征相关肿瘤的肿瘤DNA表现出特征性的MSI,在这些病例中,与MMR基因相关的一种或多种蛋白的IHC表达通常缺失。已采用MSI和/或IHC分子检测作为普遍筛查方法,以诊断最新确诊的CRC和子宫内膜癌患者的Lynch综合征。IHC检测结果可以潜在指导基因特异性种系检测。许多基因检测实验室提供多基因(组合)检测,同时检测所有Lynch综合征相关基因(通常还有与遗传性癌症易感性相关的其他基因)的致病性变异。
结直肠肿瘤标本中存在MSI是Lynch综合征的标志性特征,可引起对种系致病性MMR基因变异的怀疑。微卫星是位于整个基因组中的短串联重复DNA(单核苷酸、二核苷酸、三核苷酸或四核苷酸)序列,主要位于内含子或基因间序列中。
在结直肠、子宫内膜或转移性肿瘤DNA与正常组织相比在微卫星区域显示插入或缺失时,
将使用术语MSI。MSI表示MMR基因中可能存在缺陷,可能是由于体细胞突变、种系变异或表观遗传学改变所致。
在大多数情况下,MSI与一种或多种MMR蛋白(MSH2、MLH1、MSH6和PMS2)的蛋白表达缺失相关。但是,并非在所有携带MSI的肿瘤中可见到蛋白表达缺失,而且并非所有在IHC上蛋白表达缺失的肿瘤均为微卫星不稳定性。
某些组织病理学特征强烈提示了MSI表型,包括存在肿瘤浸润淋巴细胞(参见图4)、克罗恩样反应、粘液组织学、无脏性坏死和组织学异质性。
结直肠腺癌微卫星不稳定的最初命名是基于在美国国立卫生研究院(NIH)共识会议上选择的一组3个二核苷酸和2个单核苷酸重复序列的不稳定基因座的特定百分比检测结果,称为Bethesda基因检测。 如果超过30%的肿瘤标志物不稳定,则评分为MSI-H;如果至少有一个但少于30%的标志物不稳定,则将肿瘤指定为MSI-低水平(MSI-L)。 如果基因座稳定,则将肿瘤指定为微卫星稳定(MSS)。 在Lynch综合征中的大多数肿瘤将是MSI-H。
MSI-L肿瘤的临床相关性仍存在争议;这些肿瘤与MMR基因种系致病性变异相关的概率极小。
最初的Bethesda基因检测已被5个单核苷酸重复序列的pentaplex基因检测取代,
提高了MSI-H肿瘤的检测水平。
(有关MSI检测的治疗意义的更多信息,请参见本总结“MSI的预后和治疗意义”章节。)
(有关疑似Lynch综合征的患者诊断检查中MSI状态利用的信息,请参见本总结“使用普遍肿瘤检测筛查Lynch综合征”章节。)
作为MSI的替代方法,IHC方法更便宜、更易理解、使用更广泛,出于这些原因,在大多数机构中已取代了基于聚合酶链反应(PCR)的MSI检测。使用针对MLH1、MSH2、MSH6和PMS2蛋白的单克隆抗体,
在结直肠或子宫内膜肿瘤(或转移部位)中采用IHC法进行蛋白表达水平检测。这些蛋白中任何一种单独表达缺失可能提示特定患者的特定MMR基因突变。
然而,某些蛋白可形成异二聚体(或具有其他结合伴侣),并导致IHC上表达的两种蛋白丢失。
MSI可导致核苷酸配对滑移(成环),其中引入单核苷酸错配。形成MMR蛋白的异二聚体来识别错误并结合这些位点的DNA。
例如,MSH2蛋白与MSH6蛋白复合形成MutSα,其主要作用是修复单核苷酸重复序列复制过程中可发生的单个碱基对错配和单个碱基对成环病变。在MSH6蛋白缺失的情况下,MSH2蛋白会与MSH3蛋白二聚化形成MutSβ复合物,该复合物可引发较大环出DNA错配修复,同时也具有一定的重叠活性来修复通常由MutSα修复的病变。
因此,当MSH2基因存在种系致病性变异时,肿瘤IHC可能不会同时表达MSH2和MSH6,因为后者需要与MSH2结合才能保持稳定。在这种情况下,如果在任一基因中均未发现致病性变异,则应考虑对EPCAM进行种系致病性变异检测(如果尚未纳入)。通过IHC检测MSH2和MSH6蛋白表达缺失且未发现MSH2或MSH6致病性变异的患者中,约20%的EPCAM存在种系缺失。
后一种机制约占所有Lynch综合征病例的5%。
EPCAM(TACSTD1)基因外显子9的一个等位基因缺失,紧邻MSH2起始位点的上游,方向相同,可导致MSH2启动子的转录通读和甲基化,随后在任何表达EPCAM的组织中MSH2沉默。EPCAM致病性变异的存在显示了类似的甲基化介导的MSH2丢失,这在许多家族中已有报道。
根据上述观察结果,在经CRC IHC检测发现MSH2蛋白表达缺失且无可检测的MSH2种系致病性变异的患者中进行了种系EPCAM检测,并且进行所有结肠癌基因组合检测中的MSH2检测。
对于这些基因中无变异的患者,肿瘤测序可能发现双体细胞MSH2突变。(如需了解更多信息,请参见本总结“和Lynch样或HNPCC样综合征”章节。)
同样,MLH1缺失(通过种系致病性变异或MLH1启动子的超甲基化)导致肿瘤中MLH1和PMS2蛋白均无表达。结直肠腺癌中DNA MMR蛋白最常见的异常IHC模式为MLH1和PMS2表达缺失。 PMS2和MLH1作为一种稳定的异源二聚体(称为MutLα)发挥作用。MutLα与MutSβ结合,引导新合成DNA链的切除修复。
MLH1功能缺陷导致MLH1和PMS2降解,而PMS2缺陷只对PMS2表达产生负面影响。 因此,MLH1和PMS2缺失表明MLH1突变(启动子超甲基化或种系变异),而PMS2表达缺失表明种系PMS2变异。 然而,在88例PMS2缺陷型CRC患者中,PMS2种系致病性变异检测后进行MLH1种系致病性变异检测,发现49例患者(74%)存在致病性PMS2变异,8例患者(12%)存在MLH1致病性变异。
83%的MLH1突变是错义变异,但两个亲属携带相同的MLH1变异,一例发生发生了两个保留了MLH1表达的肿瘤,携带了一个内含子变异体,导致跳过了外显子8。
因此,在PMS2表达孤立性缺失的CRC中,如果未发现PMS2种系变异,应寻找MLH1种系致病性变异。MSI和MSH2和MSH6蛋白表达缺失的肿瘤通常提示潜在的MSH2种系变异(提示MSH2致病性变异)。 与MLH1不同,MSH2缺失的MSI很少与启动子体细胞甲基化有关。
与MLH1和MSH2(均与其他蛋白二聚化或具有其他结合伴侣)不同,MSH6和PMS2的种系致病性变异导致这些特异性蛋白孤立性缺失(通过IHC法测定)。然而,来自MSH6致病性变异携带者的肿瘤显示MSI表型的频率可能低于MLH1和MSH2携带者(尽管存在一个无活性的DNA MMR系统),因为致病性错义变异不能完全消除蛋白表达,从而产生假阴性结果(通过IHC检测)。
一项研究报告通过结肠癌家族登记研究入组的MMR种系携带者的肿瘤检测结果,显示24例携带MSH6致病性变异的患者中有7例(28%)的肿瘤在IHC染色上显示正常的蛋白表达。更多信息提示对MLH1和MSH2致病性变异携带者进行IHC肿瘤检测,其中93%的MLH1携带者出现相关的MLH1蛋白表达缺失,96%的MSH2携带者出现MSH2蛋白表达缺失。
在某些情况下,肿瘤表现为MSI和/或IHC显示DNA MMR蛋白表达缺失,但未发现种系致病性变异。该疾病称为Lynch样(或HNPCC样)综合征,肿瘤表型主要是由于DNA MMR基因的双等位基因体细胞失活所致,而非致病性种系突变。(如需了解更多信息,请参见本总结的Lynch综合征相关综合征章节。)
蛋白表达缺失 | 通过IHC蛋白表达缺失预测种系MMR缺陷 | ||||
---|---|---|---|---|---|
MLH1 | MSH2 | MSH6 | PMS2 | EPCAM | |
MLH1/PMS2 | X | ||||
MSH2/MSH6 | X | X | |||
MSH6 | X | ||||
PMS2 | X | X | |||
MLH1 | X | ||||
MSH2 | X | ||||
IHC = 免疫组织化学;MMR = 错配修复。 |
需提醒人们认识到,MLH1启动子的超甲基化(一例局限于肿瘤的体细胞事件)可导致MLH1异常蛋白表达(通过IHC检测)。由于MLH1超甲基化,大约10%-15%的散发性CRC病例具有微卫星不稳定的肿瘤表型,且不可遗传。这些散发性MSI结肠癌
具有普遍过量的DNA甲基化(称为CIMP)。
(如需了解更多信息,请参见本总结前言章节“CIMP和锯齿状息肉途径”章节。)由于在Lynch综合征和散发性肿瘤中MLH1蛋白表达缺失(通过IHC检测),其预测种系MMR基因变异的特异性低于其他MMR蛋白,通常需要额外的分子检测来明确MLH1缺失的病因。
在68%的MLH1启动子超甲基化的CRC肿瘤中检测到BRAF致病性变异,而在Lynch综合征患者的CRC中检测到BRAF致病性变异非常罕见(如果曾有)。
上述信息表明检测到CRC中的体细胞BRAF V600E突变可能有助于排除一部分无需进行种系变异检测的患者。 因此,BRAF V600检测和/或MLH1超甲基化检测在所有类型Lynch综合征检测中的应用越来越广泛,以区分超甲基化引起的MLH1蛋白表达缺失和种系MLH1致病性变异。在排除患者进行种系检测时,进行上述区分也是一种符合成本效益的方法。
很少有MMR基因变异的患者其两个亲本等位基因中均携带这种变异在发现两个变异等位基因时,无论是纯合子还是复合杂合子,均称为双等位基因错配修复缺陷(BMMRD)或体质性错配修复缺陷(CMMRD)。在近亲血缘中,发生纯合子MMR基因致病性变体的BMMRD的可能,将不可避免地更高。在农村和其他地理和/或文化隔离的人口中,血缘关系的发生率可能更高。
肿瘤研究产生特征性异常结果。有关28例BMMRD患者的一系列研究显示
除肿瘤细胞外,17例脑肿瘤显示正常基质细胞中MMR蛋白染色缺失,这与Lynch综合征患者的肿瘤形成了鲜明对比,其中非肿瘤细胞保留正常染色。与通过IHC检测观察到的这一特征相反,基于PCR的MSI分析并不可靠,因为28个肿瘤中有20个为MSS。在MSI-H的肿瘤中基本均为结肠癌。
在BMMRD病例中PMS2基因明显过表达。 有人认为,在其他MMR基因中存在纯合子变异是一种产前致死状态,而当两个亲本等位基因中均表达时,PMS2表达减少的程度与生存期保持一致。
(如需了解有关BMMRD临床表型的更多信息,请参见Lynch综合征相关患病率、临床表现和癌症风险章节中的BMMRD章节。)
临床表型 | DNA MMR中致病性种系变异 | DNA MMR的体细胞失活 | 肿瘤表型 |
---|---|---|---|
Lynch综合症 | 存在于一个等位基因中 | 存在于一个等位基因中 | MSI |
伴有MLH1启动子超甲基化的散发性CRC | 不存在 | +BRAF | MSI |
BMMRD | 存在于两个等位基因中 | 不存在 | MSI(肿瘤和正常组织) |
Lynch样 | 不存在 | 存在于两个等位基因中 | MSI |
FCCX | 不存在 | 不存在 | MSS |
BMMRD = 双等位基因错配修复缺陷;FCCX = 家族性X型结直肠癌;MMR = 错配修复;MSI = 微卫星不稳定性;MSS = 微卫星稳定性。 | |||
改编自Carethers et al. |
虽然MLH1启动子的体细胞超甲基化为获得性且并不少见,但MLH1启动子超甲基化的实例已在种系中进行了描述,且通常与稳定的孟德尔氏遗传无关。MMR基因的体质甲基化最常发生在MLH1,在MSH2中少见,称为体质性。
体质表观突变(也称为原发性表观突变)是正常组织中的一种获得性突变,可使活性基因沉默或激活失活基因。
这种表观突变最常发生在母体等位基因中。在某些情况下,所有体细胞均受累,而在其他情况下,存在嵌合体的证据。原发性表观突变患者的肿瘤通常与其他典型的Lynch综合征种系变异携带者难以区分,包括发病年龄、肿瘤谱以及是否存在异常的MSI和IHC。由于这些并非以孟德尔式遗传,先前的肿瘤家族史是最少,对后代的某些风险不可预测。新发病例中的表突变似乎在配子发生的过程中被“抹去”,不会传递给下一代。报道了极为罕见的遗传性MLH1表突变病例。
在解读肿瘤中的分子改变并区分可能的主要表观突变病例与散发性MSI病例方面,是一个重大挑战。大多数MLH1表达缺失情况是由MLH1启动子的散发性超甲基化所致。 MLH1中最新体质表观突变
或遗传种系MLH1甲基化的罕见实例
对MLH1表达缺失相关的MSI解读增加一些复杂性。 与散发性MSI相似,原发性表观突变肿瘤显示MLH1启动子甲基化,也可能显示BRAF变异。如上所述,这些病例的癌症家族史往往极少或没有,与真实的散发性MSI相同。区分这类病例和散发病例可能需要分析正常组织(如血液或正常结肠粘膜)中是否存在MLH1甲基化,而这在真实的散发性病例中不存在,在常规Lynch综合征MMR致病性变异携带者中也不存在。
这种以MLH1为主的原发性表观突变与继发性表观突变不同,例如MSH2因上游EPCAM基因的遗传变异而发生甲基化时出现的表观突变。(更多信息请参见本总结的相关章节)
虽然许多分子病理学实验室可以同时评估MSI和IHC,但使用IHC检测作为MMR活性缺陷的初始筛查方法受到了青睐,因为其能轻松完成,成本效益更高。
部分依据是IHC提供的信息可能针对一个特定MMR基因(MLH1表达缺失除外)的种系基因检测,而非针对所有单独使用MSI指导的所有Lynch综合征相关MMR基因的综合检测策略。
虽然MSI检测因其预后和治疗意义最初在CRC患者的肿瘤学评估中受到青睐,但通过IHC检测可以更有效地指导Lynch综合征的筛查。
在所有最新确诊的CRC病例中使用MSI和/或IHC检测均可增加Lynch综合征初步筛查的灵敏度(不考虑诊断时年龄或癌症家族史),尤其对于MSH6和PMS2致病性变异携带者。这种方法比现有临床标准更敏感,因为许多Lynch综合征患者在年龄较大(> 50 岁)时被确诊,而且CRC家族史也较以前明显减少。许多专业组织推荐并广泛采用这种使用MSI或IHC检测的结直肠(和子宫内膜)肿瘤普遍检测。
在最新确诊CRC的患者中进行遗传风险评估和MMR基因变异检测可改善患者和处于风险的家庭成员的结局。 追溯到2009年,基因组应用于实践和预防评估(EGAPP,一种由美国疾病控制和预防中心(CDC)公共卫生基因组学办公室开发的项目)报告显示,有充分证据建议对最新确诊CRC的患者的Lynch综合征进行肿瘤筛查,以降低亲属的发病率和死亡率。
当前推荐采取MSI和IHC中任何一个作为特定的检测策略。
多项研究证明了普遍筛查Lynch综合征的可行性。来自一家机构的初步经验发现,在使用MSI和IHC筛查的1566例患者中,44例患者(2.8%)出现Lynch综合征。 对于每个先证者,随后平均有3个额外家庭成员确诊Lynch综合征。
随后对10,206例接受MSI/IHC检测的新发CRC患者进行汇总分析(作为4项大型研究的一部分),发现致病性变异检出率为3.1%。
该研究比较了用于诊断Lynch综合征的4种肿瘤检测策略:(1)对符合Bethesda指南中至少一项标准的所有患者进行检测;(2)对符合Jerusalem建议的所有患者进行检测;
(3)对70岁或70岁以下所有CRC患者,以及70岁以上符合Bethesda指南中至少一项标准的患者进行检测(4)对所有CRC患者进行普遍检测。
MSI肿瘤检测包括每个机构的个体化检测组,IHC包括在所有机构检测Lynch综合征相关的所有4个DNA MMR基因。对所有70岁或70岁以下确诊CRC的患者和70岁以上符合修订后Bethesda指南之一的患者进行肿瘤检测,灵敏度为95.1%,特异性为95.5%,诊断率为2.1%。该策略漏检了4.9%的Lynch综合征病例,但需要IHC/MSI检测的病例减少了34.8%,接受种系检测的病例比普通方法减少了28.6%。
考虑进一步按年龄(即70岁)对分子肿瘤检测进行分层的建议值得关注,因为其影响了普遍筛查策略的成本效益。
由于体细胞超甲基化导致的MLH1和PMS2缺失并不少见,并且随着CRC诊断时年龄增加,检测到的频率也随之增加。
因此,在MLH1和PMS2表达缺失(通过IHC检测)的情况下,建议进行额外的分子肿瘤检测,包括BRAF和MLH1超甲基化检测,从而减少转诊进行不必要种系基因检测的患者数量。 在一项基于人群的1117例CRC患者的研究证实,对通过IHC检测证实MLH1缺失的70岁或70岁以下CRC患者进行MLH1超甲基化分析的检测策略是经济有效的。
基于评价与Lynch综合征相关的4个MMR基因的蛋白表达水平的IHC肿瘤检测结果,通常逐步对CRC患者的Lynch综合征进行筛查。一项拟定策略总结见图6。该框架不包括同时评价多个癌症易感基因(多基因[组合]检测)的种系检测方法,这可能对选定的患者人群有用。(如需了解更多信息,请参见该总结的“多基因[组合]检测”章节。)
临床医生越来越多地利用肿瘤测序,以更个体化的方式推进治疗决策,特别是在转移性疾病患者中。一项多中心、基于人群的研究中招募的419例CRC病例研究,比较了CRC新一代肿瘤测序(NGS)检测Lynch综合征的性能与现有的筛查方案,包括MSI检测和IHC染色(BRAF p.V600E检测)。
通过种系DNA检测,将12名受试者确定为Lynch综合征携带者,经肿瘤测序鉴定无误,而MSI加BRAF检测和IHC加BRAF检测分别漏检了5例和6例Lynch综合征病例。 肿瘤测序的灵敏度高于IHC加BRAF检测(100%vs. 89.7%;P = 0.04)和MSI加BRAF检测(100%vs. 91.4%;P = 0.07),而所有策略的特异性相当(肿瘤测序为95.3%,IHC加BRAF为94.6%,MSI加BRAF为94.8%;P = 不显著)。 在46名已知Lynch综合征致病性变异的CRC携带者的验证队列中,肿瘤测序得到了相似的结果,并正确识别了100%的携带者。此外,作者报道了通过肿瘤测序确定的体细胞突变,从而强调了在283名受试者中的潜在治疗意义。 该研究表明,肿瘤测序是一种识别Lynch综合征的高效模式;然而,这种策略的成本效益仍有待确定。
2019年一项回顾性研究使用了来自一个大型、基于社区的综合美国卫生保健系统的数据,比较了通过通过对所有CRC的反射MMR IHC检测以确定Lynch综合征的年龄限制性筛查策略与无年龄上限的普遍筛查策略的诊断效能。
在70-75岁后,Lynch综合征的识别率显著下降,在80岁之后增量最小。在50岁或50岁以下确诊CRC的患者中,需要筛查以确定1例Lynch综合征病例的CRC数量为20例,但在71-80岁确诊CRC的患者中增加至208例,在80岁后确诊CRC的患者中增加至668例。
从Markov模型中获得结果,其中该模型纳入了结直肠癌、子宫内膜癌和卵巢癌的风险,以评估在70岁或70岁以下最新确诊CRC的患者中用于识别Lynch综合征的策略的有效性和成本效益。
模型中包含的策略主要基于临床标准、预测算法和肿瘤检测或预先的种系致病性变异检测,其次是定向筛查和降低风险的手术。 本研究的首选策略是首先进行HC检测,随后进行BRAF致病性变异检测。 通过这一策略,每个生命年获得的增量成本效益比为36,200美元。 在该模型中,每个先证者检测的亲属数量(3-4)是有效性和成本效益的关键决定因素。这些结果与EGAPP早期进行的分析结果相似,发现最具成本效益的方法是检测所有肿瘤中是否存在MSH2、MLH1、MSH6和PMS2蛋白表达缺失,然后根据缺失蛋白对MSH2、MLH1或MSH6进行靶向种系检测。如果不存在MLH1,则对BRAF变体阴性肿瘤进行检测。
NCCN 2019指南支持使用IHC和/或MSI和/或全面的肿瘤NGS组合检测或种系多基因(组合)检测对所有CRC进行普遍筛查。
与仅筛查70岁以下的人相比,所有人不分年龄的普遍筛查使得每一被挽救的生命年中的增量成本翻了一番。
该分析的作者得出结论,筛查70岁以下患者具有合理性,而根据支付意愿,不考虑年龄因素筛查所有患者也可以接受
然而,值得注意的是,这项研究的结论取决于在家族CRC索引病例中鉴定出一种生殖系MMR基因变体的基础上,进行种系检测(通过目前已知的途径)的高危亲属的数量。 在该模型中,为了达到认可的50,000美元成本效益阈值,至少需要对3-4个亲属进行检测。
这强调了医生与患者沟通、家庭沟通的重要性,以及确保在具有已知致病基因的Lynch综合征家族中更好地实施种系检测的必要性。(如需了解有关家庭沟通和Lynch综合征家族中基因检测摄取的更多信息,请参见本总结的“遗传性结肠癌综合征的社会心理问题”章节。)
另一项研究阐述了检测Lynch综合征相关基因致病性变异的成本效益,并评估了21种筛查策略,包括临床标准、临床Lynch综合征预测模型的使用和分子肿瘤检测。
该模型包括两个步骤:(1)测算新发现的Lynch综合征诊断数量;(2)测算在健康携带者中确认Lynch综合征所获得的生命年。在所有建模的策略中,用预测模型如PREMM(1,2,6)筛选先证者,然后采用IHC进行MMR蛋白表达和种系基因检测是最佳的方法,每获得一个生命年的增量成本效益比为35,143美元。对所有先证者进行种系基因检测是最有效的方法,但每获得一个生命年的成本为996,878美元。作者认为,在先证者中Lynch综合征筛查的最初步骤应利用预测模型,普遍检测和一般人群筛查策略均非具有成本效益的Lynch综合征筛查策略。
确立普遍肿瘤检测的年龄上限仍存在争议。一些专家支持仅对70岁以下CRC患者进行检测(仅对符合修订后Bethesda标准的≥70岁患者进行预留检测;采用该策略,5%的携带者将被漏诊)。
然而,也有人主张反对设置检测的年龄上限,因为通过级联筛查对年轻一代有潜在的好处,并有机会对发现携带已知家族致病性变体的个人进行更多的监测和其他预防性干预。
另一项成本效益分析是使用179名70岁或70岁以前确诊的子宫内膜癌患者的数据进行的,这些患者使用MMR-IHC和反射性MLH1启动子高甲基化进行筛查,其中7名Lynch综合征携带者(3.9%)被确定。
在确诊子宫内膜癌时,7名Lynch综合征先证者中仅1名的年龄为50岁或50岁以下。作者计算得出结果,在51-70岁时筛查确诊子宫内膜癌的妇女,额外获得了29.3个生命年(高于小于50岁确诊女性筛查中获得的45.4个生命年),筛查70岁或70岁以下所有诊断筛查与50岁或50岁以下诊断筛查的增量成本效益比为每生命年5252欧元。与使用Bethesda指南指导MMR和MSI检测的策略相比,对所有70岁或70岁以下女性进行基于肿瘤的普遍筛查也具有成本效益,每获得一个生命年的增量成本效益比为6668欧元。
在CRC和子宫内膜癌中普遍肿瘤检测的成本效益在很大程度上是由级联筛查的假设驱动,通过级联筛查,将识别、检测其他处于风险的家庭成员,从而随后使自身的癌症风险降低。
随着DNA突变分析的进步,种系基因检测的成本不断降低,包括通过多基因(组合)检测同时检测与恶性肿瘤相关的多个种系变异。因此,将需要使用更多种系检测相关的最新数据进行额外的成本效益分析。多基因(组合)检测可能成为未来更有利且成本效益更高的方法。
虽然全国范围内继续采用普遍筛查,但分子检测的采用和方法存在显著差异。2011年美国国家遗传咨询师协会的一项调查发现,超过25%的受访者在其中心实施了某种形式的普遍筛查。肿瘤筛查方法各不相同;53个中心中,对于最新确诊的结直肠肿瘤,34个(64.2%)开始仅行IHC检测,11个(20.8%)开始仅行MSI检测,8个(15.1%)开始即同时进行两种检测
2012年的一项调查表明,71%的美国国家癌症研究所(NCI)综合癌症中心正在常规进行某种形式的普遍筛查,但在社区医院癌症项目的随机样本中,利用率下降到15%。
由于对Lynch综合征普遍筛查的依从性可能较差(许多患者未被转诊进行基因评估和检测),因此利用Six Sigma概念框架进行了一项前瞻性质量改善研究,以改善在年轻的CRC患者中普遍基因筛查的实施情况
该研究的主要目的是增加早发性CRC患者(18-50岁)中MMR缺陷的肿瘤研究比例。除了在治疗点提供明确的提示,干预还涉及患者和提供者接受的教育。该研究表明,与干预前相比,干预后的12个月中,患CRC的年轻成年人中IHC检测率提高了21.5%。
对Lynch综合征进行基因检测相关报告的研究,重点关注根据家族史或临床特征选择有潜在Lynch综合征风险的患者和家庭。虽然普遍的肿瘤筛查越来越多地被用来鉴别,新诊断的可能有种系变异的患者,但是很少有研究调查了在普遍的肿瘤检测之后进一步行基因检测的情况。Lynch综合征普遍筛查的一个重要意义是,不会导致合适患者进行自动种系检测。在临床环境中,医疗保健团队进行更多的随访,以促进肿瘤筛查结果异常患者转诊至遗传咨询,从而提高基因检测的完成情况。
异常肿瘤筛查后患者基因检测完成情况的水平较高可能与遗传咨询师参与这一过程以披露筛查阳性结果、在肿瘤检测后提供咨询或便于转诊有关。
随后的遗传咨询需要病理学家、转诊外科医生或肿瘤学家和癌症遗传学服务机构之间的进行协调。举例说明,一项基于人群的筛选研究发现,仅54%的IHC缺陷肿瘤(BRAF致病性变异阴性)患者最终同意并继续进行种系MMR检测。
一家机构在CRC诊断后接受常规MSI和IHC检测的1100例患者中发现了21种致病性变异。该研究发现,当外科医生和遗传咨询师收到异常MSI/IHC结果的资料时,特别是在遗传咨询师在患者随访中发挥积极作用时,接受遗传咨询和种系MMR基因检测的比例显著增加
与通常在未获得患者事先许可的情况下进行的肿瘤检测相比,种系基因检测(如MMR致病性变异的种系检测)一般包括遗传咨询,在进行之前需要患者许可。一项美国癌症项目的现况调查(20个NCI指定的综合癌症中心和49个社区医院癌症项目)发现,对于所有或选定病例中在结肠癌诊断时进行MSI和/或IHC检测(作为标准病理学评价一部分)的患者,肿瘤检测前均不需要书面知情同意。
鉴于MMR致病性变异携带者中子宫内膜癌的患病率增加,对子宫内膜癌患者进行Lynch综合征筛查的共识与日俱增。
一项对所有子宫内膜癌进行肿瘤筛查的可行性和必要性的研究显示,最新确诊的患者中至少有2.3%(95%CI,1.3%–4.0%)患有Lynch综合症(不考虑诊断时年龄或癌症家族史)。
确诊Lynch综合征的13例病例中的8例为50岁或50岁以上,其中8例不符合已公布的Lynch综合征家族史标准,MSI漏检2例。由于子宫内膜癌的患病率增加以及本研究的结果,作者支持对子宫内膜癌进行Lynch综合征的普遍筛查。(有关对MMR蛋白表达进行IHC检测的更多信息,请参见该总结的IHC章节。)
对242例连续子宫内膜病例进行的另一项较小研究表明,缺乏体细胞MML1启动子超甲基化的MMR缺陷病例发生率为4.5%(11/242),其中包括4例(1.7%)发生种系MMR突变,4例(1.7%)发生两种体细胞MMR突变(通过NGS检测),另有2例(0.8%)发生无法解释的MMR缺陷。
该结果表明,对子宫内膜癌进行普遍的MMR肿瘤筛查将确定具有潜在Lynch综合征的患者和具有各种形式MMR缺陷的非Lynch综合征病例谱。
另一项研究前瞻性地评估了CRC和子宫内膜癌病例的IHC普遍筛查,不考虑诊断时年龄。
三级机构和社区,2011年至2013年间筛查了1290例CRC和484例子宫内膜癌病例。 该研究还计算了所有检测到种系致病性变异的患者的PREMM(1,2,6)和PREMM5评分。22%的子宫内膜癌和18.8%的CRC可见异常染色。排除因存在BRAF和/或MLH1超甲基化确定为散发性病例后,10.8%的CRC患者和6.6%的子宫内膜癌患者被转诊进行遗传咨询。24例患者(1.4%)确诊Lynch综合征,其中66%患有CRC。 在子宫内膜癌病例中Lynch综合征的总体检出率为1.7%,在CRC病例中总体检出率为1.2%。在阿姆斯特丹标准、Bethesda指南、PREMM(1,2,6)和PREMM5中,表现最好的模型是PREMM5,将检测到82%通过普遍筛查确定的病例。
在基于模型的模拟研究中检查了确诊子宫内膜癌的女性肿瘤检测的成本效益,并将IHC检测纳入以下情况:(1)50岁前诊断;(2)60岁前诊断;(3)诊断时任何年龄,存在患任何Lynch综合征相关癌症的FDR;(4)所有病例,不考虑诊断年龄和家族史。符合阿姆斯特丹II标准的女性或50岁前确诊的至少有一个伴任何Lynch综合征相关癌症FDR的女性,直接转诊进行遗传咨询和基因检测,而不进行IHC检测。据报告,在所有子宫内膜癌且具有一名患任何Lynch综合征相关癌症FDR的患者中,IHC检测MMR蛋白表达的策略在Lynch综合征检测中具有成本效益。
相对于成本最低的方案,这一方案获得的每生命年增量成本比为9,126美元,该方案是对所有50岁前确诊子宫内膜癌且至少有一名患Lynch综合征相关癌症的FDR的女性进行基因检测。在所有子宫内膜癌女性中,采用最具包容性的IHC检测策略时预期寿命最高(不考虑诊断时年龄或家族史),但每获得一个生命年最不利的增量成本比为648,494美元。NCCN建议用IHC和/或MSI(一种全面的肿瘤NGS组合检测)进行肿瘤检测,或对所有子宫内膜癌进行种系多基因(组合)检测。
尽管有这些建议,但对新诊断为子宫内膜癌的妇女进行普遍筛查的情况尚不清楚。
(如需了解有关Lynch综合征中子宫内膜癌的更多信息,请参见PDQ关于乳腺癌和妇科癌症遗传学总结。)
在所有肿瘤类型中使用MSI检测,已成为筛选对免疫检治疗有良好反应的病例的重要筛查工具。 这些结果可用于CRC以外的肿瘤中Lynch综合征的筛查。 一项研究评估了MSI在多种恶性肿瘤中的应用,并将其用作识别Lynch综合征的一种潜在手段,不考虑肿瘤类型。
在一项单中心研究中评估了超过15000例患者的50种以上癌症,对标记良好的肿瘤和匹配的正常DNA测序结果和配对生殖系MMR基因检测的数据用于确定MSI状态。使用软件工具确定MSI,以不稳定微卫星的百分比作为配对肿瘤-正常基因组测序数据的评分,并允许同时对MSI位点进行全面研究。 据报道,与使用PCR对5个单核苷酸微卫星病灶进行MSI检测相比,该方法在通常不筛查MMR-缺陷(dMMR)的癌症中更敏感。
在本研究中,MSI-H肿瘤中,大多数是CRC和子宫内膜癌,但38%(125/326)的MSI-H肿瘤和超过90%的中等水平MSI肿瘤为其他癌症类型。种系检测证实MSI-H和中等水平MSI的肿瘤中分别有16.3%和1.9%诊断为Lynch综合征,此外还有0.3%的病例缺乏MSI。重要的是,在所有出现MSI-H/中等肿瘤的Lynch综合征携带者中,50%患有原发性癌症(CRC或子宫内膜癌除外),其中许多恶性肿瘤与Lynch综合征无关。基于其癌症诊断或家族癌症史,在患有非典型Lynch综合征癌症的患者中,近一半患者未能满足Lynch综合征检测的临床标准。此外,在与Lynch综合征无典型相关性的癌症和具有种系PMS2变异的患者中时常观察到中等水平的MSI和MSS表型。 该研究支持了其他与Lynch综合征可变表型表达相关的结果,这些结果是基于改变的MMR基因及其广泛的相关恶性肿瘤,使得仅通过临床标准很难识别。 此外,研究者进一步分析了每个肿瘤中独特的基因变异特征,并将结果与观察到的MSI表型和种系MMR状态相关联,以提供一些间接数据,说明基因变异携带者的癌症是由Lynch综合征和MMR缺陷所致还是可能是偶然结果。这在评估与Lynch综合征的相关性尚不清楚且值得商榷的癌症(如乳腺癌和前列腺癌)方面具有一定作用。作者发现,在这个非常大的队列中,Lynch综合征的乳腺癌患者未出现MSI肿瘤,这支持了这样的假设,即这些患者的种系MMR基因变异可能只是偶然的发现,而非其癌症诊断的病因学。
对MLH1、MSH2、MSH6、PMS2和EPCAM种系致病性变异进行基因检测,有助于为受累的变异阳性患者和高危家庭成员制定适当的干预策略,其中许多人可能未受癌症影响。
如果在受累患者中发现致病性变异,则应向所有处于风险的家庭成员提供相同致病性变异的检测。家族中已确定的致病性变异检测结果为阴性的高危亲属患CRC或其他Lynch综合征相关恶性肿瘤的风险未增加,可以遵循适用于一般人群的监测建议。 携带家族性致病性变异的家庭成员则参考Lynch综合征的监测和管理指南。(有关更多信息,请参见该总结的“Lynch综合征治疗”章节。)
如果在受累的家庭成员中未发现致病性变异,则该患者的Lynch综合征检测结果被视为阴性。随着DNA测序技术的进步,目前的基因检测不够灵敏,无法检测到待测基因中的致病性变异。随着检测到某些改变(如大片段缺失或基因组重排)以及PMS2中存在假基因PMSCL,检测技术(包括大多数商业检测实验室常用的NGS)不断进展。
可能无法检测到致病性变异的原因包括:
未能检测到致病性变异可能意味着该家族确实不存在遗传风险,尽管临床表现表明存在遗传基础(例如,患者可能存在MMR基因的双体细胞突变)。 如果在受累的家庭成员中无法确定变异,则不应向高危成员提供检测,因为结果对亲属无意义。 由于家族史,患者患结直肠癌的风险仍然增加,应继续进行推荐的密切筛查。
(有关更多信息,请参见该总结的“Lynch综合征治疗”章节。)
在无法从患者获得肿瘤组织进行MSI和/或MMR蛋白IHC检测的情况下,可考虑进行MLH1、MSH2(包括EPCAM)、MSH6和PMS2的种系突变分析。随着多基因(组合)检测的出现,这种方法变得更加便宜,现在多个临床实验室提供了这种检测,其成本可能与单基因检测相当。多基因检测的成本也可能接近肿瘤筛查的成本,研究证明是CRC患者的一种具有成本效益的方法。目前,在所有最新确诊CRC患者中,多基因检测并不常规推荐用于Lynch综合征的普遍筛查,但在选择性的人群中可能非常有用,如早发性CRC患者
或来自家族性、高危临床人群。值得注意的是,在Lynch综合征以外的其他癌症相关基因中可能检测到致病性变异。一项对1112例符合NCCN Lynch综合征检测标准并接受25个基因组合的多基因检测的患者进行的研究显示,发现114例患者(9.0%)的MMR基因存在致病性变异;然而,发现71例患者(5.6%)的非Lynch综合征癌症易感基因存在致病性变异,如BRCA1、BRCA2、APC、MUTYH(双等位基因)和STK11,与预期结果一致。最后,多基因检测产生了高比例的VUS。在上述研究中,总计479例患者(38%)出现一次或多次VUS。
已证明早发性CRC患者具有高频率和广泛的种系致病性变异谱,表明在该人群中进行组合检测可能获益。一项针对450例早发性CRC(诊断时平均年龄42.5岁)且有家族史(包括至少一名患结肠癌、子宫内膜癌、乳腺癌、卵巢癌和/或胰腺癌的FDR)患者的研究显示,在72例患者(16%)中发现了75种种系致病性或潜在致病性变异。
发现的变异谱包括Lynch综合征和非Lynch综合征相关基因,包括一些传统上与CRC无关的基因(如BRCA1/BRCA2、ATM、CHEK2、PALB2和CDKN2A)。考虑到已确定的遗传性癌症综合征的高频率和多样性,作者认为有必要在该人群中进行多基因检测。同样,对151例CRC患者进行的另一项较小单机构分析显示在9.9%患者中发现了致病性种系变异。
在一项更大规模的研究中,还对1058例全部结直肠癌患者进行了多基因检测,而不考虑诊断年龄、个人或家族史或MSI/MMR检测结果。
在105例患者(9.9%)中发现癌症易感基因的种系致病性变异。而33例患者(3.1%)携带Lynch综合征基因的致病性变异,74例患者(7.0%)携带非Lynch综合征相关基因的致病性变异,包括APC、MUTYH、BRCA1/、PALB2、CDKN2A、TP53和CHEK2。这些数据表明在未经选择的CRC患者中可能发现变异的广度;因此,需使用全面的多基因检测。
2017年的一项研究检测了2012年至2015年间在美国单个商业实验室进行多基因检测的患者中致病性Lynch综合征相关基因变异的频率,并报道了确定为Lynch综合征携带者的特征。
该研究报告了迄今为止通过多基因检测的最大患者队列;共包含34,980例患者的相关数据,这些患者接受了包括MMR和EPCAM基因在内的各种多基因组合检测,其检测的适应症不限于Lynch综合征。 在612例患者(1.7%)中共发现618种致病性变异,并对其中579例受试者进行了分析(排除了33例具有Lynch综合征相关变异和第二种MMR变异或其他癌症易感基因中其他致病性变异的受试者)。大多数携带者受癌症累及,包括非Lynch综合征相关恶性肿瘤,其中乳腺癌发生率最高(124/423,23.5%)。 MSH6变异最普遍(29.3%),其次为PMS2(24.2%)、MSH2(23.7%)、MLH1(21.6%)和EPCAM(1.2%)。这一发现与先前数据不同,先前认为MSH2和MLH1变异更普遍,这是因为由于CRC的个人和/或家族史,患者被选择进行Lynch综合征特异性检测的频率更高。
该研究报道了528名Lynch综合征携带者的基因型-表型相关性,其中大多数患有CRC(186例,35.2%)和子宫内膜癌(136例,25.8%),其次是乳腺癌(124例,23.5%)和卵巢癌(74例,14%)。
145例携带者表现为乳腺癌或卵巢癌,作为前哨肿瘤,并且在多基因检测之前未事先诊断CRC或子宫内膜癌。当检测MMR基因变异在肿瘤特异性亚组中的分布时,仅在乳腺癌携带者中检测到的MSH6和PMS2变异发生率高于MLH1和MSH2,其中仅CRC受试者中检测到的后者致病性变异发生率更高。 仅对于乳腺癌患者,PMS2基因变异的发生率显著高于人群估计值,而MLH1、MSH2或MSH6并非如此。一项类似的回顾性研究报告了相似的结果。比较423例MMR基因致病或可能致病性变异女性人群与普通人群中观察到的乳腺癌发生率,从而计算乳腺癌的标准化发病率比(SIR)。作者报道MSH6携带者(SIR=2.11;95%CI,1.56-2.86)和PMS2携带者(SIR=2.92;95%CI,2.17-3.92)的乳腺癌年龄标准化风险具有统计学意义。
上述两项研究的一个关键局限性是整体转诊人群中乳腺癌病例过多,以及已知的MSH6和PMS2种系致病性变异的高背景人群患病率。
识别Lynch综合征的临床标准,包括阿姆斯特丹标准、修订后Bethesda指南或PREMM(1,2,6)风险预测模型,在本研究中未能识别27.3%的Lynch综合征携带者。
考虑到乳腺癌和卵巢癌的患病率增加,58.9%符合NCCN BRCA1/BRCA2检测指南,其中36.7%也符合NCCN Lynch综合征检测指南。最后,仅在18.8%的致病性变异携带者中获得数据,因此关于肿瘤检测结果的数据有限,其结果通常与改变的基因不一致,常见于MSH6和PMS2携带者。该研究的结果支持对Lynch综合征进行多基因检测,并进一步研究相应的癌症风险,因为目前的检测方案限制了对Lynch综合征携带者和相关恶性肿瘤的识别。
最后,在接受与Lynch综合征无关的癌症(如乳腺癌和前列腺癌)多基因检测的患者中意外检测到种系MMR基因。因此,与Lynch综合征相关的癌症谱可能比以前所认识的更广泛。(更多信息请参见本总结的乳腺癌和前列腺癌章节以及前列腺癌遗传学总结。)
(如需了解多基因检测的更多信息,包括遗传教育和咨询考虑,以及检查多基因检测使用率的研究,请参见PDQ癌症遗传学风险评估和咨询总结中的多基因[组合]检测章节。)
随着基因检测成为常规而非例外,关于检测成本的问题不可避免。在过去,每质量调整生命年(QALY)的成本效益比为50,000美元,这是良好护理价值的基准。
随着时间的推移,有人提出,这一阈值太低,可采用其他阈值,如100,000美元或150,000美元。
2015年的一项研究评估了在癌症遗传学诊所就诊的患者中进行CRC和息肉综合征多基因检测的成本效益。
这些作者建立了一个决策模型来估计转诊进行评估的患者的直接和后续成本,并在确定为致病性变异携带者的家庭成员中进行CRC监测。根据CDC和学术性分子遗传学实验室发表的模型估算成本。根据CRC的遗传模式和外显率对综合征进行分类。将4种自定义检测与标准治疗进行比较。进行了4种组合检测:(1)仅Lynch综合征相关基因(MLH1、MSH2、MSH6、PMS2和EPCAM);(2)组合检测1中的基因和与常染色体显性遗传和CRC外显率高相关的其他基因(APC、BMPR1A、SMAD4和STK11);(3)组合检测1和2中的基因以及与具有高CRC外显率(MUTYH)的常染色体隐性遗传相关的基因;或(4)前三个组合检测中的所有基因以及与具有低外显率的常染色体显性疾病相关的基因(PTEN、TP53、CDH1、GALNT12、POLE、POLD1、GREM1、AKT1和PIK3CA)。相应的费用如下所示:组合检测1,每QALY 144,235美元;组合检测2,每QALY 37,467美元;组合检测3,每QALY 36,500美元;组合检测4,每QALY 77,300美元(与组合检测3相比)。作者得出结论,使用NGS多基因检测(包括高外显率CRC和息肉综合征以及Lynch综合征癌症基因)是最有可能提供具有成本效益及临床意义结果的方法。
自进行该模型分析以来,种系基因检测的成本随着技术的进步而持续降低;需要额外的研究来继续评估该检测方法的成本效益。
Lynch综合征是一种常染色体显性遗传综合征,其特征为CRC发病年龄早,过多的同步和异时性结直肠肿瘤,右侧为主,结肠外肿瘤,特别是子宫内膜癌。Lynch综合征是由以下DNA MMR基因的致病性变异引起,即染色体3p21上MLH1(mutLhomolog1);
染色体2p22-21上MSH2(mutS同源物2);
染色体2p16上MSH6
和染色体7p22上PMS2(减数分裂后分离2)。
这些基因的功能是在复制过程中维持DNA的保真度。 Lynch综合征还与染色体2p21上EPCAM(上皮细胞粘附分子,前称TACSTD1)基因的致病性变异有关,引起位于该基因下游的MSH2的表观遗传沉默。
Lynch综合征约占所有最新确诊CRC病例的3%。
在早期的研究中,Lynch综合征致病性变异携带者在诊断CRC时的平均年龄为44-52岁
而散发性CRC诊断时为71岁。
在随后的研究中,校正了确定偏倚以确定癌症相关风险评估和基因型-表型相关性,据报道,Lynch综合征相关致病性变异携带者中诊断CRC的平均年龄为61岁。
与Lynch综合征总体和基因特异性患病率估计值相关的原始报告在很大程度上依赖于全球家族性癌症登记研究的回顾性数据。Lynch综合征中报告的早期CRC(和子宫内膜癌)风险预估值存在确定偏倚和高估,因为数据主要来自家族性癌症登记研究,病例的确定通常基于年轻发病的CRC或亲属中CRC病例的增加。通过改良的分离分析,可以校正这些癌症风险预估值,其中统计方法提供了更准确的预估值,并调整了确定偏倚。相反,可能易低估与结肠外恶性肿瘤相关的风险预估值,除了子宫内膜癌,因为许多家庭可能低估了亲属中的这些癌症,且Lynch综合征相关的肿瘤可能会较晚发生。
在一项对来自美国、澳大利亚和加拿大的5744例CRC病例(不考虑家族癌症史)进行的基于人群的大型研究中,估计人群中每279例患者中就有1例携带与Lynch综合征相关的MMR致病性变异。
在另一项针对450例CRC患者(但仅限于50岁前诊断的年轻患者)的基于人群的研究中,通过多基因(组合)检测遗传性癌症易感基因,在450例患者的72例(16%)中发现了种系致病性变异。如预期,在已知与CRC相关的基因中发生大多数已确定的变异,主要是Lynch综合征(72例患者中的37例,51.4%)。然而,72例患者中13例(18.1%)存在传统上与CRC无关的基因致病性变异,包括但不限于BRCA1/BRCA2,其占已确定变异的8%。 由于发现了高频率和广泛的致病性变异,作者建议考虑对所有早发性CRC患者进行多基因检测。
最初认为MLH1和MSH2基因是Lynch综合征中发现的MMR基因的大多数致病性变异。然而,随着DNA突变分析和所有CRC的普遍肿瘤筛查的改进,MSH6和PMS2致病性变异的发生率不断增加。
MSH6和PMS2变异可能在未经选择的CRC(和子宫内膜癌)病例中更常见,
与MLH1和MSH2变异相比,MLH1和MSH2变异在高危CRC诊所的患者中更常见。
来自前瞻性Lynch综合征数据库(PLSD)的多篇论文描述了前瞻性随访(主要是欧洲)登记研究患者的癌症结局。 其中关键发现是PMS2携带者患CRC的风险较低,尤其是在50岁以下的人群中,因此作者得出结论,PMS2携带者的监测强度可以安全地降低。 根据这些发现,正在逐渐在后期开始结肠镜检查(可能间隔时间更长)。
PMS2携带者患结肠外癌症的相对风险不大于或仅略高于人群预期,这使得作者普遍建议不要对PMS2携带者进行任何外结肠监测。
上述数据共同支持了一种更自由化的筛查PMS2携带者的方法,尽管目前的临床实践指南并未反映这种变化。
筛查PMS2致病性变异患者的方法,以及在较小程度上筛查MSH6致病性变异患者的方法,是正在进行的临床争论的问题。
在早期研究中,Lynch综合征患者中MLH1致病性变异的发生率为41.7%-
50%,
使MLH1成为Lynch综合征家族中最常见的改变的MMR基因。直到有关基于人群的Lynch综合征发生率报道,我们才发现MLH1致病性变异估计为1/1946,位居第三,仅次于PMS2(1/714)和MSH6(1/758),正如在一项纳入5744例CRC病例的大型国际研究中所估计。
MLH1致病性变异与Lynch综合征相关的整个恶性肿瘤谱相关。
在MLH1致病性变异携带者中,发现到70岁时患任何Lynch综合征相关癌症的终生风险为59%-65%。
致病性MLH1变异携带者中风险最高的是CRC,估计发生率为41%-68%,
在一项纳入137例受累患者的研究中,CRC诊断时的平均年龄为42.8岁(范围:16-81岁)。
在最近的一项前瞻性研究中,使用了944例无癌症的MLH1携带者的汇总欧洲登记数据,尽管进行了结肠镜监测(尽管间隔不同),但70岁时CRC的累积发病率为46%。
在患Lynch综合征的患者或家族中MSH2致病性变异的发生率在各研究中各不相同。在包括大型癌症登记研究和早发性CRC(55岁以下)队列研究中,38%-54%的Lynch综合征家族报告了MSH2致病性变异。
国际胃肠道遗传性肿瘤学会(InSiGHT,一个致力于家族性GI癌合作研究的大型专业组织)数据库中2012年报道的MSH2致病性变异的发生率为32.8%,
基于MSH2相关Lynch综合征中存在结肠外癌症,易确定该家族。然而,在美国、澳大利亚和加拿大招募的5,744例基于人群的CRC病例队列中,MSH2致病性变异的发生率估计为1/2841;
在与Lynch综合征相关的MMR基因变异中MSH2发生率最低。
在MSH2致病性变异携带者中,发现到70岁时患任何Lynch综合征相关癌症的风险为57%-近80%。
与MSH2致病性变异相关的结肠癌终生风险估计为48%-68%。
在Lynch综合征患者的病例分析中,携带种系MSH2致病性变异的患者(49例患者,女性占45%)患结肠外癌症的终生(截止年龄,60岁)风险为48%,而MLH1携带者为11%(56例患者,女性占50%)。
在最近的一项前瞻性研究中,使用了616例无癌症的MSH2携带者的汇总欧洲登记数据,尽管进行了结肠镜监测,但70岁时CRC的累积发病率为35%。
MSH2携带者确诊CRC的平均年龄与MLH1携带者相当。一项包括143例MSH2致病性变异的受累患者的研究发现CRC诊断时的平均年龄为43.9岁(范围:16-90岁)。同一研究报道137例MLH1致病性变异携带者中CRC诊断时的平均年龄为42.8岁(16–81岁)。
大多数研究报道了来自高危诊所的约10%的Lynch综合征家族中种系MSH6致病性变异的发生率,以及较高比例的非选择性的CRC患者(约50%)的发生率。
2012年InSiGHT数据库中报告的MSH6致病性变异发生率为18%。
致病性MSH6变异的广泛发生率估计值是由于以下原因所致:相对于MLH1和MSH2相关Lynch综合征家族,MSH6相关Lynch综合征家族的样本量小、确定偏倚、CRC发病年龄较晚和家族史不太显著。
这与一项基于人群的研究结果一致,该研究纳入了42例有害MSH6种系致病性变异携带者,其中30例(71%)有不符合阿姆斯特丹II标准的家族癌症史。
在最近一项对5744例CRC病例进行的国际人群研究中,MSH6致病性变异的发生率估计为1/758,居MMR基因变异发生率第二位,仅次于PMS2。
MSH6致病性变异携带者中任何Lynch综合征相关癌症的终生风险约为25%,
CRC终生风险估计为12%-22%
MSH6携带者确诊CRC的年龄晚于MLH1和MSH2携带者。一项早期研究纳入来自20个家庭的146名MSH6携带者(59名男性和87名女性),其中这些携带者存在MSH6截断致病性变异,在MLH1、MSH2和MSH6携带者中,到70岁时CRC的发生率相似(P=0.0854)。然而,确诊结直肠癌时的平均年龄为,(a)男性MSH6携带者为55岁(n=21;范围,26-84岁),而MLH1和MSH2致病性变异携带者分别为43岁和44岁;(b)女性MSH6携带者为57岁(n=15;范围,41-81岁),MLH1和MSH2致病性变异携带者分别为43岁和44岁。
迄今为止报道的最大型MSH6致病性变异携带者相关研究纳入来自5个国家的113个家庭,通过家庭癌症诊所和基于人群的癌症登记确定研究。
与普通人群的发生率相比,MSH6致病性变异携带者的CRC发生率增加了8倍(风险比[HR],7.6;95%CI,5.4-10.8),不考虑性别和年龄。到70岁时,MSH6致病性变异男性携带者的CRC发生率为22%(95%CI,14%-32%),而女性携带者发生率为10%(95%CI,5%-17%)。 到80岁时,确诊CRC的MSH6致病性变异男性携带者的CRC发生率翻倍,达44%(95%CI,28%-62%),而女性携带者为20%(95%CI,11%-35%)。
在最近的一项前瞻性研究中,使用了305例无癌症的MSH6携带者的汇总欧洲登记数据,尽管进行了结肠镜监测,但70岁时CRC的累积发病率为20%。
PMS2是MMR基因家族中最后一个发现的基因。这是因为家庭中的外显率较低,从而难以使用临床标准进行识别,
也因为假基因干扰导致的DNA突变分析的局限性。
在早期对CRC和疑似Lynch综合征患者的研究中,PMS2致病性变异的发生率从2.2%到5%不等,
2012年InSiGHT数据库报告显示增至7.5%。
在瑞士一项对未经选择的结直肠癌病例的普遍肿瘤检测结果进行检查的研究中,1000例连续病例的IHC评价发现,在所有肿瘤中1.5%孤立地不表达PMS2。如果PMS2缺陷型CRC的发生率代表了所有PMS2相关的Lynch综合征,则PMS2将是与Lynch综合征相关的最常见基因。
来自一个基于人群的大型CRC队列的结果发现,在所有MMR变异中,PMS2致病性变异的发生率最高,其中714人中有1人携带致病性PMS2基因变异。
对于杂合的PMS2致病性变异携带者,发现任何癌症的终生风险为25%-32%。
对3项基于人群的研究和一项基于临床的研究进行的荟萃分析估计,对于PMS2致病性变异的携带者,男性在70岁前患CRC的风险为20%,女性为15%,患子宫内膜癌的风险为15%。
同样,一个欧洲的临床登记联盟重点关注纠正确认偏倚,发现PMS2致病性变异的男性和女性的累积终生(至70岁)CRC风险仅为19%和11%。
此外,PMS2致病性变异的患者出现CRC的时间比MLH1和MSH2致病性变异患者晚7-8年。
在一项前瞻性研究中,使用了77例无癌症的PMS2携带者的汇总欧洲登记数据,尽管进行了结肠镜监测,但70岁时CRC的累积发病率为10%。
对来自欧洲联合会的284个PMS2家族的近5,000例患者进行分析,并辅以另外两项登记研究的数据,旨在提供更稳健的PMS2相关癌症风险估计。
与一般人群风险估计值6.6%和4.7%相比,男性在80岁前患CRC的风险为13%(95%CI,7.9%-22%),女性为12%(95%CI,6.7%-21%)。 发现子宫内膜癌风险为13%(95%CI,7%-24%)。 在这些队列中未发现其他Lynch综合征谱系肿瘤的额外风险。 作者得出结论,这些数据证明了考虑延迟开始结肠镜检查(直至35-40岁)以及更长随访间隔(2-3年)的合理性,尽管未对此进行特定研究。 与来自欧洲前瞻性Lynch综合征数据库的原始报告相同,不可能评估这种结肠镜检查和息肉切除术降低CRC检出率的程度。
PLSD是一项正在进行的评估Lynch综合征癌症风险的重大举措。 尽管其缺乏关于筛查实践的具体细节,但包括来自许多欧洲项目的结局数据,按年龄、性别和MMR基因进行分类。
由于认识到较大PLSD的局限性,提供了具有更详细监测数据的子集。
这些来自德国、荷兰和芬兰的前瞻性结肠镜检查数据包括2,747例患者,其中62例在监测开始时既往无癌症。 由于监测实践的差异,德国的结肠镜检查间隔约为1年,荷兰约为2年,芬兰约为3年。 结肠镜检查的中位次数为5次,每例患者的中位观察时间约为8年。 尽管监测间隔存在差异,但在有癌症史(14%)和无癌症史(15.6%)的患者中发现了相似的腺瘤检出率。 在10年随访时,异时性肿瘤的首次癌症检出率为8.4%和14%。 增加风险的因素包括男性、既往CRC、存在MLH1或MSH2致病性变异、年龄大于40岁和结肠镜检查时检出腺瘤。 值得注意的是,在1年、2年或3年的筛查间隔之间,未观察到CRC检测或检测时分期的显著差异。
值得注意的是,在双等位基因PMS2致病性变异的携带者中观察到更严重的表型。(如需了解更多信息,请参见该总结的Lynch综合征遗传学章节中的BMMRD章节。)
这些致病性变异携带者患CRC和子宫内膜癌的终生风险总结见表11。
基因 | 结直肠癌的终生风险(%) | 子宫内膜癌的终生风险(%) |
---|---|---|
MLH1 | 41-50 | 34-54 |
MSH2 | 35-56 | 21-51 |
MSH6 | 10-22 | 16-49 |
PMS2 | 10 | EPCAM |
Lynch综合征患者子集(约1%)具有EPCAM致病性变异,从而导致MSH2启动子超甲基化和失活。
在一项针对194例EPCAM缺失携带者的欧洲研究中,70岁前患CRC的累积风险为75%,平均发病年龄为43岁。这与MSH2携带者的风险相当(到70岁时高达68%)。然而,在该研究中,EPCAM缺失的女性患子宫内膜癌的风险仅为12%,而MSH2携带者的风险高达71%。
相关表型取决于EPCAM基因3’端缺失变异的位置;如果缺失较大且包括MSH2的部分启动子,则表型将与其他MSH2相关Lynch综合征家族相似。
当缺失涉及EPCAM的终止信号但保留所有MSH2基因和启动子时,表型主要局限于CRC。
具有相同的仅限于基因的3’端且未延伸到MSH2的启动子的EPCAM缺失的两个家族
然而,随后的一项研究证明,由EPCAM变异引起的MSH2蛋白表达缺失的女性也有患子宫内膜癌的风险。
如上所述,患者可能在两个亲代等位基因中均携带MMR基因变异,这种情况称为BMMRD。(如需了解更多信息,请参见该总结的Lynch综合征遗传学章节中的BMMRD章节。)
这种双等位基因变异与一个特征性但不是诊断性的临床表型有关。临床特征包括儿童血液系统恶性肿瘤和脑肿瘤。当GI肿瘤发生时,发病年龄显著较低,有时在20岁之前。Café au lait spots和其他提示神经纤维瘤病的特征具有特征性。偶有患者表现为多发性腺瘤。
不同人群之间MMR变异的发生率无显著差异,在许多不同国家发现发生率相似。 与遗传性乳腺癌和卵巢癌(HBOC)一样,在特定种族中存在某些发生率较高的变异。 在HBOC中值得注意的是经常重复出现Ashkenazi犹太人变异,这种情况非常普遍,因此可以对这些常见变异进行直接面向消费者的检测。(如需了解更多信息,请参见PDQ乳腺癌和妇科癌症遗传学总结中BRCA1或BRCA2致病性变异概率的人群预估值章节,以及PDQ癌症遗传学风险评估和咨询总结中直接面向消费者(DTC)基因检测章节。) 通常通过单倍型分析确定表观建立者变异的精确性。 在某些情况下,建立者变异只是一种频繁复发的新生变异。
关于Lynch综合征MMR基因的首项人群研究结果显示,芬兰发现了两种十分常见的MLH1变异,占该国Lynch综合征病例的大多数。
从那以后,在大多数人群中发现了先证者变异,其中相对未经选择的CRC患者接受了变异检测。 许多报告来自欧洲。 与芬兰一样,在移民人数较少的种族相当同质的情况下,这些可能很容易确定。已在英国、瑞典、瑞士、意大利、
葡萄牙、法国、西班牙和匈牙利发现了欧洲的先证者变异,且可能存在于所有种族中。 来自亚洲,
拉丁美洲、中东和非洲的此类报告较少。
在美国,MSH2基因外显子1-6缺失估计占该基因变异的20%之多。 这种所谓的美国先证者突变可通过单倍型分析进行确定,这种变异可追溯到大约500年前。
一项南美研究结合了乌拉圭、哥伦比亚、巴西、阿根廷和智利的数据,还根据阿姆斯特丹和Bethesda特征选择了相关的病例,其中MLH1的发生率为60%,MSH2的发生率为40%。 未评价MSH6和PMS2。 选择偏倚可能影响变异的发生率,并可能影响MLH1和MSH2的相对作用。有人指出,哥伦比亚可能存在先证者变异。
尽管在采用逐步方案的情况下,认为在特定种族/地理区域中对常见的先证者变异进行检测是产生成本效益的第一步,但当采用越来越普遍的广泛组合检测方法作为基本方案时,可能没有必要。
与种族有关的一个考虑因素是某些人群中血缘关系的增加以及随之而来的BMMRD风险。 (如需了解更多信息,请参见本总结双等位基因错配修复缺陷[BMMRD]章节。)
在本节中,列出了探索MMR基因变异在美国不同种族中分布的数据。 考虑到存在选择和确认偏倚,这些研究的解释具有挑战性。 此外,即使是基于人群的研究也受到许多种族的小样本量和种族/人种自我报告的限制。
很少有数据表明,根据地理或种族,Lynch综合征发生率存在很大差异。 在一个较小和/或同质的种族群体中,先证者变异的存在可增加该特定基因变异的发生率。 MLH1和MSH2变异的比例在不同人群之间存在轻微差异。 尚未在群体水平下对MSH6和PMS2进行充分研究,因此无法对其相对发生率进行推断。
美国最具代表性的基于人群的研究,如俄亥俄州哥伦布的研究,以白人为代表,但总数更高。 因此,因此,像西班牙裔和非洲裔美国人这样的少数民族的数据受到的影响较小,代表性不强。
在波多黎各进行的一项研究根据阿姆斯特丹标准或Bethesda指南,考虑了89例疑似Lynch综合征的加勒比西班牙患者的变异。
患者接受立即的种系检测或从肿瘤MSI/IHC开始逐步评价。 MSH2、MLH1和MSH6的基因变异发生率分别为67%、25%和8%。 无明显的建立者变异。 很明显,根据临床家族史标准选择受试者可能导致低外显率的MSH6和PMS2基因的漏报。
来自加利福尼亚州、德克萨斯州和波多黎各的临床研究显示,总体变异发生率与所描述的相似,MLH1略多于MSH2,但也包括MSH6和PMS2。有人认为存在可追溯至西班牙和欧洲的潜在先证者变异。
关于拉美裔Lynch综合征的最接近人群的信息是基于加利福尼亚肿瘤登记中心进行的南加利福尼亚州研究,其中确定了265例患者。
在携带MSI-H肿瘤的患者中,13例(62%)出现MMR变异。MLH1、MSH2、MSH6和PMS2的MMR变异发生率分别为46%(6/13)、31%(4/13)、15%(2/13)和8%(1/13)。
在美国进行的更真实的基于人群的研究的结果重点强调了数量较少的问题。 在俄亥俄州哥伦布的一项研究中,仅8%的连续系列患者为非裔美国人,未指明西班牙裔作为白人亚组的比例。
在另一项涉及在Dana-Farber癌症研究所接受治疗的几乎所有CRC患者的组合检测的研究中,5%以下为非裔美国人,3%以下为西班牙裔,这突出了从小样本中提取有意义数据的挑战。
在非洲裔美国人中评估与MMR变异相关的Lynch综合征患病率和癌症风险的问题与西班牙裔(一组研究不足的异质性人群)相似。 对美国13个转诊中心临床研究数据进行研究,确定了51个Lynch综合征家族,MMR基因变异发生率如下所示:61%MLH1、21%MSH2、6%MSH6和12%PMS2。癌症发作年龄分布曲线与白人人群非常相似。
与大多数对西班牙裔的研究一样,没有根据任何一致的、程序性的评估(如普遍的肿瘤检测)来确定病例。
Lynch综合征的一个标志性特征是致病性MMR基因变异携带者发生同步和异时性结直肠肿瘤的风险增加。一项对来自结肠癌家族登记的382例Lynch综合征患者进行的研究显示,节段性结肠切除术后10年、20年和30年的异时性CRC发生率分别为16%、41%和63%。
每切除10 cm结肠,异时性CRC的风险逐步下降31%,50例行广泛结肠切除术的患者均未确诊异时性CRC。 另一项对1273例既往患有癌症的Lynch综合征患者进行的前瞻性研究显示,MLH1携带者随后CRC的累积发生率为46%,MSH2携带者为48%,MSH6携带者为23%。这只比既往没有诊断为癌症的致病性变异携带者患新发癌症的风险稍高。我们再次看到良好的生存率,这归因于有利的肿瘤病理学和监测效果的结合。
Lynch综合征患者患其他癌症的风险增加,尤其是子宫内膜癌。据估计,86个家庭中1018名女性在70岁时患结肠外癌症的累积风险为20%,而一般人群为3%。
某些证据表明,患者癌症的发病率因亲缘关系而异。
Lynch综合征中最常见的结肠外恶性肿瘤是子宫内膜腺癌,在约50%的Lynch综合征家族中至少有1名女性成员受累。此外,50%携带MMR基因致病性变异的女性其首发恶性肿瘤为子宫内膜癌。
据估计,MLH1致病性变异携带者患子宫内膜癌的终生风险为44%,MSH2致病性变异携带者为71%,尽管一些早期的研究可能由于确定偏倚而高估了风险。
在70岁时,113个家庭中MSH6致病性变异携带者患子宫内膜癌的终生风险估计为26%,在80岁时估计为44%;
总体而言,MSH6致病性变异的女性携带者患子宫内膜癌的风险是普通人群中女性的25倍(HR,25.5;95%CI,16.8–38.7;P<0.001)。
在另一项研究中,MSH6携带者(71%)患子宫癌的累积终生风险高于MLH1(27%)和MSH2(40%)致病性变异携带者(P=0.02),MSH6致病性变异携带者诊断时平均年龄较大,为54岁(n=29;范围,43-65岁),而MLH1携带者为48岁,MSH2致病性变异携带者为49岁。
在PMS2致病性变异携带者中,据报道70岁时患子宫内膜癌的风险为15%。
根据结肠癌家族登记项目中收集的前瞻性数据得出以下结论,MMR基因致病性变异女性中5年子宫内膜癌风险约为3%,10年子宫内膜癌风险约为10%。
一项使用1942例既往无癌症的MMR携带者的汇总欧洲登记数据进行的前瞻性研究显示,MLH1携带者子宫内膜癌的累积发生率为34%,MSH2携带者为51%,MSH6携带者为49%,PMS2携带者为24%。
由EPCAM致病性变异引起的MSH2蛋白表达缺失的女性也有患子宫内膜癌的风险,这取决于变体在EPCAM中的位置。一项研究发现EPCAM缺失携带者患子宫内膜癌的累积风险为12%(95%CI,0%-27%)。
一项对127名以子宫内膜癌为指标癌症的Lynch综合征妇女进行的研究发现,患其他癌症的风险显著增加。据报告,以下风险升高:CRC,48%(95%CI,27.2%-58.3%);肾癌、肾盂癌和输尿管癌,28%(95%CI,11.9%-48.6%);膀胱癌,24.3%(95%CI,8.56%-42.9%);乳腺癌,2.51%(95%CI,1.17%-4.14%)。
一项对113个来自结肠癌家族登记的携带MSH6致病性变异的家族进行的研究显示,与普通人群相比,女性MSH6携带者中子宫内膜癌的发生率增加了26倍(HR,25.5;95%CI,16.8-38.7)。 与普通人群相比,与Lynch综合征相关的其他癌症的发生率增加了6倍(HR,6.0;95%CI,3.4–10.7),但在男性MSH6携带者中未观察到。
Lynch综合征相关子宫内膜癌并不局限于子宫内膜样亚型,Lynch综合征的子宫肿瘤谱可能包括透明细胞癌、子宫乳头状浆液性癌和恶性混合性Müllerian肿瘤。
同时,子宫内膜癌最常见于子宫下段。 (如需了解有关筛查方法的信息,请参见本总结Lynch综合征的子宫内膜癌筛查章节。)
多项研究显示,与Lynch综合征相关的其他恶性肿瘤的风险增加,包括胃癌、胰腺癌、卵巢癌、小肠和脑癌、膀胱、输尿管和肾盂移行细胞癌以及皮肤皮脂腺瘤。
此外,一些研究表明与乳腺癌、前列腺癌和肾上腺皮质癌相关。
其中许多恶性肿瘤的相关性强度受到以下限制:大多数研究样本量较小(因此,与相对风险[RR]相关的CI较宽)、分析的回顾性性质以及转诊或确认偏倚。
迄今为止最大的前瞻性研究针对来自结肠癌家族登记的446例未受累的致病性变异携带者。
结肠癌家族登记研究是一个国际队列,从北美和澳大利亚的6个中心招募基于人群和基于诊所的受试者。对照受试者为来自已知MMR致病性变异家庭的非携带者。使用三个亚队列分析CRC(365例携带者,903例非携带者)、子宫内膜癌(215例携带者,523例非携带者)和其他癌症(446例携带者,1029例非携带者)的风险。随访长达10年的受试者患CRC(SIR,20.48;95%CI,11.71-33.27;P < 0.01)、子宫内膜癌(SIR,30.62;95%CI,11.24-66.64;P < 0.001)、卵巢癌(SIR,18.81;95%CI,3.88-54.95;P < 0.001)、胃癌(SIR,9.78;95%CI,1.18-35.30;P = 0.009)、肾癌(SIR,11.22;95%CI,2.31-32.79;P < 0.001)、膀胱癌(SIR,9.51;95%CI,1.15–34.37;P = 0.009)、胰腺癌(SIR,10.68;95%CI,2.68–47.70;P = 0.001)和女性乳腺癌(SIR,3.95;95%CI,1.59-8.13;P = 0.001)的SIR增加。
Lynch综合征的一个得到充分描述的变种,其表型为多发性皮肤肿瘤(包括皮脂腺腺瘤、皮脂腺癌和角化棘皮瘤)和CRC,称为Muir-Torre综合征。
在Muir-Torre家族中发现了MLH1、MSH2和MSH6基因的致病性变异,在MSH2携带者中该综合症的发生率增加
对1,914例无亲缘关系的MLH1和MSH2先证者进行研究,发现MSH2在具有Muir-Torre综合征表型的患者中更常见。在15例皮脂腺皮肤肿瘤患者中,13例(87%)发生MSH2致病性变异,而2例发生MLH1致病性变异(P=0.05)。
在163个随机收集的皮脂腺肿瘤的69个(42%)中,使用IHC或MSI检测发现DNAMMR活性缺陷,提示这是这些病变发生的共同机制,检测皮脂腺肿瘤中MMR缺陷将对筛查Lynch综合征或Muir-Torre综合征无效。
(有关Muir-Torre综合征中皮肤肿瘤的更多信息,请参见PDQ皮肤癌遗传学总结中的皮脂腺癌章节。)
癌症发生部位 b | 一般人群风险c | 林奇综合征患者的患病风险d |
---|---|---|
胃部 | <1 | 0.2-13 |
卵巢 | 1.3 | 3.4-22 |
肝胆管 | <1 | 0.02-4 |
泌尿道 | <1 | 0.2-25.5 |
小肠 | <1 | 0.4-12 |
脑/CNS | <1 | 1.2-3.7 |
皮脂腺肿瘤 | <1 | 9.0 |
胰腺 | 1.6 | 0.4-3.7 |
CNS = 中枢神经系统。 | ||
a 改编自Syngal et al. | ||
b 不断变化的数据表明Lynch综合征与乳腺癌和前列腺癌之间存在潜在相关性。(如需了解有关这些癌症的更多信息,请参见本总结中可能与Lynch综合征相关的其他癌症章节。) | ||
cHowlader et al. | ||
d 癌症风险估计值范围因研究样本量、受试者确定和统计方法而异。 |
其他肿瘤被认为是林奇综合征疾病谱的一部分,但这是有争议的。乳腺癌和前列腺癌被认为是可能的林奇综合征相关肿瘤,因此目前对MMR基因进行多基因(组合)检测,以确定是否存在这些癌症。
Lynch综合征中的乳腺癌风险问题一直存在争议。回顾性研究结果不一致,但多项研究证明Lynch综合征患者的一部分乳腺癌中存在微卫星不稳定性;
其中一项研究评估了Lynch综合征患者的乳腺癌风险,发现其并未升高。
但是,迄今为止在来自结肠癌家族登记的446例未受累的致病性变异携带者且随访长达10年的患者中开展的最大型前瞻性研究
报告乳腺癌SIR升高为3.95(95%CI,1.59-8.13;P=0.001)。
该研究小组随后分析了764名既往确诊结直肠癌的MMR基因致病性变异携带者的相关数据。 结果显示,结直肠癌后乳腺癌的10年风险为2%(95%CI,1%-4%),SIR为1.76(95%CI,1.07-2.59)。
来自英国的一系列由临床推荐的林奇综合征家族组成的研究表明,157例MLH1携带者患乳腺癌的风险增加了两倍,而其他MMR变异携带者患乳腺癌的风险未增加。
15项具有分子肿瘤检测结果的研究中Lynch综合征的乳腺癌风险荟萃分析结果显示,MMR致病性变异携带者中122个乳腺癌中的62个(51%;95%CI,42%-60%)为MMR缺陷型。此外,在总共21项研究中的乳腺癌风险预估显示,在8项比较MMR变异携带者与非携带者的研究中,患乳腺癌的风险增加了2-18倍,而在13项研究中未观察到乳腺癌风险与Lynch综合征相关的统计学证据。
随后多项研究表明,乳腺癌风险高于先前发表的结果,
尽管尚未持续观察到这种情况。
一项有关325个加拿大Lynch综合征家庭的研究(主要包括MLH1和MSH2携带者)显示MSH2携带者患乳腺癌的终生累积风险为22%。
同样,在一项对423名Lynch综合征女性进行的研究中,乳腺癌风险升高,与MLH1和MSH2致病性变异相比,MSH6和PMS2致病性变异患者的风险显著更高。
事实上,在60岁前,PMS2、MSH6、MSH2和MLH1患乳腺癌的风险分别为37.7%、31.1%、16.1%和15.5%。 这些结果与另一项针对528例Lynch综合征相关致病性变异(包括MLH1、MSH2、MSH6、PMS2和EPCAM)患者的研究一致,其中仅患乳腺癌的患者中PMS2和MSH6变异发生率高于仅患结直肠癌的患者(P=2.3 x 10-5)。
通过对美国10,000多例接受基因检测的癌症患者进行研究,提供了支持MSH6与乳腺癌相关性的额外数据。
结果表明,MSH6与乳腺癌相关,OR为2.59(95%CI,1.35-5.44)。综上所述,这些研究强调了随着更多患者通过多基因组合检测进行检测,Lynch综合征患者的风险特征如何继续发展,与先前研究相比,具有PMS2和MSH6致病性变异的患者数量更多。 在缺乏明确风险估计的情况下,根据家族史对Lynch综合征患者进行乳腺癌筛查。
在对来自两个美国Lynch综合征登记的198个家庭的研究中发现前列腺癌与Lynch综合征相关,其中前列腺癌最初并非家庭选择标准的一部分。在60岁时,MMR基因致病性变异携带者亲属患前列腺癌的风险为6.3%,在80岁时为30%,而在60岁时人群风险为2.6%,在80岁时为18%,总体HR为1.99(95%CI,1.31-3.03)。
2014年的一项荟萃分析支持该相关性,发现在已知MMR致病性变异的男性中前列腺癌的RR估计值为3.67(95%CI,2.32-6.67)。
在MSH2致病性变异的患者中,这种风险可能增加。
尽管围绕常规前列腺特异性抗原(PSA)筛查存在普遍争议,但作者认为,在男性MMR基因携带者中,从40岁开始,通过PSA和直肠指检进行筛查是“合理考虑”因素。
一项在692名未根据癌症家族史或诊断时年龄选择的转移性前列腺癌男性中进行的研究显示,在4名男性(0.5%)中发现了种系MMR致病性变异。
目前,分子和流行病学证据支持前列腺癌是Lynch综合征癌症之一。与乳腺癌一样,
在为MMR致病性变异携带者制定前列腺癌监测指南之前,还需要更多的研究来确定绝对风险和年龄分布。(有关前列腺癌和Lynch综合征的更多信息,请参见PDQ“前列腺癌遗传学”总结的“MMR基因”章节。)
在114例ACC病例分析中,其中94例患者进行了详细的家族史评估并排除了Li-Fraumeni综合征,3例患者有提示Lynch综合征的家族史。 94个家庭中MMR基因致病性变异的发生率为3.2%,与未经选择的结直肠癌和子宫内膜癌患者中Lynch综合征的比例相似。 在对来自同一项目的135个MMR基因致病性变异-阳性Lynch综合征家族的回顾性综述中,发现2例先证者有ACC病史。 在可进行MSI检测的4个ACC中,均为MSS。这些数据表明,如果在ACC索引病例中怀疑Lynch综合征,采用MSI或IHC检测对ACC进行初步评估可能会产生误导。
在一些研究中发现另外几种癌症与Lynch综合征相关,但仍需进一步探讨。表12比较了普通人群与Lynch综合征患者患这些癌症的风险。
与针对普通人群中一般风险人群的建议相比,Lynch综合征患者中CRC及其前体病变腺瘤息肉的生物学行为的多个方面支持在该人群中实行不同的CRC筛查方案。目前,由于MMR系统中致病性种系缺乏,针对Lynch综合征的癌症筛查和监测的建议考虑了相对于一般人群的癌症风险差异。以下生物学差异构成了目前实施的Lynch综合征筛查策略的基础:
Lynch综合征中的CRC比散发性癌症发生得更早;然而发病年龄的差异取决于发生突变的MMR基因。(如需了解有关CRC基因特异性发病年龄的更多信息,请参见本总结Lynch综合征相关的患病率、临床表现和癌症风险章节。)
Lynch综合征致病性变异携带者发生结肠腺瘤的风险增加,该携带者的腺瘤发病年龄小于同族致病性变异阴性患者。
据报道,MMR致病性变异携带者发生腺瘤的风险是非携带者的3.6倍。
到60岁时,70%的携带者发生腺瘤,而非携带者为20%。与对照受试者的腺瘤相比,携带者的大部分腺瘤不存在MMR蛋白表达,更可能出现发育不良的特征。
在一项研究中,携带者诊断腺瘤时的平均年龄为43.3岁(范围:23-63.2岁),诊断癌症时的平均年龄为45.8岁(范围:25.2-57.6岁)。
较大比例的Lynch综合征CRC(60%-70%)发生在右半结肠,提示单纯乙状结肠镜检查并非合适的筛查策略,结肠镜检查可对结肠进行更完整的结构检查。 已有对Lynch综合征行监测性结肠镜检查的循证综述。
Lynch综合征患者一生中CRC的发生率明显增高,提示应使用最灵敏的检测方法。(有关可用的结肠监测建议,请参见表13。)
从正常黏膜到腺瘤再到癌的进展加快,
提示应在较短间隔时间(每1-2年)进行筛查,并辅以结肠镜检查。
已经证明MMR基因致病性变异携带者发生可检测腺瘤的年龄小于非携带者。
目前尚不清楚这是否反映了腺瘤的发生率更高,或Lynch综合征中存在更大的腺瘤且检测效果更好。
在芬兰的一项筛查研究中,对Lynch综合征中CRC的风险进行了研究和更新,研究从20世纪80年代初持续至今。
在本试验过程中,纵向研究的设计不断发展。在最早研究阶段,有关每例患者的变异状态的信息未知,受试者若符合临床标准则可以入组研究;该研究包括一些先前确诊癌症或腺瘤的患者以及其他无此类病史且正在接受无症状筛查的患者,而对照组包括来自拒绝筛查的相同家庭的患者。其中许多患者(68%)使用x线造影/钡灌肠进行了筛查。当可获得该信息时,结肠镜检查是用于MMR致病性变异携带者的方法,根据研究结果,检查间隔从5年缩短至3年至2年。
在多项研究中进行的一项为期15年的对照筛查试验表明,来自Lynch综合征家族的患者使用结肠镜检查检测的CRC发生率、CRC特异性死亡率和总死亡率均降低。
对来自Lynch综合征家族的133例患者每隔3年进行一次结肠筛查,未对来自这些家族的119例对照患者进行筛查。在筛查的受试者中,8例受试者(6%)出现CRC,而出现CRC的对照受试者为19例(16%),筛查后风险降低了62%。此外,筛查组的所有CRC均为局部CRC,未引起死亡,而对照组有9例CRC引起的死亡。筛查组的总死亡率也有获益,其中筛查组10例死亡,对照组26例死亡(P=0.003)。
随后,该研究重点关注了既往未确诊腺瘤或癌症的受试者。符合条件的420名致病性变异携带者的平均年龄为36岁,接受结肠镜检查的平均次数为2.1,中位随访时间为6.7年。在28%的受试者中检测到腺瘤。在60岁时,男性中出现一个或多个腺瘤的累积风险为68.5%,女性中为48.3%。值得注意的是,在60岁时,在基线检查时无癌症的男性患者中检测到癌症的风险为34.6%,女性患者中为22.1%,因此视为间期癌症。在60岁时,男性中腺瘤或癌症的合并累积风险为81.8%,女性中为62.9%。对于腺瘤和癌,约50%位于脾曲近端。尽管结肠镜监测的CRC发生率较高,但在该非随机研究中,推荐的短时间间隔并没有得到定期遵守。这些作者建议每隔2年进行一次监测。这符合大多数共识指南(参见表13),其中适当的结肠镜筛查间隔保持每1-2年一次。检测后10年对242名致病性变异携带者的结肠镜监测数据进行分析,结果显示CRC和子宫内膜癌的监测程序的依从性为95%。尽管未预防所有CRC,但死亡率与变异阴性亲属相当。然而,这可能是由于研究的样本量较小所致。
考虑到结肠镜检查是结肠癌监测的公认措施,初步数据表明,使用色素内镜(如靛胭脂)可能增加微小、组织学晚期腺瘤的检出率。
当检测到腺瘤时,就提出了是否采用MSI/IHC检测腺瘤的问题。一项对既往患有CRC和已知MMR致病性变异的患者进行的研究发现,12个腺瘤中的8个同时存在MSI和IHC蛋白丢失。
然而,本研究的作者强调,腺瘤中MSI/IHC检测正常并不能排除Lynch综合征。MSI/IHC异常在最小的腺瘤中不常见,在大于8mm的腺瘤中更常见,这也表明MMR缺陷是在生长过程中获得的。
由于基因特异性CRC风险的变异性,该领域的专家提出了基因特异性筛查和监测建议。例如,欧洲联合体
提出了临床建议,即将结直肠癌和子宫内膜癌筛查的开始时间推迟到30岁,与对MSH6致病性变异携带者后期开始筛查的建议一致。 此外,参与Lynch综合征患者和家庭护理的美国虚拟工作组2015年的一篇综述得出结论,尽管多项研究表明单等位基因PMS2携带者的外显率降低,但不建议对该组的Lynch综合征癌症监测指南进行任何变更。
虽然初始数据可能支持通过特定MMR基因突变启动和监测CRC和其他结肠外癌症的不同策略,
但在获得更多数据之前,与(a)医学界和受累患者总体遵守建议有关的问题
和(b)与特定筛查方式相关的局限性
已阻止实施基因特异性指南。
组织 | 开始年龄筛查 | 筛选间隔 | 推荐的筛查方式 | 注释 |
---|---|---|---|---|
NCCN(2019) | 家族中最年轻的CRC病例(如果在25岁之前)确定前20-25年或2-5年 | 1-2年 | 结肠镜检查 | 对于MSH6携带者,考虑开始结肠镜检查的年龄较大,例如在30岁或比任何CRC亲属小10岁。由于PMS2基因携带者的数据有限,专家组无法对推迟启动结肠镜检查的年龄提出具体建议 |
美国结直肠癌多协会工作组(2014)b | 家族中最年轻的CRC病例(如果在25岁之前)确定前20-25年或2-5年 | 1-2年(每年针对MMR致病性变异携带者) | 结肠镜检查 | 对于MSH6和PMS2携带者,考虑分别在30岁和35岁开始筛查,除非家族中发生早发性癌症。也可采取针对BMMRD患者的建议。 |
Mallorca组(2013) | 20-25年 | 1-2年 | 结肠镜检查 | |
ESMO(2013)c | 家族中最年轻的CRC病例(如果在25岁之前)确定前20-25年或5年;未确定上限 | 1-2年 | 结肠镜检查 | |
BMMRD=双等位基因错配修复缺陷;CRC=结直肠癌;ESMO=欧洲肿瘤内科学会;IHC=免疫组织化学;MMR=错配修复;MSI=微卫星不稳定性;NA=未处理;NCCN=美国国家综合癌症网络。 | ||||
a 本表总结了2010年及以后的现有指南。其他组织(包括美国癌症协会)在2010年之前已发布了指南。 | ||||
b 美国结直肠癌多协会工作组包括以下组织:美国家庭医学协会、美国胃肠病学会、美国医师学会-美国内科医学会、美国放射学会、美国胃肠病协会、美国结直肠外科医师学会和美国胃肠内镜学会。 | ||||
c 美国临床肿瘤学会和日本肿瘤内科学会已认可表中所示的ESMO指南。 |
注:还可获得PDQ有关一般人群中子宫内膜癌筛查的单独总结。
子宫内膜癌是Lynch综合征家族中观察到的最常见的结肠外癌症,在约50%的Lynch综合征家族中至少1名女性受累。(有关MMR致病性变异携带者患子宫内膜癌的基因特异性风险的更多信息,请参见本总结的“子宫内膜癌”章节。)
在一般人群中,在女性出现包括异常或绝经后出血在内的症状时,一般确诊为子宫内膜癌。进行子宫内膜取样以提供用于诊断的组织学标本。80%的子宫内膜癌妇女表现为I期疾病,目前尚无数据表明Lynch综合征妇女的临床表现与一般人群不同。
鉴于其患子宫内膜癌的风险显著增加,建议对Lynch综合征妇女进行子宫内膜筛查。拟定的筛查方式包括经阴道超声(TVUS)和/或子宫内膜活检。在没有数据支持的情况下,TVUS继续被广泛推荐使用;目前NCCN指南认为,无明确证据支持对Lynch综合征进行子宫内膜癌筛查。
关于Lynch综合征,有两项研究检测了TVUS在女性子宫内膜筛查中的应用。
在一项对来自Lynch综合征家族或“Lynch样综合征/HNPCC样”家族的292例女性患者的研究中,未见由TVUS检查所检出的子宫内膜癌病例。此外,有症状的女性患者中出现了两例间期癌。
在另一项研究中,41例Lynch综合征女性患者被纳入TVUS筛查项目。在进行的179次TVUS程序中,有17次见异常结果。对这17例行子宫内膜取样检查,3例为复杂非典型增生,14例为正常子宫内膜取样。然而,有1例TVUS未能确诊患者,在TVUS结果正常后8个月出现异常阴道出血,并发现患有IB期子宫内膜癌。
这两项研究均表明,TVUS既不具有敏感性也不具有特异性。
一项对 175 例 Lynch 综合征妇女(包括子宫内膜取样和TVUS)的研究显示子宫内膜取样检查比TVUS检查敏感性高。在14例确诊为子宫内膜癌的患者中,子宫内膜取样检查检出11例。另外3例患者中的2例有症状女性患者发展为间隔期癌,1例为子宫切除术时发现的隐匿性子宫内膜癌。子宫内膜取样检查同时还确诊了另外14例子宫内膜增生。在14例子宫内膜癌患者组中,10例行TVUS筛查的患者同时也行子宫内膜取样检查。其中4例TVUS为异常,6例为正常。
这项队列研究显示子宫内膜取样检查比TVUS在子宫内膜筛查中获益更多,但是没有数据可以预测,使用任何其他筛查方式对合并Lynch综合征的子宫内膜癌患者生存率有获益。
有些研究提示,临床或遗传诊断为Lynch综合征的女性患者并没有接受完整的妇科筛查。
(有关更多信息,请参阅本总结“遗传性结肠癌综合征的社会心理问题”一章中的“Lynch综合征”一节中关于妇科癌症筛查的信息。)
Lynch综合征患者的卵巢癌累计终生风险估计在3.4%-22%范围内。
然而,目前对于Lynch综合征家族,没有卵巢癌筛查的有效性研究。对那些没有行能预防妇科癌症的风险降低手术的女性患者,用于子宫内膜癌筛查的TVUS检查在临床实践中已扩展至用于卵巢癌筛查。然而,由于现有筛查方法缺乏敏感性和特异性,NCCN坚称数据不支持Lynch综合征患者行常规卵巢癌筛查。
证据级别:未指定
Lynch综合征患者为预防子宫内膜癌或卵巢癌的一项有效措施是行风险降低的手术。一项对315例致病性MMR基因变体的妇女进行回顾性研究,比较子宫切除术和卵巢切除术前后子宫内膜癌和卵巢癌的发生率。在随访的子宫内膜癌女性患者中,手术组平均随访13.3年,非手术组平均随访7.4年;在随访的卵巢癌女性患者中,手术组平均随访11.2年,非手术组平均随访10.6年。手术组未检出癌症,相对而言,非手术组子宫内膜癌检出率为33%,卵巢癌检出率为5.5%。
一项关于成本-效益分析的数学建模研究中,在30岁携带与Lynch综合征相关的MMR基因变体人群中,将实施风险降低手术(预防性子宫切除术和双侧输卵管卵巢切除术)和非手术方式筛查进行比较,结果显示,预防性手术是有成本效益的,成本较低,QALY较高。
随后的一项数学建模研究评估了多种筛查和手术策略,结果提示30岁开始行年度筛查并于40岁后行风险降低手术是最有效的策略。
筛查其他Lynch综合征相关癌症的决定是在个体基础上进行的,并依赖于FDR和Lynch综合征二级亲属中确诊的癌症。
男性Lynch综合征患者发生胃癌的终生风险约8%,女性患者约5%。
最近的流行病学数据显示,胃癌的诊断率比以前高达13%的数据相比,呈下降趋势。大多数与Lynch综合征相关的胃癌的组织学特征为肠型,因此可采用食管胃十二指肠镜(EGD)筛查来确诊。
虽然没有明确的数据支持对胃、十二指肠和远端小肠癌的监测,但是Lynch综合征患者40岁行基线检查时,在结肠镜检查同时可使用EGD行可视化十二指肠的检查。一旦发现幽门螺杆菌,需进行测定和治疗。尽管关于适当监测间隔时间的数据有限,但是普遍认为应每3-5年监测一次,特别是对于有胃、十二指肠和远端小肠癌家族史和亚洲血统的人群。
与Lynch综合征相关小肠癌的终生风险报道不一,从小于1%至12%不等。
大多数小肠恶性肿瘤局限于十二指肠和回肠,分别在EGD和结肠镜(专用回肠插管法)的检查范围内。评估小肠病变的其他方法包括CT肠造影和胶囊内镜检查,但成本效益分析不支持将这些评估用于Lynch综合征的常规筛查项目。
尿路恶性肿瘤包括肾盂、输尿管的移行细胞肿瘤和膀胱肿瘤。这些恶性肿瘤的相关终生风险各不相同,范围从小于1%至25%不等,较高的估计值与尿路内不同位置(包括膀胱)发现的癌症合并相关。
以尿液细胞检测作为潜在筛查方式的研究显示,因其低敏感性和高假阳性率,故最终往往需要行额外的侵入式检查(例如膀胱镜检查)。目前没有用于无症状Lynch综合征患者常规筛查的有效方法。
有两项校正确认偏移的队列研究支持Lynch综合征患者的胰腺癌风险升高。一项研究显示,70岁时胰腺癌的累积风险为3.7%,比普通人群高8.6倍。
另一项使用结肠癌家族登记数据的前瞻性研究显示SIR为10.7,累积风险为0.95%。
这些研究结果支持一个专家共识,即推荐对Lynch综合征和具有胰腺癌FDR的患者进行胰腺癌筛查,而对于其他有类似风险的高危人群也一样。
需要注意的是,共识中不推荐对普通人群进行尿路、膀胱、肝胆系统和胰腺的癌症筛查。然而,NCCN建议对于有尿路上皮癌家族史和MSH2致病性变体患者(特别是男性),考虑进行尿路上皮癌监测。
直肠腺瘤/癌症预防计划(CAPP2)是一项双盲、安慰剂对照、随机试验,对于一些国际中心监测项目中的Lynch综合征患者,确定阿司匹林在预防其CRC中的作用。
这一长达4年的研究随机将861例参与者进行分组:阿司匹林组(600 mg/天),阿司匹林安慰剂组,抗性淀粉组(30g/天),或抗性淀粉安慰剂组。平均随访55.7个月(范围:1-128个月),48例参与者中发现53例原发性CRC(阿司匹林组427例中发现18例,阿司匹林安慰剂组434例中发现30例。)未分配至阿司匹林组的76例患者(因阿司匹林敏感或消化性溃疡病史),被随机分配至抗性淀粉组或抗性淀粉安慰剂组。意向治疗分析得出结论:CRC的HR为0.63(95% CI, 0.35–1.13;P=0.12)。然而,确诊为CRC的患者中有5例发生了两处原发性结肠癌。Poisson回归分析阐述了多发性原发CRC的影响,且阿司匹林对其产生了保护作用(发病率比[IRR],0.56;95% CI, 0.32–0.99; P=0.05)。对于完成至少2年治疗的参与者,符合方案分析得出结论:HR为0.41 (95% CI, 0.19–0.86; P=0.02) 和IRR为0.37 (0.18–0.78; P=0.008)。关于Lynch综合征癌症(子宫内膜癌、卵巢癌、胰腺癌、小肠癌、胆囊癌、输尿管癌、胃癌、肾癌和脑癌)的分析显示,与安慰剂组相比,阿司匹林有保护作用。阿司匹林组和安慰剂组不良事件无显著差异,并且任何治疗均未发现严重不良反应。作者的结论是,平均25个月每天使用阿司匹林600mg,可大大降低Lynch综合征患者的癌症发病率。CAPP2未能显示出每日抗性淀粉摄入的任何作用。研究的局限性是,各中心的不同监测频率没有被标准化。2008年发表了一项入组746例Lynch综合征患者的早期CAPP2研究结果,
对于结肠腺瘤/癌,短期平均随访29个月(范围:7-74个月)后,未显示出显著的保护作用(相对风险, 1.0; 95% CI, 0.7–1.4)。2015年一项对结肠癌家族登记系统中的1858例参与者进行调查的结果显示,阿司匹林和布洛芬可能对MMR基因的致病性变体有化学预防作用。
CAPP3试验用于评估低剂量阿司匹林(100mg、300mg、600mg阿司匹林肠溶片设盲)的效果,试验开始于2013年,预期至2021年将纳入近3000致病性变体患者。
尽管证据等级有1级,但是专家认为关于阿司匹林用于Lynch综合征化学预防的证据还不够充分或成熟,无法推荐其规范使用。
Lynch综合征的一个标志性特征存在同时性和异时性 CRC。据报道,节段性结肠切除术后10年异时性CRC发生率为16%,20年为41%,30年为63%。
由于同时性和异时性肿瘤发病率的升高,对结肠肿瘤性病变的Lynch综合征患者,推荐的手术治疗一般为扩大结肠切除术(全部或几乎全部)。然而,治疗方案必须个体化,通常包括节段性结肠切除术。数学建模研究显示,67岁以上患者比早发性癌症的年轻患者行扩大手术获益少。一项Markov决策分析模型中,对于早发性CRC的年轻患者,行扩大手术的生存优势增加,可比行节段性切除术的相同患者生存时间延长4年。
推荐扩大手术时需要权衡患者的并发症、疾病的临床分期、患者的意愿、和手术技能。没有前瞻性或回顾性研究显示,Lynch综合征患者行扩大切除术比节段性切除术的生存优势。
有两项研究表明,与行节段性切除术患者相比,行扩大切除术的患者的异时性CRC和CRC相关的其他手术较少。
权衡扩大切除术与节段性切除术的功能结果至关重要。尽管大部分患者行结肠切除术后适应良好,但是有些病人需使用止泻药物。一个决策模型对比了30岁患者接受腹部结肠切除术和节段性结肠切除术患者的QALY。
在这个模型研究中,扩大手术与节段性手术之间没有太大差异,行节段性手术比行扩大手术患者的QALY多0.3年。
就手术方式来说,重要的是认识到次全或全结肠切除术并不能消除直肠癌风险。据报道,结肠切除术后12年,腹部结肠切除术后直肠残端发生癌变的终生风险是12%。
另外,扩大结肠切除术的常见并发症为潜在的排尿和性功能障碍,以及腹泻;随着吻合口越来越靠近远端,风险越来越高。因此,手术的选择必须由外科医生和患者根据个体情况决定。
对于Lynch综合征和直肠癌的患者,需给予相同的手术选择(扩大和节段性切除术)和注意事项。扩大手术包括保存括约肌的恢复性直肠结肠切除术和IPAA术,以及不保存括约肌的直肠结肠切除术联合回肠造口术。据报道,以直肠癌作为基准,节段性切除术后异时性结肠癌风险在15%至27%之间。
两项回顾性研究显示,Lynch综合征患者,直肠癌节段性切除术后患者异时性结肠癌的发病率分别为15%和18%。
在其中一项研究中显示,直肠切除术后中位随访时间101.7个月时,异时性高危腺瘤和癌症的合并风险为51%。
没有关于Lynch综合征患者术后生育能力的数据。据报道,在女性FAP患者中,结肠切除术和IRA术后生育能力没有差异,但是与普通人群相比,以IPPA行恢复性直肠结肠切除术患者的生育能力下降54%。
另一项对FAP患者进行问卷调查的研究表明,在IRA、IPAA和直肠结肠切除术加末端回肠造口术后,生育问题的发生率相似。在该研究中显示,手术时年龄越早,生育问题就越多。
对于Lynch综合征的治疗,当确诊CRC时,多数临床医生支持扩大切除术。但是,如上所述,手术的选择必须由外科医生和患者根据个体情况决定。
如前面章节所讨论的,MSI不仅是Lynch综合征的分子特征,还出现在10%-15%的散发CRC病例中(主要由于MLH1高甲基化或MMR基因的双等位基因体细胞突变)。虽然MSI检测最初是用于筛查可能携带致病性MMR基因变体的,但是人们越来越认识到MSI其对预后和治疗的重要意义。MSI 检测的实用性超越了识别 Lynch 综合征,使得普遍 MSI 筛查变得更加引人注目,并导致其被广泛应用。有几项研究表明,与MSS癌相比,MSI-H的CRC的分期特异性生存率更高。另外,MSI-H的CRC切除术后,5氟尿嘧啶(5-FU)作为辅助化疗似乎没有效果,而在MSS CRC 中该药物被广泛用于治疗。最后,在早期1期和2期研究中,应用如免疫治疗对晚期MSI-H的CRC似乎有效,同时这些药物被作为单一疗法,至少在MSS的CRC中显示少许活性。
虽然MSI-H肿瘤占所有散发性CRC的15%,但与III期CRC相比,II期CRC更为常见,只出现在3%到4%的转移病例中。这种分期特征暗示了与潜在MSI-H状态相关的更好的预后。
随后的几项研究证实,与MSS病例相比,II期MSI-H CRC的生存率升高。一项Meta分析涵盖32项研究,7642例病例,1277例MSI-H,估计与MSI相关总生存率(OS)的合并HR为0.65(95% CI,0.59–0.71;异质性P=0.16;I2 [由于异质性而非偶然性导致的研究间变异百分比的指标] = 20%).
然而,虽然数据有限,但是MSI肿瘤在使用5-FU辅助治疗时没有获益(HR, 1.24; 95% CI, 0.72–2.14)。几项大型随机临床试验的后续数据证实了MSI-H的良好预后。其中包括“快速、简单、可靠的试验(QUASAR)”,该试验涵盖了1900例II期CRC患者,探讨对比了辅助性5-FU化疗与单纯手术治疗的优势。在这项研究中,MSI-H肿瘤的复发风险为MSS肿瘤的一半(相对危险度 [RR], 0.53; 95% CI, 0.40–0.70)。
在“泛欧结肠癌辅助治疗试验(PETACC)-3试验”中我们看到了类似的结果,该试验为对II期或III期CRC患者行切除术后使用5-FU联合或不联合伊立替康治疗的随机对照试验。
MSI-H状态与OS比值比(OR)为0.39(95%CI,0.24-0.65)相关,而这一优势在II期和III期疾病中均可见。
与先前其他的数据一致,对85例Lynch综合征相关的CRC患者和67例散发性dMMR CRC患者的临床病理分析显示,Lynch综合征患者的生存率显著升高,诊断年龄较低,肿瘤浸润淋巴细胞(TIL)数量较高。
在这项队列研究中,16例CRC肿瘤组的外显子组测序和新抗原数据表明,Lynch综合征相关性CRC患者的体细胞突变量和新抗原量显著高于散发性dMMR CRC患者;推测这是改善生存结局和 TIL 增加的来源。
鉴于MSI-H肿瘤偏向于累及右半结肠,因此关于MSI-H肿瘤累及直肠的结果和预后的数据不充足。有一些研究显示只有2%的直肠癌为MSI-H。
一项对62例来自同一机构的MSI-H直肠癌患者进行中位随访时间为6.8年的研究。对于I期和II期患者,直肠癌的5年特异性生存率为100%,III期为85.1%,IV期为60.0%,提示MSI-H的良好预后也适用于累及直肠的癌症。
另外,作者还报道了5-FU联合放疗的病理完全缓解率为26%,提示对于MSI-H肿瘤患者,5-FU联合局部放疗对直肠癌的治疗也有效。在这项研究中可观的病理完全缓解率显示,在开始治疗前需要进行充分活检来评估MSI状态。
几项关于CRC中MSI的研究结果预测使用5-FU辅助化疗在II期或III期结肠癌切除术后没有获益。
这在历史上一直是一个有争议的领域。众所周知,在人工培养的结肠癌细胞中,DNA-MMR活性的丧失对DNA损伤剂(细胞毒性化疗的常见机制)产生抵抗,它通过失去细胞周期停滞的信号以应对无法修复的DNA损伤。
由此产生的预测是,DNA dMMR肿瘤可能对烷基化剂、5-FU和含铂药物不完全敏感。
出人意料的是,在2000年发表的一篇文章显示,Dukes C(III期)CRC合并MSI的患者在给予5-FU辅助化疗时有显著的生存获益。
然而,本分析中的患者并未随机接受治疗;他们根据自身临床状况而被动选择辅助化疗方案,非故意地情况下,治疗组的中位年龄比对照组小13岁。
然而,在2003年,一项对570例结肠癌患者行辅助化疗的前瞻性随机对照试验结果显示,在MSI组中,5-FU辅助化疗没有任何获益。此外,当治疗合并MSI的结肠癌患者时,死亡率增加的趋势并不显著:III期癌症为2倍,II期癌症为3倍。
随后,10项研究证实了这一点,但是当给予CRC患者5-FU化疗后,所有的研究均不能证明CRC患者的获益。
相反,一项5-FU对比观察组的随机试验Meta分析显示,5-FU对MSI Ⅲ期疾病患者有潜在的获益。一项探索性亚组分析显示,只有那些与Lynch综合征相关性MSI患者才能有获益。本研究未对II期患者进行分析。
临床前数据表明,5-FU联合奥沙利铂可以避免MSI-H肿瘤对5-FU单一疗法的耐受性。
回顾性分析433例MSI-HⅡ期和Ⅲ期CRC(既有散发性又有继发于Lynch综合征)患者,与单纯手术相比,FOLFOX(5-FU联合奥沙利铂)对无病生存率(DFS)有优势。
在Lynch综合征相关的MSI患者亚群中,使用FOLFOX可改善DFS,但是,结果没有统计学意义。另外的研究已经证实,无论MSI状态如何,所有使用包括FOLFOX在内的辅助化疗的生存率都相似。
通过MSI途径发生的肿瘤比通过其他途径发生的肿瘤有更多的体细胞突变。这意味着dMMR肿瘤可能具有更多的潜在抗原(称为新抗原),并且可能比正常MMR(pMMR)肿瘤对免疫系统的调控更敏感。显微镜下,MSI-H肿瘤常常表现出大量的肿瘤淋巴细胞浸润,而有时会导致Crohn样反应。长期以来,这一组织学特征提示MSI-H肿瘤可增强免疫监测的可能性,是MSI-H肿瘤较MSS肿瘤具有更好的分期特异性生存率的主要假设之一。
为了验证免疫调节在MSI-H肿瘤中有效性的假设,在一个涵盖MSI-H或MSS癌症患者的小型队列研究中进行了抑制程序性细胞死亡-1(PD-1)的2期试验。对各种化疗方案失败的转移性疾病患者使用抗PD-1免疫检查点抑制剂——帕博利珠单抗进行治疗。
在这个小型的2期研究中,32例CRC患者(11例为dMMR,21例为pMMR,另外9例为非结直肠的dMMR肿瘤)每14天静脉注射一次帕博利珠单抗。可评估的患者中,dMMR-CRC肿瘤的免疫相关缓解率为40%(4/10),pMMR-CRC肿瘤免疫相关缓解率为0%(0/18),非CRC-dMMR肿瘤免疫相关缓解率为71%(5/7)。免疫相关的20周无进展生存率在dMMR-CRC肿瘤患者中为78%(7/9),在pMMR-CRC肿瘤患者中为11%(2/18),在非CRC-dMMR肿瘤患者中为67%(4/6)。dMMR肿瘤的体细胞突变平均是pMMR肿瘤的24倍。此外,在本研究中,体细胞突变量与PFS延长有关。作者的结论是,MMR状态预测了使用帕博利珠单抗阻断免疫检查点的临床获益。
对74例以前行过细胞毒性化疗(包括5-FU、伊立替康和奥沙利铂)的MSI-H/dMMRCRC患者进行了另一种PD-1抑制剂——纳武利尤单抗的单臂2期研究(CheckMate 142)。
总的来说,31%的患者(74例中的23例)对治疗有客观缓解,69%(74例中的51例)至少在12周内控制了疾病。在对纳武利尤单抗治疗后有缓解的患者中,在研究分析时未达到中位缓解持续时间(中位随访时间为12个月)。在本研究中,Lynch综合征相关转移性MSI-H/dMMR-CRC与非Lynch转移性MSI-H/dMMR-CRC的个体缓解率没有显著差异。研究中20%的参与者出现了3级或更高的毒性,最常见的是淀粉酶和/或脂肪酶升高,但是没有可归因于纳武利尤单抗的致死病例。
基于这些数据,2017年5月,FDA批准每3周静脉注射200mg帕博利珠单抗用于治疗任何对标准治疗无效的MSI-H/dMMR转移癌;2017年8月,FDA批准每2周静脉注射240mg纳武利尤单抗用于治疗对细胞毒性化疗无效的MSI-H/dMMR CRC。
在另一组纳入142例患者的研究中,119例转移性dMMR CRC患者接受纳武利尤单抗联合伊匹单抗治疗。
客观缓解率为55%,12周疾病控制率为80%,12个月PFS为71%,而中位缓解时间尚未达到。32%的受试者发生3级和4级毒性(最常见的是肝功能检查值升高),而13%的受试者因毒性而停止治疗。这是一项非随机研究,因此关于联合免疫检查点阻断剂是否优于单用PD-1抑制剂,特别是考虑到联合治疗的毒性明显增加,仍然存在疑问。基于这些数据,在2018年7月,FDA批准纳武利尤单抗联合伊匹单抗用于治疗dMMR/MSI-H转移性CRC,并且该疗法已通过先前氟嘧啶、奥沙利铂和伊立替康的化疗取得进展。
MSI-H相关性CRC免疫治疗的另一种替代方法是应用肿瘤导向疫苗。迄今为止,最有希望的方法包括,应用肿瘤相关性新抗原作为表位来提高肿瘤特异性T细胞的免疫。目前正在进行关于III期CRC切除术后患者的辅助治疗(NCT01461148)、转移性疾病患者(NCT01885702)和Lynch综合征患者的CRC预防(NCT01885702)等方面的研究。
Lynch样综合征在Lynch综合征疑似患者中占70%,但是种系检测未能确定致病性MMR基因变体。
与Lynch综合症中的肿瘤表型相似,CRC显现出MMR DNA蛋白的MSI和IHC缺失。然而,MMR缺陷的CRC是由MMR DNA的双等位基因体细胞失活引起的,
其中,相对于两个体细胞序列突变,MMR基因的一个等位基因的体细胞突变和另一个等位基因杂合度的丧失是最有可能的。(更多有关Lynch样综合征肿瘤表型的信息,请参阅表10。)
导致Lynch样综合征的原因可能包括:(1)可能存在一些不被当前检测所发现的种系DNA变异;(2)受影响的个体可能存在除了目前已知与Lynch综合征相关的MMR DNA基因外的种系致病性变体;或(3)除了与种系相关的机制外,还有其他使MMR DNA失活的机制。
越来越多的证据表明,Lynch样综合征先证者和家族成员的CRC风险较低,SIR为2.12,而Lynch综合征的SIR为6.04。
初步估计显示,Lynch样综合征患者的结肠外癌症风险较低,SIR为1.69,而Lynch综合征SIR为2.81。另一项对14例早发性(<50y)CRC和dMMR的回顾性研究表明,43%的患者有Lynch综合征,57%有Lynch样综合征。
在缺乏纵向随访的大规模研究的情况下,除了有与Lynch样综合征的肿瘤进展率相关的数据外,目前还强烈推荐行癌症筛查,这与Lynch综合征指南中相似。
家族性结直肠癌X型或FCCX是指符合阿姆斯特丹标准但无MSI/IHC异常的家族。
约50%符合阿姆斯特丹标准的家族缺乏致病性MMR基因变体,因此被称为FCCX家族。目前正在进行关于确定FCCX遗传病因的研究,但在很大程度上,它仍是未知的,并被认为是一种异质性疾病。然而,区分Lynch综合征和FCCX对于癌症风险评估和对受累个体和高危亲属的筛查建议具有重要意义。虽然CRC的风险增加到普通人群的两倍,但与Lynch综合征患者的CRC风险相比要低(风险增加>6倍),而且没有显著的结肠外转移癌风险。因此,对癌症筛查建议进行了改良,建议每5年进行一次CRC监测。
随着50岁以前被确诊的人越来越多,CRC的诊断年龄在流行病学上正在发生变化。
(有关普通人群中CRC发病趋势的更多信息,请参阅关于结直肠癌预防的PDQ总结。)一项研究检测了450例年轻发病的CRC(平均诊断年龄42.5岁)患者中高外显率致病性变体的患病率,并在包含至少一例结肠、子宫内膜、乳腺、卵巢和/或胰腺癌的FDR家族史中72例患者(16%)内鉴定出75个种系致病性变体或类致病性变体。
已鉴定的变体谱包括Lynch综合征和非Lynch综合征相关基因,包括几种传统上与CRC无关的基因(如BRCA1/BRCA2、ATM、CHEK2、PALB2和CDKN2A)。鉴于遗传性癌症综合征的高频率和多样性,作者认为在这一人群中进行多基因(组合)检测是有必要的。
在没有Lynch综合征的额外家族史或个人史的情况下,36岁以前确诊CRC的单发病例与MMR基因致病性变体相关。一项研究发现只有6.5%的人有MMR致病性变体,
而另一项关于50岁以下不超过一位FDR的CRC患者的研究发现,21%肿瘤中存在异常MSI,PMS2和MSH6基因缺陷的比例过高。
因此,对于非常早发的单发CRC病例,应采用MSI/IHC进行肿瘤筛查,而不是直接进行种系致病性变体分析。
FAP和Lynch综合征腺瘤的内镜治疗效果,以及关于手术推荐和计划的决策,需要腺瘤病灶的准确评估。在AFAP和Lynch综合征中,微小腺瘤的存在对AFAP中的微腺瘤和Lynch综合征中的扁平腺瘤(尽管有时较大)提出了特殊的挑战。
对内镜下检测微小息肉的敏感性需求已经增加,因为不仅在一般风险受试者中发现扁平腺瘤和无蒂锯齿状息肉,还在AFAP患者中发现衰减型腺瘤表型,以及在Lynch综合征患者中发现微小扁平腺瘤。 现代高分辨率内窥镜提高了腺瘤的检出率,但使用各种活体染料,特别是靛蓝胭脂红染料喷雾,进一步提高了检出率。某些研究表明,靛蓝胭脂红可提高粘膜对比度,从而提高腺瘤的检出率。无论有无家族史,详细的临床评估包括使用染色喷雾结肠镜(靛蓝胭脂红或亚甲蓝)
放大或非放大成像,或可能更新的成像技术,如窄带成像,
可揭示更多微腺瘤的特征性右半结肠内聚集。如果发现十二指肠腺瘤或胃底息肉伴表面不典型增生,可使用上消化道内窥镜检查了解更多信息。如果进行APC或MUTYH检查,这些发现将可能增加变体的检出率。
在各种大型系列的平均风险人群中,约有5%到10%的病例检出微小扁平病变,包括高度发育不良的腺瘤和浸润性腺癌。
其中一些研究包括串联程序——白光检查后行随机“强化”(从盲肠撤回时间> 20分钟)检查对比色素内镜检查——色素内镜组检出更多的腺瘤。
然而,在一些随机试验中,结果没有发现显著差异。
在Lynch综合征患者的随机试验中,
使用标准结肠镜,如图所示息肉切除术后,然后行靛蓝胭脂红行色素内镜检查或反复“强化”白光结肠镜检查(如上面所提到的,设计相近的一般风险筛选组)。在这个系列研究中,色素内镜组和强化白光组在腺瘤检出率上没有显著差异。然而,这些患者年龄较小,而且在多数情况下,他们曾接受过几次可能导致息肉清除的检查。
在德国的一项研究中,
一组Lynch综合征患者接受白光检查后行染色内镜检查;而另一组患者接受窄带成像结肠镜检查后行色素内镜检查。在两组中,色素内镜检查对扁平息肉的检出有显著优势,尽管一些被检出的病变是增生性的。在法国的一系列Lynch综合征病例研究中,同样采用白光检查后行色素内镜检查,色素内镜可检出的腺瘤显著增多。
AFAP比Lynch综合征患者进行的色素内镜检查评估更少。一项研究应用白光检查,检测了4例疑似AFAP患者和少于20个腺瘤。
所有患者均在色素内镜下发现1000多个小腺瘤,与结肠切除术后的病理评估结果一致。
对于FAP患者的十二指肠,色素内镜也有相似的作用效果。荷兰的一项研究显示,使用靛蓝胭脂红染色喷雾剂检测十二指肠腺瘤的数量和大小有所增加,其中包括一些较大的腺瘤。总体Spigelman评分未受到显著影响。
PJS和JPS患者出现疾病相关性小肠并发症(例如出血、梗阻、肠套叠或癌变)的风险更高。虽然FAP患者合并十二指肠肿瘤风险很高,但其空肠受累的相对风险较低。Lynch综合征患者出现小肠恶性肿瘤的RR很高,但绝对危险不到10%。尽管小肠肿瘤的风险很高,以至于需考虑对每种疾病进行监测,但这样做的技术挑战很大。由于技术上的挑战和相对较低的患病率,Lynch综合征患者行小肠筛查是没有实际证据基础的。
从历史上看,内镜不能到达的小肠中段和远端,需要对其行X线评估,包括小肠钡剂造影或一种小肠造影,通过放置一根经鼻胃十二指肠管,使得所有造影剂进入小肠利于更准确的成像。这些措施对小病灶均不敏感。任何治疗都需要剖腹探查。大多数病例都需要切除,尽管术中肠镜检查已经应用了很多年,这种术中肠镜可不切开肠管检查也可以通过肠管切口进入检查。经口肠镜检查(使用携带两个气囊、一个气囊或旋脊的加强套管来辅助)已被用于克服过度循环的技术问题,从而使得空肠深部的病变(息肉切除)获得治疗的可能。
多数资料表明,PJS采用双气囊肠镜是小肠内窥镜检查的首选方法。
这可能仅涉及经口肠镜检查,即使随后的逆行肠镜检查可以对整个小肠进行更完整地评估。由于这些检查耗时长且有发生并发症的风险,深部肠镜检查之前往往会进行更多的无创影像学检查,包括传统的钡剂检查、胶囊内镜检查、CT或磁共振肠造影检查。
在FAP中,来自胶囊内镜的数据
显示,在Spigelman III期或IV期十二指肠受累的患者中,空肠和/或回肠息肉的患病率为50%-100%,但Spigelman I期或II期中几乎没有此类息肉。所有息肉均小于10毫米,且均未作活检或切除。因此,他们的临床意义仍然不确定,但很可能是有限的,因为在FAP空肠癌的报告很少。
以上描述的PJS患者小系列的胶囊内镜检查
显示息肉出现的频率相似(50%-100%),但息肉普遍比FAP患者大得多,且更有可能出现症状,需要内镜或手术切除。由于胶囊内镜检查的敏感性,胶囊检查被认为是影像学检查的一种合适替代方法。
遗传研究已经证实在家族性CRC家族中,结肠癌、腺瘤和癌症有一个共同的常染色体显性遗传模式,
腺瘤和结直肠腺癌的基因频率为0.19。
在一个MSI阴性家族性结直肠肿瘤家族的亚组中发现与染色体9q22.2-31.2相关。
最近的一项研究将家族性CRC家族与11、14和22号染色体上的三个潜在基因位点联系起来。
十多年来,关于这些家族性癌基因位点的研究进展甚微。
符合Amsterdam-I Lynch综合征标准的家族,如果MSI检测没有显示MMR缺陷的证据,其患结直肠癌或其他癌症的风险似乎与那些患典型的Lynch综合征且有明显MMR缺陷证据的家族相同。这些符合Amsterdam-I标准的家族具有完整的MMR序列,被称为FCCX,
有人认为这些家族应归类为一个独特的群体。
FCCX的遗传病因尚不清楚。利用全基因组连锁分析和外显子组测序,在4例FCCX家族CRC患者中鉴定出核糖体蛋白S20(RPS20)基因的截短变体。
该变体与CRC在家族中共分离,优势值的对数为3。此外,在292例对照组中未发现该变体。肿瘤样本中未观察到LOH,且体外成熟RNA序列分析证实RPS20单倍体不足模型。在另外25个FCCX家族的研究中没有发现RPS20的种系变体,这表明RPS20变体是一个FCCX的罕见原因。同一组先前在18个FCCX家族中的2个受累个体中发现了骨形态发生蛋白受体1A(BMPR1A)基因的变体。
有必要进行进一步的研究,以确定或反对RPS20或BMPR1A在FCCX中的作用。
在这些初步研究之后,在家族性CRC、非Lynch综合征组中发现了几个其他假定的FCCX基因,包括多肽N-乙酰半乳糖氨基转移酶12(GALT12)基因,
BUB1和BUB3,
SEMA4A基因,
RINT1,
FAN1,
一个大家族中HNRNPA0和WIF1致病性变体的联合作用。
由于潜在的候选基因列表将继续增长,使得对待这些家族的任何简单方法都复杂化。
Lynch综合征的CRC发病年龄范围从44岁(登记系列)到平均52岁(基于人群的系列)不等。
FCCX没有相应基于人口的数据,因为根据定义FCCX至少需要一个早发病例,几乎可以肯定非常具有异质性,而且在可预见的未来不太可能出现任何基于人口的数字。直接对比FCCX和Lynch综合征发病年龄的研究表明FCCX的发病年龄稍大,
且CRC的终生风险大大降低。在一项大型研究中,完整MMR家族(FCCX家族)中CRC的SIR为2.3(95%CI,1.7-3.0),相对而言,有缺陷MMR家族(Lynch综合征家族)的SIR为6.1(95%CI,5.7-7.2)。
在FCCX家族中,也没有发现结肠外肿瘤的风险升高,这表明对CRC的加强监测是足够的。尽管需要进一步的研究,但FCCX家族中的肿瘤似乎也有不同的病理表型,肿瘤浸润淋巴细胞比Lynch综合征家族中的少。
Cowden综合征和Bannayan-Riley-Ruvalcaba综合征(BRRS)均是PTEN错构瘤综合征的组成部分。约85%的患者被诊断为Cowden综合征,约60%的BRRS患者具有可识别的PTEN致病性变体。
此外,PTEN致病性变体已经在多种临床表型患者中被识别出来。
PTEN错构瘤综合征一词是指具有PTEN致病性变体的任何患者,无论其临床表现如何。
PTEN有双特异性磷酸酶的作用,可将酪氨酸、丝氨酸和苏氨酸中的磷酸基团去除。PTEN的致病性变体是多种多样的,包括无义、错义、移位和剪切位点变体。约40%的变体存在于编码磷酸酶核心基序的第5外显子中,并且发现一些复发的致病性变体。
变体在5 '末端或PTEN的磷酸酶核心内的PTEN患者,有更多器官系统受累的趋势。
Cowden综合征的诊断操作标准已经公布并随后更新。
包括主要、次要和病理诊断标准,涵盖某些皮肤粘膜表现和成人型小脑发育不良性神经节细胞瘤(Lhermitte-Duclos病)。在系统文献综述的基础上提出了一套更新的标准
且目前用于国家癌症综合网络(NCCN)指南。
与之前的标准相反,作者的结论是,没有足够的证据可以将任何特征归类为病理学特征。随着基因检测的应用增加,特别是多基因面板的应用,Cowden综合征的临床标准需要与记录有不符合这些标准的种系PTEN致病性变体的个体的表型统一。在此之前,Cowden综合征和其他PTEN错构瘤综合征是否根据临床或基于基因检测结果定义尚不明确。美国医学遗传学和基因组学学会(ACMG)建议,对于有个人病史或一级亲属出现以下情况时应考虑转诊进行遗传学咨询:1)成人型Lhermitte-Duclos病,或2)Cowden综合征诊断标准中,符合任何三个主要或次要标准。
包括Cowden综合征的诊断标准在内的详细的建议,均可以在NCCN和ACMG指南中找到。
此外,使用临床标准来评估PTEN致病性变异率的预测模型是可行的;成本效益分析表明,如果变异率大于10%,那么PTEN种系检测的成本效益高。
在10年的时间里,国际Cowden联合会(ICC)在美国、欧洲和亚洲,前瞻性地招募了一系列符合放宽ICC PTEN检测标准的成人和儿童患者。
大多数患者不符合Cowden综合征或BRRS的临床诊断标准。在招募和测试的3399例患者中,295例先证者(8.8%)和另外73名家庭成员被发现携带种系PTEN致病性变体。除乳腺癌、甲状腺癌和子宫内膜癌外,作者还得出结论,根据癌症的风险,黑色素瘤、肾癌和结直肠癌应被视为由种系PTEN致病性变体引起的癌症谱的一部分。另一项对约100例PTEN致病性变体患者的研究证实了这些发现,并提示70岁时累积的癌症风险为85%。
在这两项研究中,年龄校正后,致病性变体携带者的CRC风险增加(SIR,5.7-10.3)。
此外,一项研究发现,在接受过至少一次结肠镜检查的PTEN致病性变体个体中,有93%的患者有息肉。
最常见的组织学表现为增生,但也有腺瘤和无蒂锯齿状息肉。PTEN致病性变体携带者CRC风险增加,因此建议对这些患者进行结肠镜监测。
然而,基于不同专家的意见,开始行结肠镜检查的年龄(30-40岁)和随后结肠镜检查的频率(每两年一次至每3-5年一次)都有很大的不同。
癌症 | 年龄校正后的SIR(95%CI) | 年龄相关的外显率评估 |
---|---|---|
乳腺 | 25.4 (19.8–32.0) | 85%从30岁左右开始b |
结直肠 | 10.3 (5.6–17.4) | 9%从40岁左右开始 |
子宫内膜 | 42.9 (28.1–62.8) | 28%从25岁左右开始 |
肾脏 | 30.6 (17.8–49.4) | 34%从40岁左右开始 |
黑色素瘤 | 8.5 (4.1–15.6) | 6%,最早发病年龄为3岁 |
甲状腺 | 51.1 (38.1–67.1) | 35%,出生时和一生中 |
CI=置信区间;DIR=标化发病率比。 | ||
a改编自Tan等人。 | ||
b 其他的历史研究表明乳腺癌的终生风险较低,在25%-50%之间。(了解更多信息,请参阅PDQ总结乳腺癌和妇科癌症的遗传学中的PTEN错构瘤综合征[包括Cowden综合征]一节。) |
PJS是一种早发的常染色体显性遗传病,其特征是嘴唇、口周和颊部出现黑色素细胞斑疹;以及多发性胃肠道息肉,包括错构瘤和腺瘤。
在绝大多数PJS家族中,已发现19p13.3号染色体STK11基因的种系致病性变体。
PJS中最常见的癌症是胃肠道癌。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%到54%
卵巢癌(主要是卵巢性索瘤)为21%。
据估计,胰腺癌的风险比普通人群高出100多倍。
一项系统回顾发现,PJS患者终生累积的癌症风险,所有部位的总和,高达93%。
表15显示了这些肿瘤的累积风险。
患有PJS的女性更易患子宫颈恶性腺瘤,它是一种罕见且侵袭性很强的子宫颈腺癌。
此外,患有PJS的女性通常会发展为伴环状小管的良性卵巢性索瘤,而患有PJS的男性则易发睾丸支持细胞瘤;
虽然这两种类型的肿瘤都不是恶性的,但它们可以引起与雌激素分泌增加相关的症状。
虽然根据已发表的文献,PJS患者的恶性肿瘤风险似乎非常高,但选择和转诊偏移可能导致其对这些风险的估计过高。
部位 | 年龄(y) | 累积风险% b |
---|---|---|
任何癌症 | 60–70 | 37–93 |
胃肠道癌 c,d | 60–70 | 38–66 |
妇科癌症 | 60–70 | 13–18 |
每个来源 | ||
胃 | 65 | 29 |
小肠 | 65 | 13 |
结直肠 | 65 | 39 |
胰腺 | 65–70 | 11–36 |
肺 | 65–70 | 7–17 |
乳腺 | 60–70 | 32–54 |
子宫 | 65 | 9 |
卵巢 | 65 | 21 |
子宫颈 e | 65 | 10 |
睾丸 e | 65 | 9 |
GI=胃肠道。 | ||
a 经Macmillan出版有限公司许可转载:肠胃病学,2010年版权所有。 | ||
b 与一般人群相比,所有癌症的累积风险均增加(P<0.05),但子宫颈癌和睾丸癌除外。 | ||
c GI癌包括结直肠癌、小肠癌、胃癌、食管癌和胰腺癌。 | ||
d Westerman等人:胃肠道癌不包括胰腺癌。 | ||
e不包括子宫颈恶性腺瘤或睾丸支持细胞瘤。 |
每个来源
PJS是由位于19p13号染色体上的STK11(也称为LKB1)肿瘤抑制基因的致病性变体引起的。
与家族性腺瘤性息肉病不同,PJS中的息肉是错构瘤。对错构瘤性息肉和PJS癌症的研究表明,等位基因失衡(LOH)符合双击假说,表明STK11是一个抑癌基因。
然而,杂合子STK11基因敲除小鼠在未失活其余野生型等位基因的情况下发生错构瘤,这表明单倍体也足以在PJS中发生肿瘤。
随后,在STK11+/-的癌症小鼠中出现LOH;
事实上,STK11+/-致病性变体杂合子和TP53-/-致病性变体纯合子的复合突变小鼠加速了错构瘤和癌症的发展。
STK11基因的种系变体表现出一系列无义、移位、错义变异,以及剪接位点变异和大片段缺失。
约85%的变体定位于表达蛋白的激酶结构域区域。没有发现强的基因型-表型相关性。
高达30%的变体是STK11的一个或多个外显子的大片段缺失,强调了缺失分析在疑似PJS病例中的重要性。
STK11已经被明确证实会导致PJS。尽管先前直接使用DNA测序的评估显示,STK11的致病性变体检出率为50%,但增加检测大片段缺失技术的研究发现,高达94%的符合PJS的临床标准的患者可检出致病性变体。
鉴于这些研究的结果,其他主要基因不太可能引起PJS。
PJS患者癌症的累积风险高,在PDQ总结结直肠癌遗传学中发表了Peutz-Jeghers综合征(PJS)诊断和监测建议表中总结的各种筛查建议。
JPS是一种遗传异质性、罕见、儿童至成人早期发病的、常染色体显性遗传病,尽管结直肠息肉占优势,但在其整个胃肠道表现为典型的错构瘤性息肉病。
JPS可伴随腹泻、消化道出血、蛋白丢失性肠病和息肉脱垂。
JPS是一种特殊类型的错构瘤性息肉,称为幼年性息肉,通常在有JPS家族史的情况下发生。青少年息肉的诊断是基于其组织学表现,而不是发病时的年龄。在婴幼儿中偶尔可见结肠或直肠的孤立性幼年性息肉,但并不意味着可确诊JPS。符合下列一项或多项标准的患者符合JPS的临床诊断:
由SMAD4基因(也称为MADH4/DPC4)18q21号染色体上的种系致病性变体引起的JPS
在近15%到60%的病例中,
而由位于染色体带10q22上编码骨形态发生蛋白受体1A(BMPR1A)基因的致病性变体引起的JPS,约占25%到40%。
由于已知SMAD4和BMPR1A中的致病性变体可解释青少年息肉病,临床医生会建议少于5个息肉的年轻患者进行基因检测。对77例(共84例)息肉患者进行的研究发现,在数量有限的息肉中,患者基因检测的结果是极少;在检测到的种系变体型中,没有一个被归类为明确的致病性或可能的致病性。
基因型/表型相关性提示SMAD4变体可能与重度胃息肉病的风险更高
和遗传性出血性毛细血管扩张症(HHT)的特征相关(见下文HHT的特征)。
据报道,JPS中CRC的终生风险为39%。
胃癌的风险增加,但还是远低于CRC的风险。
12%的JPS患者存在心脏瓣膜异常,这些患者通过单一机构的息肉病登记进行随访,
且所有可识别的致病性变体都携带SMAD4变体。
携带SMAD4致病性变体的JPS患者也可能有HHT的体征和症状,如动静脉畸形、皮肤粘膜毛细血管扩张、杵状指、骨关节病、肝动静脉畸形和小脑海绵状血管瘤,这表明这两种综合征是部分重叠的。
当临床发现患者同时具有JPS和HHT的特征时,致病性变体将出现在SMAD4基因中。大多数仅患HHT的患者在激活素受体样激酶1(ALK1)基因或内皮素(ENG)基因中都有致病性变体,尽管非常罕见(约1%-2%的HHT患者),但有报道称,SMAD4致病性变体也见于此疾病。
一系列研究表明,未经选择的HHT患者中SMAD4致病性变体的发生率略高。在这项研究中,30例未经JPS临床诊断的HHT患者中有3例(10%)在SMAD4中发现了种系变体。
相反,需要注意的是,在两项对JPS临床诊断的个体研究中显示,21%到22%的SMAD4致病性变体携带者出现了HHT的特征表现。
在对9个JPS家族21例SMAD4致病性变体携带者的研究中,81%(17/21)的患者有HHT特征。
本研究中的高患病率可能是由于纳入了来自单个家族的几个亲属和纳入了具有相同致病变体的几个家族的结果。
对携带SMAD4致病性变体的JPS患者,建议行HHT监测。
另一方面,在ALK1或ENG中没有种系变体的HHT患者可考虑进行SMAD4种系遗传检测;如果证实有SMAD4种系致病性变体,则应进行胃肠道评估。
(更多信息,请参阅表17:已发布的JPS诊断和监测建议。)
JPS 的一种严重形式在出生后最初几年发病,被称为婴儿型JPS。婴儿型JPS往往由10q22-23号染色体的微缺失引起,该区域包括BMPR1A和PTEN。(了解更多关于PTEN的信息,请参阅本总结的PTEN错构瘤综合征[包括Cowden综合征]一节。) 表型通常包括头颅畸形和发育迟缓等特征,可能是由于PTEN功能丧失所致。
复发性胃肠道出血、腹泻、渗出性肠病以及相关的发育迟缓,均与这些婴儿的死亡率高相关,从而限制了此类病例的遗传力。
约15%到60%的JPS是由SMAD4基因的种系致病性变体引起的,而约25%到40%是由BMPR1A基因的致病性变体引起的。
变异频率的巨大变化可能反映出个体研究中报告的患者数量相对较少。符合JPS临床标准的个体组在SMAD4或BMPR1A中均不具有可识别的致病性变体。
SMAD4编码一种蛋白质,它是转化生长因子(TGF)-β信号通路中的一个组成部分,将生长抑制信号从细胞表面传导到细胞核内。SMAD4中的种系致病性变体易使个体出现幼年性息肉和癌症,
且11个外显子中有6个外显子已发现了种系变体。大多数变体是独一无二的,但在多个独立的家族中发现了几个复发性致病性变体。
携带SMAD4致病性变体的患者也有很高风险出现胃癌等结肠外消化道肿瘤,通常与胃息肉病状况有关。
BMPR1A是TGF-β超家族的丝氨酸-苏氨酸激酶I型受体,激活后可导致SMAD4磷酸化。BMPR1A基因首先通过连锁分析在未识别SMAD4致病性变体的JPS家族中鉴定。BMPR1A变体包括无义、移位、错义和剪接位点变体。
在JPS患者的BMPR1A和SMAD4中,MLPA检测到基因组大片段缺失。
罕见的JPS家族已证实ENG和PTEN基因中的变体,但这些还没有在其他研究中得到证实。
几项研究初步表明,部分遗传性乳腺癌和结肠癌家族可能有由CHEK2基因的致病性变体引起的癌症家族综合征。
然而,随后的研究表明CHEK2变体只与CRC风险轻度升高有相关性(例如低外显率)。一项大型研究表明,CHEK2中的截短变体与CRC无显著相关性;然而,一种特异性错义致病性变体(I157T)与CRC风险中度增加有相关性(OR, 1.5; 95% CI, 1.2–3.0)。
在波兰进行的另一项研究也得到了类似的结果。
在这项研究中,463例来自Lynch综合征和Lynch综合征相关家族的先证者和5496例对照组被分为四种CHEK2致病性变体分型,包括I157T。在MMR变体阴性病例中,I157T等位基因的错义仅与Lynch综合征相关性癌症有相关性(OR, 2.1; 95% CI, 1.4–3.1)。未发现与截断变体相关。需要进一步的研究来证实这一发现,并确定它们是否与FCCX有关。
(了解更多信息,请参阅PDQ总结乳腺癌和妇科癌症的遗传学一节。)
HMPS是一种罕见的家族癌症综合征,其特征是伴有多种结肠息肉类型,包括锯齿状腺瘤、非典型幼年性息肉和腺瘤以及结肠腺癌。虽然最初定位为6q16-q21号染色体之间的一个位点,但现在认为HMPS位点在15q13-q14号染色体上。
虽然JPS和HMPS之间有相当多的重叠表型,但有一个大家族与15号染色体上的一个位点相连,提出了这可能是一种独特的疾病。德系犹太人家族与HMPS的连锁分析显示,在15q13.3染色体上有共同的单倍体型。
在种系1(GREM1)上游发现了一个不寻常的杂合子40kb单拷贝复制,它与具有HMPS的个体和家庭成员完全分离,而与未受影响的对照组没有分离。
这种HMPS患者中的复制与正常肠上皮中GREM1转录水平的表达增加有关。
GREM1是一种骨形态发生蛋白(BMP)拮抗剂,理论上可以促进肠内干细胞表型的形成。导致BMP信号缺陷的种系变体也引起JPS,因此HMPS和JPS之间存在潜在联系。
尽管极为罕见,GREM1致病性变体已在多个其他德系犹太血统的家族中被发现,具有不同的临床表现。虽然息肉病是大多数家族的一个统一特征,但在息肉数量、组织学和发病年龄方面存在高度的变异性。此外,在一些致病性变体携带者中已经发现了结肠外恶性肿瘤,因为受累个体的数量很少,故未明确证明与GREM1致病性变体存在因果关系。在相对有限的数据基础上,考虑对患有不明原因息肉病和/或家族性CRC的德系犹太人家族进行GREM1变体分析是合理的。
在这些家族中,有必要进行全面的变体分析,包括对GREM1的非编码区的分析。
孤立性和多发性增生性息肉(HP)(通常为白色、扁平且小)在普通人群中很常见,但它们的存在并不意味着潜在的遗传疾病。历史上,世界卫生组织定义的SPS临床诊断,必须满足以下标准之一:
尽管绝大多数SPS患者缺乏HP家族史,但约半数SPS患者有CRC阳性家族史。
一些研究表明,在有明确符合SPS标准的患者中,结直肠腺癌的患病率为大于50%。
一项研究使用不同的世界卫生组织SPS标准(SPS定义为乙状结肠附近至少5处组织学诊断为HP和/或无蒂锯齿状腺瘤(SSA),其中2处直径大于10 mm,或整个结肠内分布多于20处HP和/或SSA),发现来自57个家族中347例FDR(41%男性)患者CRC的RR为5.4(95%可信区间,3.7-7.8)。
世界卫生组织的标准是以专家意见为基础的;而且,没有已知的易感基因或基因组区域与这种疾病有可重复性的联系,所以进行基因诊断。两项研究报告了SPS个体中潜在的致病性种系变体。
在一项对38例患者的研究中,所有患者均符合多于20处HP,一处大(>1cm)HP,或结肠附近HP,发现在碱基切除修复基因MBD4和MUTYH中发现了分子错位变化。
一例患者被发现携带双等位基因MUTYH致病性变体,因此被诊断为MUTYH相关性息肉病。27例患者MBD4中未检测到致病性变体。然而,6例患者存在不确定意义的单核苷酸多态性。只有两例患者有已知的SPS家族史,38例患者中有10例合并CRC。这一系列可能包括散发性HP患者和其他疑似SPS的患者。
在40例被定义为具有5处以上HP或3处以上HP(其中2个直径大于1cm)的SPS患者的队列研究中,在1例患者在EPHB2基因(D861N)中发现种系变体。
58岁时发现结肠有锯齿状腺瘤和多于100处HP的患者,其母亲36岁时死于结肠癌。在另外100例有CRC个人史的患者或200例与之匹配的健康对照组中未发现EPHB2种系变体。
关于对SPS患者的结肠肿瘤中发现的体细胞分子遗传改变的研究更为深入。在一项对每个结肠超过20处HP、超过4处HP直径大于1cm或者每个结肠有多处(5-10)HP的患者的研究中,在息肉组织中发现了一种特殊的体细胞BRAF突变(V600E)。
这些患者的HP中有50%(20/40)检出了V600E-BRAF致病性变体。这些患者的HP也显示出明显高于左侧散发HP的CpG岛甲基化表型(CIMP高),且KRAS变异较少。在该组以往的一项研究中,来自SPS组患者的HP显示21%(16/76)的1p染色体丢失,而来自大HP(>1cm)组或者只有5-10处HP患者的HP染色体丢失为0%。
SPS患者HP的许多遗传和组织学改变与结直肠腺癌的CIMP通路共同存在。散在的锯齿状息肉是CIMP通路中CRC的前体。(了解更多信息,请参阅本总结中引言一章中“CIMP和锯齿状息肉通路”一节。)
患有PJS和JPS的人患CRC和结肠外癌症的风险增加。因为这些综合征很少见,所以没有基于证据的监测建议。由于这些综合征中结直肠癌和其他癌症的风险显著增加,已经根据回顾性研究和病例系列研究(即,仅基于专家意见)发表了一些指南。
基于已发表的指南,需使用临床判断来提出筛查建议。
组织机构 | 推荐进行STK11基因检测 a | 开始行结肠筛查的年龄 | 频率 | 方法 | 结肠外的筛查建议 | 备注 |
---|---|---|---|---|---|---|
Johns Hopkins(2006年) | 确诊,8岁时 | 18岁 | 2-3岁 | C | 乳腺、妇科(宫颈、卵巢、子宫)、胰腺、小肠、胃、睾丸 | |
Johns Hopkins(2007年) | 确诊,未指定年龄 | 青少年晚期或出现症状时 | 3岁 | C | 乳腺、妇科(宫颈、卵巢、子宫)、胰腺、小肠、胃、睾丸 | 在青少年晚期或出现症状时进行基因检测。 |
ACPGBI(2007年) | 18岁 | 3岁 | C或FS+BE | 未提及结肠外筛查 | 没有进行基因检测的建议;需要考虑行STK11/LKB1检测。 | |
克利夫兰诊所(2007年) | 18岁 | 3岁 | C | 乳腺、妇科(宫颈、卵巢)、胰腺、小肠、胃、睾丸 | ||
伊拉斯谟大学医学中心(2010年) | 25–30岁 | C | 乳腺、妇科(宫颈、卵巢、子宫)、胰腺、小肠、胃 | |||
NCCN(2019年) | 没有特殊建议 | 青少年晚期 | 2-3岁 | C | 乳腺(女性)、妇科(宫颈、卵巢、子宫)、肺、胰腺、小肠、胃、睾丸 | 向专业团队咨询。 |
ACPGBI=大不列颠及爱尔兰结直肠学会;BE=钡灌肠;C=结肠镜检查;FS=软质乙状结肠镜检查;NCCCN=国家综合癌症网络。 | ||||||
a STK11检测包括测序后的缺失分析(例如,多重连接依赖式探针扩增),前提是测序没有发现变体。 | ||||||
b 肺癌的风险增加,但除了戒烟和提高症状意识之外,没有其他建议。 | ||||||
(有关PJS和乳腺癌及卵巢癌风险的更多信息,请参阅PDQ总结乳腺癌及妇科癌遗传学一章中与乳腺癌及/或妇科癌症相关的其他高外显率综合征一节。) |
组织/作者 | 推荐SMAD4/BMPR1A检测 | 开始筛查的年龄 | 频率 | 方法 | 备注 |
---|---|---|---|---|---|
ACPGBI(2007年) | 15–18岁 b | 1–2岁 | C或FS+BE | 对基因携带者和 70 岁前患病的监测和预防性手术的讨论。 | |
克利夫兰诊所(2007年) | 15岁 | 3岁 | C,EGD | 部分携带SMAD4致病性变体的家族也患有HHT;这些人需进行HHT筛查。 | |
Johns Hopkins(2007年) | 确诊,基因筛查首选C以上 | 15岁或出现症状时 | 每年一次直至无息肉,之后每2-3年一次 | C | 超过50-100个息肉的预防性手术,不能耐受内窥镜检查,严重的胃肠道出血,JPS伴腺瘤样改变,明显的CRC家族史。 |
St. Mark's(2012年) | 确诊,4岁时行基因检测 | 12岁 | 1-3岁,基于严重程度 | C,EGD | 考虑HHT的相关检查。 |
NCCN(2019年) | 确诊 | 15岁 | 2-3岁或1岁,如果发现息肉 | C | 参考专业团队。在没有明确致病性变体的家族中,考虑从20岁开始每5年和从40岁开始每10年对未发现息肉的患者进行内镜检查。 |
ACPGBI=大不列颠及爱尔兰结直肠学会;BE=钡灌肠;C=结肠镜检查;CRC=结直肠癌;EGD=食管胃十二指肠镜;FS=软质乙状结肠镜检查;GI=胃肠道;HHT=遗传性出血性毛细血管扩张症;NCCN=国家综合癌症网络。 | |||||
a SMAD4/BMPR1A测试包括测序后的缺失分析(例如,多重连接依赖式探针扩增),前提是测序没有发现变体。 | |||||
b 年轻人,如果患者出现了症状。 |
Originally described in the 1800s and 1900s by their clinical findings, the colon cancer susceptibility syndrome names often reflected the physician or patient and family associated with the syndrome (e.g., Gardner syndrome, Turcot syndrome, Muir-Torre syndrome, Lynch syndrome, Peutz-Jeghers syndrome [PJS], Bannayan-Riley-Ruvalcaba syndrome, and Cowden syndrome). These syndromes were associated with an increased lifetime risk of colorectal adenocarcinoma. They were mostly thought to have autosomal dominant inheritance patterns. Adenomatous colonic polyps were characteristic of the first four, while hamartomas were found to be characteristic in the last three.
With the development of the Human Genome Project and the identification in 1990 of the adenomatous polyposis coli (APC) gene on chromosome 5q, overlap and differences between these familial syndromes became apparent. Gardner syndrome and familial adenomatous polyposis (FAP) were shown to be synonymous, both caused by pathogenic variants in the APC gene. Attenuated FAP (AFAP) was recognized as a syndrome with less adenomas and extraintestinal manifestations due to an APC pathogenic variant at the 3’ or 5’ ends of the gene. MUTYH-associated polyposis (MAP) was recognized as a separate adenomatous polyp syndrome with autosomal recessive inheritance. Once the pathogenic variants were identified, the absolute risk of colorectal cancer (CRC) could be better assessed for carriers of pathogenic variants (refer to Table 3).
Syndrome | Absolute Risk of CRC in Carriers of a Pathogenic Variant |
---|---|
90% by age 45 y | |
69% by age 80 y | |
10% to 56% by age 75 y, depending on the gene involved | |
35% to 53% | |
39% by age 70 y | |
17% to 68% by age 60 y | |
FAP = familial adenomatous polyposis; JPS = juvenile polyposis syndrome; PJS = Peutz-Jeghers syndrome. | |
aCancer risk estimates quoted here predate the widespread use of surveillance and prophylactic surgery. | |
With these discoveries genetic testing and risk management became possible. Genetic testing refers to searching for variants in known cancer susceptibility genes using a variety of techniques. Comprehensive genetic testing includes sequencing the entire coding region of a gene, the intron-exon boundaries (splice sites), and assessment of rearrangements, deletions, or other changes in copy number (with techniques such as multiplex ligation-dependent probe amplification [MLPA] or Southern blot). Despite extensive accumulated experience that helps distinguish pathogenic variants from benign variants and polymorphisms, genetic testing sometimes identifies variants of uncertain significance (VUS) that cannot be used for predictive purposes.
By 1900, several reports had demonstrated that patients with a large number of polyps (later subclassified as adenomas) were at very high risk of CRC and that the pattern of transmission in families was autosomal dominant. In the 20th century, the adenoma-to-carcinoma progression was confirmed, and FAP was recognized as the prototypical model for this progression.
Classic FAP is characterized by numerous (hundreds to thousands) adenomatous polyps in the colon and rectum developing after the first decade of life (refer to Figure 3).
There is also a subset of classic FAP that has an attenuated phenotype. AFAP is a heterogeneous clinical entity characterized by fewer adenomatous polyps in the colon and rectum than in classic FAP. (Refer to the Attenuated Familial Adenomatous Polyposis [AFAP] section of this summary for more information.)
FAP is one of the most clearly defined and well understood of the inherited colon cancer syndromes.
It is an autosomal dominant condition, and the reported incidence varies from 1 in 7,000 to 1 in 22,000 live births.
The presence of ethnic differences in the prevalence of FAP has been suggested
but a large study did not find significant differences in ethnic variation in more than 6,169 individuals with a personal and/or family history of CRC and polyps who were referred for genetic testing at a large reference laboratory.
Most cases of FAP result from pathogenic variants in the APC gene on chromosome 5q21. (Refer to the Genetics of FAP section of this summary for more information about the APC gene and genetic testing.)
In addition to a high risk of colon adenomas in FAP patients, various extracolonic manifestations have also been described, including upper gastrointestinal (GI) tract adenomas and adenocarcinomas; fundic gland stomach polyps; nonepithelial benign tumors (osteomas, epidermal cysts, dental abnormalities); desmoid tumors; congenital hypertrophy of retinal pigment epithelium (CHRPE); and malignant tumors (thyroid and brain tumors, hepatoblastoma). Refer to Table 4 for the risks of these extracolonic manifestations in FAP.
Malignancy | Relative Risk | Absolute Lifetime Risk (%) |
---|---|---|
Desmoid tumor | 852.0 | 15.0 |
Duodenal tumors and cancer | 330.8 | 5.0–12.0 |
Thyroid cancer | 7.6 | 2.0 |
Brain cancer | 7.0 | 2.0 |
Ampullary cancer | 123.7 | 1.7 |
Pancreatic cancer | 4.5 | 1.7 |
Hepatoblastoma | 847.0 | 1.6 |
Gastric cancer | Not defined | 0.6a |
Adapted from Giardiello et al., Jagelman et al.,Sturt et al.,Lynch et al.,Bülow et al.,Burt et al.,and Galiatsatos et al. | ||
aThe Leeds Castle Polyposis Group. |
FAP has also been known as familial polyposis coli or adenomatous polyposis coli (APC). Gardner syndrome was previously the diagnosis for FAP patients who manifested with colorectal polyposis, osteomas, and soft tissue tumors. However, Gardner syndrome has been shown genetically to be a variant of FAP, and thus the term Gardner syndrome is essentially obsolete in clinical practice.
Individuals who inherit a pathogenic variant in the APC gene have a very high likelihood of developing colonic adenomas; the risk has been estimated to be more than 90%.
The age at onset of adenomas in the colon is variable, and the median age for the appearance of colorectal adenomas is 16 years.
By age 10 years, only 15% of carriers of the APC germline variant manifest adenomas; by age 20 years, the probability rises to 75%; and by age 30 years, 90% will have presented with FAP.
The exception is AFAP, in which affected individuals typically have fewer colon polyps, which are predominantly in the right colon, and later onset of CRC. (Refer to the Attenuated Familial Adenomatous Polyposis [AFAP] section of this summary for more information.) Without any intervention, most individuals with FAP will develop CRC by the fourth decade of life.
Thus, surveillance and intervention for carriers of an APC pathogenic variant and at-risk persons have conventionally consisted of annual colonoscopy beginning around puberty for early detection of colonic polyps and to help plan when to perform colectomy.
(Refer to the Interventions for FAP section of this summary for more information.)
CHRPE are flat, darkly pigmented lesions in the retina that are present in approximately 75% of patients with FAP
compared with a general population frequency of 1.2%.
The lesions are often present at birth or in early childhood and are frequently multiple or bilateral in FAP patients.
A study of 17 individuals diagnosed with FAP and 13 at-risk family members reported a sensitivity of the presence of a CHRPE lesion in association with colonic polyps in FAP of 76%, a specificity of 92%, a positive predictive value of 93%, and a negative predictive value of 75%; thus, screening at-risk individuals for CHRPE can be a reasonable method of detecting FAP.
Desmoid tumors are proliferative, locally invasive, nonmetastasizing, fibromatous tumors in a collagen matrix. Although they do not metastasize, they can grow very aggressively and be life threatening.
Desmoids may occur sporadically, as part of classical FAP, or in a hereditary manner without the colon findings of FAP.
Desmoids have been associated with hereditary APC pathogenic variants even when not associated with typical adenomatous polyposis of the colon.
Most studies have found that 10% of FAP patients develop desmoids, with reported ranges of 8% to 38%. The incidence varies with the means of ascertainment and the location of the pathogenic variant in the APC gene.
APC pathogenic variants occurring between codons 1445 and 1578 have been associated with an increased incidence of desmoid tumors in FAP patients.
Desmoid tumors with a late onset and a milder intestinal polyposis phenotype (hereditary desmoid disease) have been described in patients with pathogenic variants at codon 1924.
A desmoid risk factor scale has been described in an attempt to identify patients who are likely to develop desmoid tumors.
The desmoid risk factor scale was based on gender, presence or absence of extracolonic manifestations, family history of desmoids, and genotype, if available. By utilizing this scale, it was possible to stratify FAP patients into low-, medium-, and high-risk groups for developing desmoid tumors. The authors concluded that the desmoid risk factor scale could be used for surgical planning. Validation of the risk factors comprising this scale was supported by a large, multiregistry, retrospective study from Europe.
The natural history of desmoids is variable. Some authors have proposed a model for desmoid tumor formation whereby abnormal fibroblast function leads to mesenteric, plaque-like desmoid precursor lesions, which in some cases occur before surgery and progress to mesenteric fibromatosis after surgical trauma, ultimately giving rise to desmoid tumors.
It is estimated that 10% of desmoids resolve, 50% remain stable for prolonged periods, 30% fluctuate, and 10% grow rapidly.
Desmoids often occur after surgical or physiological trauma, and both endocrine and genetic factors have been implicated. Approximately 80% of intra-abdominal desmoids in FAP occur after surgical trauma.
The desmoids in FAP are often intra-abdominal, may present early, and can lead to intestinal obstruction or infarction and/or obstruction of the ureters.
In some series, desmoids are the second most common cause of death after CRC in FAP patients.
A staging system has been proposed to facilitate the stratification of intra-abdominal desmoids by disease severity.
The proposed staging system for intra-abdominal desmoids is as follows: stage I for asymptomatic nongrowing desmoids; stage II for symptomatic nongrowing desmoids of 10 cm or less in maximum diameter; stage III for symptomatic desmoids of 11 cm to 20 cm or for asymptomatic slow-growing desmoids; and stage IV for desmoids larger than 20 cm, or rapidly growing, or with life-threatening complications.
These data suggest that genetic testing could be of value in the medical management of patients with FAP and/or multiple desmoid tumors. Those with APC genotypes predisposing to desmoid formation (e.g., at the 3’ end or codon 1445 of the APC gene) appear to be at high risk of developing desmoids after any surgery, including risk-reducing colectomy and surgical surveillance procedures such as laparoscopy.
The most common FAP-related gastric polyps are fundic gland polyps (FGPs). FGPs are often diffuse and not amenable to endoscopic removal. The incidence of FGPs has been estimated to be as high as 60% in patients with FAP, compared with 0.8% to 1.9% in the general population.
These polyps consist of distorted fundic glands containing microcysts lined with fundic-type epithelial cells or foveolar mucous cells.
The hyperplastic surface epithelium is, by definition, nonneoplastic. Accordingly, FGPs have not been considered precancerous. However, case reports of stomach cancer appearing to arise from FGPs have led to a reexamination of this issue.
In one FAP series, focal dysplasia was evident in the surface epithelium of FGPs in 25% of patients versus 1% of sporadic FGPs.
In a prospective study of patients with FAP undergoing surveillance with esophagogastroduodenoscopy, FGPs were detected in 88% of the patients. Low-grade dysplasia was detected in 38% of these patients, whereas high-grade dysplasia was detected in 3% of these patients. The study's authors recommended that, if a polyp with high-grade dysplasia is identified, polypectomy be considered with repeat endoscopic surveillance in 3 to 6 months.
Complicating the issue of differential diagnosis, FGPs have been increasingly recognized in non-FAP patients consuming proton pump inhibitors (PPIs).
FGPs in this setting commonly show a PPI effect consisting of congestion of secretory granules in parietal cells, leading to irregular bulging of individual cells into the lumen of glands. To the trained eye, the presence of dysplasia and the concomitant absence of a characteristic PPI effect can be considered highly suggestive of the presence of underlying FAP. The number of FGPs tends to be greater in FAP than that seen in patients consuming PPIs, although there is some overlap.
Gastric adenomas also occur in FAP patients. The incidence of gastric adenomas in Western patients has been reported to be between 2% and 12%, whereas in Japan, it has been reported to be between 39% and 50%.
These adenomas can progress to carcinoma. FAP patients in Korea and Japan are reported to have a threefold to fourfold increased risk of gastric cancer compared with the general population in those countries, a finding not observed in Western populations.
One potential explanation for a higher prevalence of gastric adenomas in Asian FAP patients than that seen in Western FAP patients may be the higher overall prevalence of Helicobacter pylori infection.
More recently, a rise in incidence of gastric adenocarcinoma was observed in a Western FAP database.
Alterations in the promoter (1B) of APC were discovered in families with gastric adenocarcinoma and proximal polyposis of the stomach (GAPPS), who express numerous, predominantly fundic gland, gastric polyps restricted to the body and fundus with regions of dysplasia or gastric adenocarcinoma, and no evidence of colorectal or duodenal polyposis. These variants segregated with the gastric phenotype in multiple GAPPS families. Although penetrance of the gastric polyposis phenotype is high, the phenotype can vary ranging from asymptomatic adults to teenagers presenting with massive symptomatic gastric polyposis, as well as unaffected carriers who had clean endoscopies at ages ranging from 42 to 77 years. However, the penetrance for gastric cancer is less clear. Promoter 1B APC alterations rarely occur in FAP families with gastric fundic gland polyps and colonic polyposis.
Whereas the incidence of duodenal adenomas is only 0.4% in unselected patients undergoing upper GI endoscopy,
duodenal adenomas are found in 80% to 100% of FAP patients. Most are located in the first and second portions of the duodenum, especially in the periampullary region.
There is a 4% to 12% lifetime incidence of duodenal adenocarcinoma in FAP patients.
In a prospective multicenter surveillance study of duodenal adenomas in 368 participants from northern Europe with FAP, 65% had adenomas at baseline evaluation (mean age, 38 y), with cumulative prevalence reaching 90% by age 70 years. In contrast to earlier beliefs regarding an indolent clinical course, the adenomas increased in size and degree of dysplasia during the 8 years of average surveillance, although only 4.5% developed cancer while under prospective surveillance.
This is a large study; however, it is limited by the use of forward-viewing rather than side-viewing endoscopy and the large number of investigators involved in the study. Intestinal polyps can also be assessed in FAP patients using capsule endoscopy.
One study of computed tomography (CT) duodenography found that larger adenoma size could be accurately measured but smaller, flatter adenomas could not be accurately counted.
A retrospective review of FAP patients suggested that the adenoma-carcinoma sequence occurred in a temporal fashion for periampullary adenocarcinomas with a diagnosis of adenoma at a mean age of 39 years, high-grade dysplasia at a mean age of 47 years, and adenocarcinoma at a mean age of 54 years.
A decision analysis of 601 FAP patients suggested that the benefit of periodic surveillance starting at age 30 years led to an increased life expectancy of 7 months.
Although polyps in the duodenum can be difficult to treat, small series suggest that they can be managed successfully with endoscopy but with potential morbidity—primarily from pancreatitis, bleeding, and duodenal perforation.
FAP patients with particularly severe duodenal polyposis, sometimes called dense polyposis, or with histologically advanced duodenal adenomas appear to be at the highest risk of developing duodenal adenocarcinoma.
Because the risk of duodenal adenocarcinoma is correlated with the number and size of polyps and the severity of dysplasia of the polyps, a stratification system that incorporates these features was developed to attempt to identify those individuals with FAP at the highest risk of developing duodenal adenocarcinoma.
According to this system, known as the Spigelman classification (refer to Table 5), 36% of patients with the most advanced stage will develop carcinoma.
Points | Polyp Number | Polyp Size (mm) | Histology | Dysplasia |
---|---|---|---|---|
1 | 1–4 | 1–4 | Tubular | Mild |
2 | 5–20 | 5–10 | Tubulovillous | Moderate |
3 | >20 | >10 | Villous | Severe |
Stage I, 1–4 points; Stage II, 5–6 points; Stage III, 7–8 points; Stage IV, 9–12 points. |
Other extracolonic tumors arising in FAP patients include papillary thyroid cancer, adrenal tumors, hepatoblastoma, and brain tumors.
Papillary thyroid cancer (cribriform morular type) has been reported to affect 1% to 2% of patients with FAP.
However, a study
of papillary thyroid cancers in six women with FAP failed to demonstrate loss of heterozygosity (LOH) or pathogenic variants of the wild-type allele in codons 545 and 1061 to 1678 of the six tumors. In addition, four of five of these patients had detectable somatic RET/PTC chimeric genes. This pathogenic variant is generally restricted to sporadic papillary thyroid carcinomas, suggesting the involvement of genetic factors other than APC pathogenic variants. Further studies are needed to show whether other genetic factors such as the RET/PTC chimeric gene are independently responsible for or cooperative with APC variants in causing papillary thyroid cancers in FAP patients.
Adrenal tumors have been reported in FAP patients, and one study demonstrated LOH at the APC locus in an adrenocortical carcinoma (ACC) in an FAP patient.
In a study of 162 FAP patients who underwent abdominal CT for evaluation of intra-abdominal desmoid tumors, 15 patients (11 women) were found to have adrenal tumors.
Of these, two had symptoms attributable to cortisol hypersecretion. Three of these patients underwent subsequent surgery and were found to have ACC, bilateral nodular hyperplasia, or adrenocortical adenoma. The prevalence of an unexpected adrenal neoplasia in this cohort was 7.4%, which compares with a prevalence of 0.6% to 3.4% (P < .001) in non-FAP patients.
No molecular genetic analyses were provided for the tumors resected in this series. A subsequent study identified adrenal lesions in 26% (23 of 90) of patients with FAP, 18% (2 of 11) of patients with AFAP, and 24% (5 of 21) of patients with MAP. Most lesions in this series followed a benign and slowly progressive course; no cases of ACC were reported.
Hepatoblastoma is a rare, rapidly progressive, and usually fatal childhood malignancy that, if confined to the liver, can be cured by radical surgical resection. Multiple cases of hepatoblastoma have been described in children with an APC pathogenic variant.
Some series have also demonstrated LOH of APC in these tumors.
No specific genotype-phenotype correlations have been identified in FAP patients with hepatoblastoma.
(Refer to the Hepatoblastoma section in the PDQ summary on Childhood Liver Cancer Treatment for more information.)
The constellation of CRC and brain tumors has been referred to as Turcot syndrome; however, Turcot syndrome is molecularly heterogeneous. Molecular studies have demonstrated that colon polyposis and medulloblastoma are associated with pathogenic variants in APC (thus representing FAP), while colon cancer and glioblastoma are associated with pathogenic variants in mismatch repair (MMR) genes (thus representing Lynch syndrome).
Medulloblastoma, a highly malignant embryonal central nervous system tumor, accounts for approximately 80% of the brain tumors found in FAP and primarily occurs in children with 70% diagnosed before age 16 years. High-grade astrocytomas and ependymomas have also been described in FAP patients. Although the relative lifetime risk of any brain tumor among members of an FAP family is increased 7-fold and that of medulloblastoma 90-fold, the absolute lifetime risk of any brain tumor is approximately 1% to 2%.
The APC gene on chromosome 5q21 encodes a 2,843-amino acid protein that is important in cell adhesion and signal transduction; the main function of the APC protein is to regulate intracellular concentrations of beta-catenin, a major mediator of the Wnt signal transduction pathway. APC is a tumor suppressor gene, and the loss of APC is among the earliest events in the chromosomal instability colorectal tumor pathway. FAP and AFAP can be diagnosed genetically by testing for germline pathogenic variants in the APC gene in DNA from peripheral blood leukocytes. More than 300 different disease-associated pathogenic variants of the APC gene have been reported.
Most of these changes are insertions, deletions, and nonsense variants that lead to frameshifts and/or premature stop codons in the resulting transcript of the gene. The most common APC pathogenic variant (10% of FAP patients) is a deletion of AAAAG in codon 1309; no other pathogenic variants appear to predominate. Variants that reduce rather than eliminate production of the APC protein may also lead to FAP.
Most APC pathogenic variants that occur between codon 169 and codon 1249 result in the classic FAP phenotype.
There has been much interest in correlating the location of the pathogenic variant within the gene with the clinical phenotype:
A low-penetrance APC variant, I1307K, has been studied for its association with CRC. (Refer to the APC I1307K section in the Colorectal Cancer Susceptibility Genes section of this summary for more information.)
Individuals who present with a classic FAP phenotype are candidates for APC testing. However, in many probands with a personal or family history of polyposis, multigene panel testing is an appropriate option to consider given the genetic heterogeneity of polyposis conditions and the phenotypic overlap among associated syndromes.
In particular, patients who develop fewer than 100 colorectal adenomatous polyps may pose a diagnostic challenge. The differential diagnosis includes AFAP, MAP, polymerase proofreading–associated polyposis (PPAP), and biallelic mismatch repair deficiency (BMMRD).
AFAP can be diagnosed by testing for germline APC pathogenic variants. (Refer to the Attenuated Familial Adenomatous Polyposis [AFAP] section of this summary for more information.) MAP is caused by biallelic germline pathogenic variants in the MUTYH gene, inherited in an autosomal recessive manner.
PPAP is caused by heterozygous pathogenic variants in POLE and POLD1.
BMMRD is a condition in which individuals inherit pathogenic variants in both alleles of one of the MMR genes (MLH1, MSH2, MSH6, PMS2, or EPCAM).
(Refer to the MUTYH-Associated Polyposis [MAP], Oligopolyposis, and Biallelic mismatch repair deficiency [BMMRD] sections of this summary for more information.)
For example, in a large cross-sectional study, pathogenic variants in APC were found in 80% (95% confidence interval [CI], 71%–87%) of individuals with more than 1,000 adenomas, 56% (95% CI, 54%–59%) in those with 100 to 999 adenomas, 10% (95% CI, 9%–11%) in those with 20 to 99 adenomas, and 5% (95% CI, 4%–7%) in those with 10 to 19 adenomas.
In this same study, the prevalence of biallelic MUTYH pathogenic variants was similar to APC for those with the attenuated phenotype (20–99 adenomas), but MUTYH pathogenic variants were also observed in a small minority (2%) of those with classic polyposis.
Most commercial laboratories perform not only full gene sequencing but also deletion/duplication analysis of the APC and other genes. However, it is important to verify the testing methodology with each laboratory. Deletion analysis is especially important for individuals with FAP because 8% to 12% of affected individuals have a whole exon deletion or promoter 1B deletion in the APC gene, which would not be detected with sequencing.
As mentioned, for patients who present with polyposis, multigene panels that include multiple polyposis genes are often ordered, which simplifies and lowers the cost of testing by assessing all genes at the same time. (Refer to the Multigene [panel] testing section in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for more information.)
In families in which a pathogenic variant in the APC gene is identified, predictive testing for at-risk relatives can definitively identify or rule out the variant. Such testing is important to determine whether at-risk relatives need to undergo aggressive screening or whether such procedures are not necessary or can be discontinued (i.e., in relatives who test negative for the familial pathogenic variant).
Most patients with FAP have an affected parent, and a pattern of autosomal dominant inheritance may be observed in the family. Accordingly, cascade genetic counseling and testing may then be extended to at-risk family members. However, it is estimated that 25% of patients with FAP have a de novo pathogenic variant in APC, meaning that the variant does not appear to be inherited from either parent.
In cases where the variant cannot be identified in leukocyte DNA of either parent, it is possible that germline mosaicism may explain the finding. Thus, siblings of an individual should always be offered APC testing, but testing aunts, uncles, and cousins of the proband would not be indicated.
The early appearance of clinical features of FAP and the subsequent recommendations for surveillance beginning at puberty raise special considerations relating to the genetic testing of minors.
In general, genetic testing of minors for hereditary cancer syndromes is not recommended unless the results are expected to inform medical management in childhood. Thus, FAP presents an example in which possible medical benefit justifies genetic testing of minors in families with a known pathogenic variant, especially for the anticipated 50% of at-risk children who will be found not to be carriers of pathogenic variants and who can thus be spared surveillance. In addition, testing infants for FAP can allow for hepatoblastoma surveillance until age 5 years. Otherwise, if at-risk minors are not tested, colonoscopy or flexible sigmoidoscopy is initiated between ages 10 to 15 years.
The psychological impact of such testing is addressed in the Psychosocial Issues in Hereditary Colon Cancer Syndromes section of this summary.
Individuals at risk of FAP, because of a known APC pathogenic variant in either the family or themselves, are evaluated for onset of polyposis by flexible sigmoidoscopy or colonoscopy. Once an FAP family member is found to manifest polyps, the only effective management to prevent CRC is colectomy. Prophylactic surgery has been shown to improve survival in patients with FAP.
If feasible, the patient and his/her family members should be included in a registry because it has been shown retrospectively that registration and surveillance reduce CRC incidence and mortality.
In patients with classic FAP identified very early in their course, the surgeon, endoscopist, and family may choose to delay surgery for several years in the interest of achieving social milestones. In addition, in carefully selected patients with AFAP (those with minimal polyp burden and advanced age), deferring a decision about colectomy may be reasonable with surgery performed only in the face of advancing polyp burden or dysplasia.
A Finnish nationwide population-based retrospective study evaluating whether surveillance of family members with FAP reduced overall mortality and improved survival demonstrated that family members of probands who were recruited to the screening program had equivalent survival to the general population up to 20 years after diagnosis of FAP.
The study included 154 families with at least one family member clinically diagnosed with FAP from 1963 to 2015. There were 194 probands and 225 family members (83 diagnosed by genetic testing and 142 by endoscopy) with a median time of follow-up of 11.8 years. In this study, the survival analysis of members of FAP families was calculated using the relative survival estimate.
This estimation compares survival among FAP probands and family members with the survival expected in the absence of FAP among individuals of the same gender and age in each calendar year. The relative survival for probands was 67% (95% CI, 60%–75%) after 10 years of follow-up and 66% (95% CI, 58%–76%) after 20 years. For family members, the relative survival was 98% (95% CI, 95%–101%) at 10 years and 94% (95% CI, 88%–100%) at 20 years. At 25 years of follow-up, the relative survival for family members was lower than the general population at 87% (95% CI, 79%–96%). The relative survival was significantly lower for probands than for family members (P < .001). In terms of mortality, the standardized mortality ratio was elevated in probands in both the 0- to 5-year and 5- to 10-year periods of follow-up whereas it remained stable for family members until 20 years of follow-up. This difference was more marked in the beginning of follow-up for probands taking into account the fact that probably most were symptomatic, and most likely had CRC at the diagnosis. The authors pointed out that if the CRC was treated successfully without recurrence, the survival of the probands approached that of the family members.
Endoscopic surveillance usually begins early (age, 10–15 y).
(Refer to the Psychosocial Issues in Hereditary Colon Cancer Syndromes section of this summary for more information on the social and emotional implications of early surveillance.) Historically, sigmoidoscopy may have been a reasonable approach in identifying early adenomas in most patients. However, colonoscopy is the tool of choice in light of (a) improved instrumentation for full colonoscopy; (b) sedation; (c) recognition of AFAP, in which the disease is typically most manifest in the right colon; and (d) the growing tendency to defer surgery for a number of years.
Individuals who have tested negative for an otherwise known family pathogenic variant do not need FAP-oriented endoscopic surveillance; they are recommended by NCCN to undergo average-risk population screening. In the case of families in which no family variant has been identified in an affected person, clinical surveillance is warranted. Colon surveillance is not stopped in persons who are known to carry an APC pathogenic variant but who do not yet manifest polyps, because adenomas occasionally are not manifest until the fourth and fifth decades of life. (Refer to the PDQ summary on Colorectal Cancer Screening for more information on these methods.)
Colon adenomas will develop in nearly 100% of persons who are APC pathogenic variant–positive; risk-reducing surgery comprises the standard of care to prevent CRC after polyps have appeared and are too numerous or histologically advanced to monitor safely using endoscopic resection.
FAP patients and their doctors should have an individualized discussion to decide when surgery will be performed. It is useful to incorporate into the discussion the risk of developing desmoid tumors after surgery, as well as fecundity for women. Timing of risk-reducing surgery usually depends on the number of polyps, their size, histology, and symptomatology.
Once numerous polyps have developed, surveillance colonoscopy is no longer useful in timing the colectomy because polyps are so numerous that it is not possible to biopsy or remove all of them. At this time, it is appropriate for patients to consult with a surgeon who is experienced with available options, including total colectomy and restorative proctocolectomy.
Rectum-sparing surgery, with sigmoidoscopic surveillance of the remaining rectum, is a reasonable alternative to total colectomy in those compliant individuals with relative rectal sparing of polyps who understand the consequences and make an informed decision to accept the residual risk of rectal cancer occurring despite periodic surveillance.
Surgical options include restorative proctocolectomy with ileal pouch anal anastomosis (IPAA), total colectomy with ileorectal anastomosis (IRA), or total proctocolectomy with ileostomy (TPC). TPC is reserved for patients with low rectal cancer in which the sphincter cannot be spared or for patients on whom an IPAA cannot be performed because of technical problems. There is no risk of developing rectal cancer after TPC because the whole mucosa at risk is removed. These procedures can be performed utilizing minimally invasive techniques.
Irrespective of whether a colectomy and an IRA or a restorative proctocolectomy is performed, most experts suggest that periodic and lifelong surveillance of the rectum or the ileal pouch be performed to remove or ablate any polyps. In earlier unselected studies, the risk of rectal cancer after total colectomy 20 years after IRA was reported to be as high as 25%.
This risk has been reported to be much lower with better selection of patients for IRA.
Factors that have been reported to increase rectal cancer risk after IRA include the number of polyps throughout the colon, the number of polyps in the rectum, the presence of colon cancer at the time of IRA, the length of the rectal stump, the duration of follow-up after IRA, and the genotype.
An abdominal colectomy with IRA as the primary surgery for FAP does not preclude later conversion to an IPAA for uncontrolled rectal polyps and/or rectal cancer. In the Danish Polyposis Registry, the morbidity and functional results of a secondary IPAA (after a previous IRA) in 24 patients were reported to be similar to those of 59 patients who underwent primary IPAA.
In most cases, the clinical polyp burden in the rectum at the time of surgery dictates the type of surgical intervention, namely, restorative proctocolectomy with IPAA versus IRA. Patients with a mild phenotype (<1,000 colonic adenomas) and fewer than 20 rectal polyps may be candidates for IRA at the time of prophylactic surgery.
In some cases, however, the polyp burden is equivocal, and in such cases, investigators have considered the role of genotype in predicting subsequent outcomes with respect to the rectum.
Pathogenic variants reported to increase the rectal cancer risk and eventual completion proctectomy after IRA include variants in exon 15 codon 1250, exon 15 codons 1309 and 1328, and exon 15 variants between codons 1250 and 1464.
In patients who have undergone IPAA, it is important to continue annual surveillance of the ileal pouch because the cumulative risk of developing adenomas in the pouch has been reported to be up to 75% at 15 years.
Although they are rare, carcinomas have been reported in the ileal pouch and anal transition zone after restorative proctocolectomy in FAP patients.
A meta-analysis of quality of life after restorative proctocolectomy and IPAA has suggested that patients with FAP do marginally better than patients with inflammatory bowel disease in terms of fistula formation, pouchitis, stool frequency, and seepage.
Celecoxib, a specific cyclooxygenase 2 (COX-2) inhibitor, and nonspecific COX-2 inhibitors, such as sulindac (a nonsteroidal anti-inflammatory drug [NSAID]), have been associated with a decrease in polyp size and number in FAP patients, suggesting a role for chemopreventive agents in the treatment of this disorder.
Although celecoxib had been approved by the U.S. Food and Drug Administration (FDA), its license was voluntarily withdrawn by the manufacturer. Currently, there are no FDA-approved drugs for chemoprevention in FAP. Nevertheless, agents such as celecoxib and sulindac are in sufficiently widespread use that chemopreventive clinical trials typically utilize one of these agents as the control arm. A randomized trial showed possible marginal improvement in polyp burden with the combination of celecoxib and difluoromethylornithine, compared with celecoxib alone.
A small, randomized, placebo-controlled, dose-escalation trial of celecoxib in a pediatric population (aged 10–14 y) demonstrated the safety of celecoxib at all dosing levels when administered over a 3-month period.
This study found a dose-dependent reduction in adenomatous polyp burden. At a dose of 16 mg/kg/day, which approximates the approved dose of 400 mg twice daily in adults, the reduction in polyp burden paralleled that demonstrated with celecoxib in adults.
Omega-3-polyunsaturated fatty acid eicosapentaenoic acid in the free fatty acid form has been shown to reduce rectal polyp number and size in a small study of patients with FAP after subtotal colectomy.
Although not directly compared in a randomized trial, the effect appeared to be similar in magnitude to that previously observed with celecoxib.
It is unclear at present how to incorporate COX-2 inhibitors into the management of FAP patients who have not yet undergone risk-reducing surgery. A double-blind placebo-controlled trial of 41 child and young adult carriers of APC pathogenic variants who had not yet manifested polyposis demonstrated that sulindac may not be effective as a primary treatment in FAP. There were no statistically significant differences between the sulindac and placebo groups over 4 years of treatment in incidence, number, or size of polyps.
Consistent with the effects of COX-2 inhibitors on colonic polyps, in a randomized, prospective, double-blind, placebo-controlled trial, celecoxib reduced, but did not eliminate, the number of duodenal polyps in 32 patients with FAP after a 6-month course of treatment. Of importance, a statistically significant effect was seen only in individuals who had more than 5% of the duodenum involved with polyps at baseline and with an oral dose of 400 mg, given twice daily.
A previous randomized study of 24 FAP patients treated with sulindac for 6 months showed a nonsignificant trend in the reduction of duodenal polyps.
The same issues surrounding the use of COX-2 inhibitors for the treatment of colonic polyps apply to their use for the treatment of duodenal polyps (e.g., only partial elimination of the polyps, complications secondary to the COX-2 inhibitors, and loss of effect after the medication is discontinued).
Because of the common clustering of adenomatous polyps around the duodenal papilla (where bile enters the intestine) and preclinical data suggesting that ursodeoxycholate inhibits intestinal adenomas in mice that harbor an Apc germline variant,
two trials that employ ursodeoxycholate have been performed.
In both studies, ursodeoxycholate did not have a significant chemopreventive effect on duodenal polyps; paradoxically, in one study, ursodeoxycholate in combination with celecoxib appeared to promote polyp density in patients with FAP.
Because of reports demonstrating an increase in cardiac-related events in patients taking rofecoxib and celecoxib,
it is unclear whether this class of agents will be safe for long-term use for patients with FAP and in the general population. Also, because of the short-term (6 months) nature of these trials, there is currently no clinical information about cardiac events in FAP patients taking COX-2 inhibitors on a long-term basis.
One cohort study has demonstrated regression of colonic and rectal adenomas with sulindac treatment in FAP. The reported outcome of this trial was the number and size of polyps, a surrogate for the clinical outcome of main interest, CRC incidence.
Preclinical studies of a small-molecule epidermal growth factor receptor (EGFR) inhibitor and low-dose sulindac in the Apcmin/+ mouse diminished intestinal adenoma development by 87%
suggesting that EGFR inhibitors had the potential to inhibit duodenal polyps in FAP patients. A 6-month double-blind, randomized, placebo-controlled trial tested the efficacy of sulindac, 150 mg twice daily, and erlotinib, 75 mg daily, versus placebo in FAP or AFAP patients with duodenal polyps.
Ninety-two patients with FAP or AFAP were randomly assigned to receive study drugs or placebo and underwent pretreatment and posttreatment upper endoscopies to determine the changes in the sum diameter of the polyps and number of polyps in a 10 cm segment of proximal duodenum. The trial was terminated prematurely because the primary endpoint was met. The intent-to-treat analysis demonstrated a median decrease in duodenal polyp burden (sum of diameters) of 8.5 mm in the sulindac/erlotinib arm while there was an 8 mm increase in the placebo arm (P < .001). Significantly higher rates of grade 1 and grade 2 adverse events occurred in the treatment arm than in the placebo arm: in the treatment arm, 60.9% developed an acneiform rash and 32.6% developed oral mucositis; in the placebo arm, 19.6% developed an acneiform rash and 10.9% developed oral mucositis. On the basis of the previously modest effects of sulindac and celecoxib on duodenal polyps in FAP patients
and the dramatic effect of genetic EGFR inhibition on intestinal adenoma development in the Apcmin/+ mouse,
it is likely that erlotinib was responsible for the success of this trial. An ongoing clinical trial is determining whether lower doses of erlotinib alone are sufficient for significantly reducing duodenal polyp burden in FAP and AFAP patients.
Patients who carry APC germline pathogenic variants are at increased risk of other types of malignancies, including desmoid tumors, gastric tumors, duodenal cancer, small bowel cancer, hepatoblastoma, thyroid cancer, and brain tumors. The management of these extracolonic tumors is described below.
The management of desmoids in FAP can be challenging and can complicate prevention efforts. There is no accepted standard treatment for desmoid tumors. Multiple medical treatments have generally been unsuccessful in the management of desmoids. Treatments have included antiestrogens, NSAIDs, chemotherapy, and radiation therapy, among others. Studies have evaluated the use of raloxifene alone, tamoxifen or raloxifene combined with sulindac, and pirfenidone alone.
Thirteen patients with intra-abdominal desmoids and/or unfavorable response to other medical treatments who had expression of estrogen-alpha receptors in their desmoid tissues were included in a prospective study of raloxifene, given in doses of 120 mg daily.
Six patients had been on tamoxifen or sulindac before treatment with raloxifene, and seven patients were previously untreated. All 13 patients with intra-abdominal desmoid disease had either a partial or a complete response 7 months to 35 months after starting treatment, and most desmoids decreased in size at 4.7 months (± 1.8 mo) after treatment. Response occurred in patients with desmoid plaques and with distinct lesions. Study limitations include small sample size and the clinical evaluation of response, which was not consistent in all patients. Several questions remain concerning the outcomes of patients with desmoid tumors not expressing estrogen-alpha receptors who have received raloxifene, as well as which patients may benefit from this potential treatment.
A second study of 13 patients with FAP-associated desmoid tumors, who were treated with tamoxifen 120 mg/day or raloxifene 120 mg/day in combination with sulindac 300 mg/day, reported that ten patients had either stable disease (n = 6) or a partial or complete response (n = 4) for more than 6 months and that three patients had stable disease for more than 30 months.
These results suggest that the combination of these agents may be effective in slowing the growth of desmoid tumors. However, the natural history of desmoids is variable, with both spontaneous regression and variable growth rates.
A third study reported mixed results in 14 patients with FAP-associated desmoid tumors treated with pirfenidone for 2 years.
In this study, some patients had disease regression, some patients had disease progression, and some patients had stable disease.
There are reports of using imatinib mesylate to treat desmoid tumors in FAP patients with some success.
Nilotinib demonstrated potential to stabilize desmoid tumor growth after treatment failure with imatinib in patients with desmoid tumors.
The benefit of the tyrosine kinase inhibitor sorafenib in the treatment of desmoid tumors was demonstrated in a phase III randomized trial comparing sorafenib (400 mg daily) with placebo in 87 patients with unresectable progressive or symptomatic desmoid tumors.
Crossover to the sorafenib group was permitted for patients in the placebo group who had disease progression on the placebo arm of the study. Objective responses were demonstrated in 16 of 49 patients treated with sorafenib (33%) compared with 7 of 35 placebo-treated patients (20%). Additionally, the two-year progression-free survival (PFS) rate was significantly higher for sorafenib (81%) than placebo (36%); the hazard ratio for progression or death was 0.13 (95% CI, 0.05–0.31; P < .001). The most frequently reported adverse events were grade 1 or grade 2 rash (73%), fatigue (67%), hypertension (55%), and diarrhea (51%). Despite a relatively favorable toxicity profile, approximately 20% of patients discontinued sorafenib due to toxicity, emphasizing the importance of appropriate dose delays and interruptions for the treatment of adverse events.
Because of the high rates of morbidity and recurrence, in general, surgical resection is not recommended in the treatment of intra-abdominal desmoid tumors. A review of experiences at one hospital suggested that surgical outcomes with intra-abdominal desmoids may be better than previously believed.
Issues of subject selection are critical in evaluating surgical outcome data.
Abdominal wall desmoids can be treated with surgical resection, but the recurrence rate is high.
It is not clear what should be done with gastric adenomas. Only retrospective case series are available and point to a relatively low prevalence of gastric adenocarcinoma development in FAP patients.
More recently, a rise in incidence of gastric adenocarcinoma was observed in a Western FAP database
suggesting that a possible change in the management of gastric tumorigenesis in FAP may be in order. One group recommends endoscopic polypectomy for the management of gastric adenomas.
The management of adenomas in the stomach is usually individualized on the basis of the size of the adenoma and the degree of dysplasia.
A baseline upper endoscopy, including side-viewing duodenoscopy, is typically performed between ages 25 and 30 years in FAP patients.
The subsequent intervals between endoscopy vary according to the findings of the previous endoscopy, often based on Spigelman stage. Recommended intervals are based on expert opinion although the relatively liberal intervals for stage 0 to stage II disease are based in part on the natural history data generated by the Dutch/Scandinavian duodenal surveillance trial (refer to Table 6 for available recommendations regarding screening frequency by Spigelman stage).
The main advantages of the Spigelman classification are its long-standing familiarity to and usage by those in the field, which allows reasonable standardization of outcome comparisons across studies.
However, the following are limitations of application of the Spigelman classification:
Spigelman Stage | NCCN (2019) | ESMO (2013) |
---|---|---|
0 (no polyps) | Endoscopy every 4 y | Not specified |
I | Endoscopy every 2–3 y | Endoscopy every 5 y |
II | Endoscopy every 1–3 y | Endoscopy every 3 y |
III | Endoscopy every 6–12 mo | Endoscopy every 1–2 y |
IV | Surgical evaluation | Endoscopy every 6-12 mo |
Complete mucosectomy or duodenectomy, or Whipple procedure if duodenal papilla is involved | ||
Expert endoscopic surveillance every 3–6 mo | Surgical options include duodenotomy with polypectomy, pancreas-sparing duodenectomy and pancreaticoduodenectomy (Whipple procedure) | |
ESMO = European Society of Medical Oncology; NCCN = National Comprehensive Cancer Network. | ||
See below for additional information about the use of surgical resection in Spigelman stage IV disease. |
The results of long-term duodenal adenoma surveillance of FAP patients in Nordic countries and the Netherlands revealed significant duodenal cancer risk in FAP patients.
According to the protocol, biennial frontal-viewing endoscopy was performed from 1990 through 2000. Subsequently, patients were followed up with surveillance according to international guidelines. The study group comprised 261 of 304 patients (86%) who had more than one endoscopy. Median follow-up was 14 years (range, 9–17 y). The lifetime risk of duodenal adenomatosis was 88%. Forty-four percent of patients had worsening Spigelman stage over time, whereas 12% improved and 34% remained unchanged. Twenty patients (7%) developed duodenal cancer at a median age of 56 years (range, 44–82 y). The cumulative cancer incidence was 18% at age 75 years (95% CI, 8%–28%). Survival in patients with symptomatic cancers was worse than those diagnosed at surveillance endoscopy.
Many factors, including severity of polyposis, comorbidities, patient preferences, and availability of adequately trained physicians, determine whether surgical or endoscopic therapy is selected for polyp management. Endoscopic resection or ablation of large or histologically advanced adenomas appears to be safe and effective in reducing the short-term risk of developing duodenal adenocarcinoma;
however, patients managed with endoscopic resection of adenomas remain at substantial risk of developing recurrent adenomas in the duodenum.
The most definitive procedure for reducing the risk of adenocarcinoma is surgical resection of the ampulla and duodenum, although these procedures also have higher morbidity and mortality associated with them than do endoscopic treatments. Duodenotomy and local resection of duodenal polyps or mucosectomy have been reported, but invariably, the polyps recur after these procedures.
In a series of 47 patients with FAP and Spigelman stage III or stage IV disease who underwent definitive radical surgery, the local recurrence rate was reported to be 9% at a mean follow-up of 44 months. This local recurrence rate was dramatically lower than any local endoscopic or surgical approach from the same study.
Pancreaticoduodenectomy and pancreas-sparing duodenectomy are appropriate surgical therapies that are believed to substantially reduce the risk of developing periampullary adenocarcinoma.
If such surgical options are considered, preservation of the pylorus is of particular benefit in this group of patients because most will have undergone a subtotal colectomy with IRA or total colectomy with IPAA. As noted in a Northern European study,
and others,
most patients with duodenal adenomas will not develop cancer and can be followed with endoscopy. However, individuals with advanced adenomas (Spigelman stage III or stage IV disease) generally require endoscopic or surgical treatment of the polyps. Chemoprevention studies for duodenal adenomas in FAP patients are under way and may offer an alternate strategy in the future. (Refer to the Chemoprevention section of this summary for more information.)
The endoscopic approach to larger and/or flatter adenomas of the duodenum depends on whether the ampulla is involved. Endoscopic mucosal resection (EMR) after submucosal injection of saline, with or without epinephrine and/or dye, such as indigo carmine, can be employed for nonampullary lesions. Ampullary lesions require even greater care including endoscopic ultrasound evaluation for evidence of bile or pancreatic duct involvement. Stenting of the pancreatic duct is commonly performed to prevent stricturing and pancreatitis. The stents require endoscopic removal at an interval of 1 to 4 weeks. Because the ampulla is tethered at the ductal orifices, it typically does not uniformly lift with injection, so injection is commonly not used. Any consideration of EMR or ampullectomy requires great experience and judgment, with careful consideration of the natural history of untreated lesions and an appreciation of the high rate of adenoma recurrence despite aggressive endoscopic intervention.
The literature uniformly supports duodenectomy for Spigelman stage IV disease. For Spigelman stage II and stage III disease, there is a role for endoscopic treatment invariably focusing on the one or two worst lesions that are present.
Reluctance to consider surgical resection is related to the short-term morbidity and mortality and the long-term complications related to surgery. Although these concerns are likely overstated,
fear of surgical intervention can lead to aggressive and somewhat ill-advised endoscopic interventions. In some circumstances, endoscopic resection of ampullary and/or other duodenal adenomas cannot be accomplished completely or safely by endoscopic means, and duodenectomy cannot be accomplished without risking a short-gut syndrome or cannot be done at all because of mesenteric fibrosis. In such cases, surgical transduodenal ampullectomy/polypectomy can be performed. However, this is associated with a high risk of local recurrence similar to that of endoscopic treatment.
Although level 1 evidence is lacking, a consensus opinion recommends annual thyroid examinations beginning in the late teenage years to screen for papillary thyroid cancer in patients with FAP. The same panel suggests clinicians could consider the addition of annual thyroid ultrasonography to this screening routine.
Although level 1 evidence is lacking, a consensus panel has suggested that liver palpation, abdominal ultrasonography, and measurement of serum alpha-fetoprotein every 3 to 6 months for the first 5 years of life in children with a predisposition to FAP be considered.
It is not necessary to continue screening after age 5 years.
Medulloblastoma is a highly malignant tumor that is usually only symptomatic 6 months or less before diagnosis; annual surveillance of asymptomatic patients may be insufficient. Thus, surveillance by means of regular CT or magnetic resonance imaging cannot be advocated. FAP family members who do not yet have polyposis, but have signs or symptoms suggestive of a brain tumor, should be evaluated with neuroimaging because brain tumors present before the diagnosis of polyposis in more than half of FAP patients. Careful evaluation is also important among FAP families in which one member already has a brain tumor because familial clustering occurs. Of such families with FAP-associated brain tumors, 40% had two affected members.
AFAP was first described clinically in 1990 in a large kindred with a variable number of adenomas. The average number of adenomas in this kindred was 30, although they ranged in number from a few to hundreds.
It has been recommended that the management of AFAP patients include colonoscopy rather than flexible sigmoidoscopy because the adenomas can be predominantly right-sided.
Adenomas in AFAP are believed to form around the age of mid-twenties to late twenties.
Similar to classic FAP, the risk of CRC is higher in individuals with AFAP; the average age at diagnosis, however, is older than classic FAP at 56 years.
Affected family members have developed CRCs with very few synchronous polyps.
Extracolonic manifestations similar to those in classic FAP also occur in AFAP. These manifestations include upper GI polyps (FGPs, duodenal adenomas, and duodenal adenocarcinoma), osteomas, epidermoid cysts, and desmoid tumors.
Because of the specific sites of APC pathogenic variants causing AFAP, these patients typically lack CHRPE lesions.
AFAP is associated with particular subsets of APC pathogenic variants. Three groups of site-specific APC pathogenic variants causing AFAP have been characterized:
In the absence of family history of similarly affected relatives, the differential diagnosis may include AFAP (including MAP), Lynch syndrome, BMMRD, germline variants in the DNA polymerase proofreading subunits (POLD1 or POLE), or an otherwise unclassified sporadic or genetic problem. A careful family history may implicate AFAP or Lynch syndrome.
APC testing is an important component of the evaluation of patients suspected of having AFAP.
If germline APC pathogenic variant testing is negative in suspected AFAP individuals, genetic testing for MUTYH, POLE, and POLD1 pathogenic variants may be warranted.
Patients found to have an unusually or unacceptably high adenoma count at an age-appropriate colonoscopy pose a differential diagnostic challenge.
The role for and timing of risk-reducing colectomy in AFAP is controversial.
Table 7 summarizes the clinical practice guidelines from different professional societies regarding surveillance of AFAP.
Organization | Condition | Screening Method | Screening Frequency | Age Screening Initiated | Comment |
---|---|---|---|---|---|
Europe Mallorca Group (2008) | AFAP | Colonoscopy | Every 2 y; every 1 y if adenomas are detected | 18–20 y | |
NCCN (2019) | Personal history of AFAP with small adenoma burden a | Colonoscopy | Every 1–2 y | If patient had colectomy with IRA, endoscopic evaluation every 6–12 mo depending on polyp burden. | |
Colectomy and IRA may be considered in patients aged ≥21 y. | |||||
NCCN (2019) | Personal history of AFAP with adenoma burden that cannot be handled endoscopically | Not applicable | Not applicable | Not applicable | Colectomy with IRA preferred. Consider proctocolectomy with IPAA if dense rectal polyposis. |
NCCN (2019) | Unaffected at-risk family member; family pathogenic variant known; APC pathogenic variant status unknown or positive | Colonoscopy | Every 1–2 y if APC positive; every 2–3 y if unknown | Late teens | If APC pathogenic variant status not tested, consider genetic testing. |
IPAA = ileal pouch–anal anastomosis; IRA = ileorectal anastomosis; NCCN = National Comprehensive Cancer Network. | |||||
aFewer than 20 adenomas that are each <1 cm in diameter and without advanced histology so that colonoscopy with polypectomy can be used to effectively eliminate the polyps. |
MAP is an autosomal recessively inherited polyposis syndrome caused by pathogenic variants in the Mut Y homolog gene. The Mut Y homolog gene, which is known as MUTYH, was initially called MYH, but was subsequently corrected because the myosin heavy chain gene already had that designation. MUTYH is located on chromosome 1p34.3-32.1.
The protein encoded by MUTYH is a base excision repair glycosylase, which repairs one of the most common forms of oxidative damage. Over one hundred unique sequence variants of MUTYH have been reported (Leiden Open Variation Database). A founder pathogenic variant with ethnic differentiation is assumed for MUTYH pathogenic variants. In white populations of northern European descent, two major variants, Y179C and G396D (formerly known as Y165C and G382D), account for 70% of biallelic pathogenic variants in MAP patients; 90% of these patients carry at least one of these pathogenic variants.
Other causative variants that have been found include P405L (formerly known as P391L) (Netherlands),
E480X (India),
Y104X (Pakistan),
1395delGGA (Italy),
1186-1187insGG (Portugal),
and p.A359V (Japan and Korea).
The MUTYH gene was first linked to polyposis in 2002 in three siblings with multiple colonic adenomas and CRC but no APC pathogenic variant.
MAP has a broad clinical spectrum. Most often it resembles the clinical picture of AFAP, but it has been reported in individuals with phenotypic resemblance to classical FAP and Lynch syndrome.
MAP patients tend to develop fewer adenomas at a later age than patients with APC pathogenic variants
but still carry a high risk of CRC (35%–75%).
A 2012 study of colorectal adenoma burden in 7,225 individuals reported a prevalence of biallelic MUTYH pathogenic variants of 4% (95% CI, 3%–5%) among those with 10 to 19 adenomas, 7% (95% CI, 6%–8%) among those with 20 to 99 adenomas, and 7% (95% CI, 6%–8%) among those with 100 to 999 adenomas.
This broad clinical presentation results from the MUTYH gene's ability to cause disease in its homozygous or compound heterozygous forms. Based on studies from multiple FAP registries, approximately 7% to 19% of patients with an FAP phenotype and without a detectable APC germline pathogenic variant carry biallelic variants in the MUTYH gene.
Adenomas, serrated adenomas, and hyperplastic polyps can be seen in MAP patients.
The CRCs tend to be right-sided and synchronous at presentation and seem to carry a better prognosis than sporadic CRC.
Clinical management guidelines for MAP range between once a year to every 3 years for colonoscopic surveillance beginning at age 18 to 30 years,
with upper endoscopic surveillance beginning at age 25 to 30 years.
(Refer to Table 8 for more information about available clinical practice guidelines for colon surveillance in MAP patients.) The recommended upper endoscopic surveillance interval can be based on the burden of involvement according to Spigelman criteria.
Total colectomy with ileorectal anastomosis or subtotal colectomy may be necessary for patients with MUTYH-associated polyposis depending on overall polyp burden.
Although MAP is the only known biallelic (recessive) adenoma cancer predisposition syndrome described to date, there are examples of biallelic cases presenting with childhood tumors in which MMR genes are involved. (Refer to the Biallelic mismatch repair deficiency section in the Lynch syndrome section of this summary for more information.)
Table 8 summarizes the clinical practice guidelines from different professional societies regarding colon surveillance of biallelic MAP.
Organization | Condition | Screening Method | Screening Frequency | Age Screening Initiated | Comment |
---|---|---|---|---|---|
Europe Mallorca Group (2008) | Carrier of MUTYH pathogenic variants | Colonoscopy | Every 2 y | 18–20 y | |
Nieuwenhuis et al. (2012) | Carrier of MUTYH pathogenic variants | Colonoscopy | Every 1–2 y | ||
NCCN (2019) | Personal history of MAP, small adenoma burden a | Colonoscopy | Every 1–2 y | If patient had colectomy with IRA, endoscopic evaluation every 6–12 mo depending on polyp burden. | |
Colectomy and IRA may be considered in patients aged ≥21 y. | |||||
NCCN (2019) | Personal history of MAP with adenoma burden that cannot be handled endoscopically | Not applicable | Not applicable | Not applicable | Colectomy with IRA preferred. Consider proctocolectomy with IPAA if dense rectal polyposis. If patient had colectomy with IRA, then endoscopic evaluation of rectum every 6–12 mo depending on polyp burden. |
NCCN (2019) | Unaffected, at-risk family member; family pathogenic variant known; MUTYH pathogenic variant status unknown or positive (biallelic) | Colonoscopy | Every 1–2 y | 25–30 y | If positive for a single MUTYH pathogenic variant, colonoscopy every 5 y beginning at age 40 y or 10 y before age of FDR at CRC diagnosis, if applicable. Evidence is needed to inform screening recommendations for the following situations: the presence of a MUTYH variant and an SDR with CRC; and a monoallelic MUTYH carrier unaffected by CRC with no family history of CRC. |
CRC = colorectal cancer; FDR = first-degree relative; IPAA = ileal pouch–anal anastomosis; IRA = ileorectal anastomosis; NCCN = National Comprehensive Cancer Network; SDR = second-degree relative. | |||||
aFewer than 20 adenomas that are each <1 cm in diameter and without advanced histology so that colonoscopy with polypectomy can be used to effectively eliminate the polyps. |
Many extracolonic cancers have been reported in patients with MAP including gastric, small intestinal, endometrial, liver, ovarian, bladder, thyroid, and skin cancers (melanoma, squamous epithelial, and basal cell carcinomas).
Additionally, noncancerous extracolonic manifestations have been reported in a few MAP patients including lipomas, congenital hypertrophy of the retinal pigment epithelium, osteomas, and desmoid tumors.
Female MAP patients have an increased risk of breast cancer.
These extracolonic manifestations seem to occur less frequently in MAP than in FAP, AFAP, or Lynch syndrome.
Similar to FAP, individuals with MAP often develop duodenal adenomas, and are at risk of developing duodenal cancer. Given the relatively recent identification of MAP compared with FAP, the incidence of duodenal polyps and risk of duodenal cancer in MAP is less well defined. Small case series have suggested the incidence of duodenal polyps in MAP to be approximately 30%, considerably lower than that of FAP. In a registry-based study the prevalence of duodenal polyps was 17%; however, only 50% of individuals in this study had undergone an upper GI endoscopy, suggesting the incidence of duodenal polyps was likely underestimated. The lifetime risk of duodenal cancer was estimated to be 4%.
A registry study from the United Kingdom and the Netherlands explored incidence of duodenal polyps and duodenal cancer in a group of patients with MAP who were undergoing regular duodenal surveillance.
Of 92 patients, 31 (34%) had evidence of duodenal polyps. The median age at duodenal adenoma detection was 50 years, and in 65% of patients duodenal adenomas were diagnosed at baseline endoscopy. Eighty-four percent of patients had Spiegelman stage I or stage II polyposis at first detection of polyps, with no patients with stage IV polyposis and no high-grade dysplasia detected. In subsequent surveillance only two patients progressed to Spiegelman stage IV polyposis, after 3.6 and 7.0 years, respectively. There additionally appeared to be sparing of the ampulla, with only two individuals having diminutive polyps without dysplasia in the ampulla. No cancers were detected in patients enrolled in upper GI surveillance programs within these registries. Two individuals with MAP were diagnosed with ampullary and duodenal cancer respectively at ages 83 and 63 years at the time of first-ever upper GI endoscopies. Therefore, duodenal polyps appear less prevalent in MAP compared with FAP, and appear at a later age. On the basis of these results, the authors suggest upper GI endoscopic screening in MAP be initiated at age 35 years.
Because MAP has an autosomal recessive inheritance pattern, siblings of an affected patient have a 25% chance of also carrying biallelic MUTYH pathogenic variants and should be offered genetic testing. Similarly, testing can be offered to the partner of an affected patient so that the risk in their children can be assessed.
The clinical phenotype of monoallelic MUTYH pathogenic variants is less well characterized with respect to incidence and associated clinical phenotypes, and its role in susceptibility to polyposis and colorectal carcinoma remains unclear. Approximately 1% to 2% of the general population carry a pathogenic variant in MUTYH.
A 2011 meta-analysis found that carriers of monoallelic MUTYH pathogenic variants are at modestly increased risk of CRC (odds ratio [OR], 1.15; 95% CI, 0.98–1.36); however, given the rarity of carriers of monoallelic pathogenic variants, they account for only a trivial proportion of all CRC cases.
A large study of 2,332 heterozygotes among 9,504 relatives of 264 CRC cases with a MUTYH pathogenic variant found that the risk of CRC at age 70 years was 7.2% for men and 5.6% for women, irrespective of family history. Among those with an FDR with a CRC diagnosis before age 50 years, the risk at age 70 years was 12.5% for men and 10% for women.
Caution should be exercised in the interpretation of this study as the vast majority of carrier status from this study was imputed and not based on genotype. The authors felt the risk for MUTYH heterozygotes with an FDR with CRC was sufficiently high to warrant more intensive surveillance than the general population (but the same as for anyone with an FDR with CRC diagnosed before age 50 y).
MMR genes may interact with MUTYH and increase the risk of CRC. An association between MUTYH and MSH6 has been reported. Both proteins interact together in base excision repair processes. A study reported a significant increase of MSH6 pathogenic variants in carriers of monoallelic MUTYH pathogenic variants with CRC compared with noncarriers with CRC (11.5% vs. 0%; P = .037).
However, a German study failed to duplicate these findings.
Additionally, a larger study found no increased cancer risk for carriers of MMR pathogenic variants with a MUTYH variant compared with those with a MMR pathogenic variant alone.
Oligopolyposis is a popular term used to describe the clinical presentation of a polyp count or burden that is greater than anticipated in the course of screening in average-risk patients but that falls short of the requirement for a diagnosis of FAP. Thus, oligo-, Greek for few, can mean different things to different observers. While conceding a lack of consensus on the matter, the National Comprehensive Cancer Network (NCCN) committee on CRC screening suggests an AFAP diagnosis is worth considering when a lifetime aggregate of 10 to 99 adenomas are present. The term oligopolyposis will be used here to describe the circumstance in which the polyp count (generally adenoma) is large enough, with or without any attendant family history, to raise in the mind of the endoscopist the possibility of an inherited susceptibility.
A majority of patients with oligopolyposis involving adenomas are not found to have an underlying predisposition when evaluated for pathogenic variants in known predisposition genes. Such cases are generally managed as if they are at an increased risk of recurrent adenomas even when the colon can be cleared of polyps endoscopically.
AFAP resulting from pathogenic germline APC variants may be the most common cause of oligopolyposis where a specific causative germline alteration cancer has been identified. Some AFAP cases with oligopolyposis will eventually develop more than 100 adenomas, albeit at a later age and often with a predominance of microadenomas of the right colon and with fewer, larger polyps in the left colon. Cases with a positive family history and an APC pathogenic variant are clearly variant cases of FAP, as the term AFAP implies.
However, patients with no immediate family history and a lesser adenoma burden may not be found to have an APC pathogenic variant. The lower the polyp count the lower the probability of having an APC pathogenic variant. Some of these cases are now known to carry biallelic MUTYH pathogenic variants or variants in other genes linked to oligopolyposis.
Pathogenic variants in related DNA polymerase genes POLE and POLD1 have been described in families with oligopolyposis, CRC, and endometrial cancer, and this condition has come to be known as polymerase proofreading–associated polyposis (PPAP).
An elegant approach was employed using whole-genome sequencing in 15 selected patients with more than ten adenomas before age 60 years. Several had a close relative with at least five adenomas who could also have whole-genome sequencing performed. All tested patients had CRC or a first-degree relative (FDR) with CRC. All had negative APC, MUTYH, and MMR gene pathogenic variant test results. No variants were found to be in common among the evaluated families. In one family, however, linkage had established shared regions, in which one shared variant was found (POLE p.Leu424Val; c.1270C>G), with a predicted major derangement in protein structure and function. In a validation phase, nearly 4,000 affected cases enriched for the presence of multiple adenomas were tested for this variant and compared with nearly 7,000 controls. In this exercise, 12 additional unrelated cases were found to have the L424V variant, with none of the controls having the variant. In the affected families, inheritance of multiple-adenoma risk appeared to be autosomal dominant.
A similar approach, whole-genome testing for shared variants, with further “filtering” by linkage analysis identified a variant in the POLD1 gene (p.Ser478Asn; c.1433G>A). This S478N variant was identified in two of the originally evaluated families, suggesting evidence of common ancestry. The validation exercise showed one patient with polyps with the variant but no controls with the variant. Somatic mutation patterns were similar to the POLE variant. Several cases of early-onset endometrial cancer were seen. The mechanism underlying adenoma and carcinoma formation resulting from the POLE L424V variant appeared to be a decrease in the fidelity of replication-associated polymerase proofreading. This in turn appeared to lead to variants related to base substitution. A subsequent study confirmed that POLE pathogenic variants are a rare cause of oligopolyposis and early-onset CRC.
All individuals in this study were negative for germline pathogenic variants in APC, MUTYH, and the MMR genes. The POLE variant L424V was found in 3 of 485 index cases with colorectal polyposis and early-onset CRC. Tumors showed microsatellite instability (MSI) and were deficient of one or more MMR proteins in two of three index cases. Somatic mutations in MMR genes, possibly the result of hypermutability secondary to POLE deficiency, were detected in these two cases. The Cancer Genome Atlas Network performed extensive sequencing analysis of 276 CRCs, and found that the presence of somatic mutations in the POLE gene was associated with a hypermutated phenotype with a substantially greater mutational burden than present in CRCs with MSI. Thus, polymerase variants appear to generate an ultra-hypermutated genotype in the tumor.
A study utilizing whole-exome sequencing in 51 individuals with multiple colonic adenomas from 48 families identified a homozygous germline nonsense pathogenic variant in seven affected individuals from three unrelated families in the base-excision repair gene NTHL1.
These individuals had CRC, multiple adenomas (8–50), none of which were either hyperplastic or serrated, and in three affected females, there was either endometrial cancer or endometrial complex hyperplasia. There were two other individuals who developed duodenal adenomas and duodenal cancer. All pedigrees were consistent with autosomal recessive inheritance. Upon examining three cancers and five adenomas from different affected individuals, none showed MSI. These neoplasms did show enrichment of cytosine to thymine transitions. Additional studies are needed to further define the phenotype. A subsequent study of 863 families with CRC and 1,600 families without CRC confirmed an association between biallelic NTHL1 pathogenic variants and inherited CRC risk.
Currently, there is no known increased risk of cancer for individuals harboring a single monoallelic pathogenic germline NTHL1 variant.
Hereditary mixed polyposis, characterized by histology that often includes adenomatous and hyperplastic polyps, has been associated with GREM1 pathogenic variants in a small number of Ashkenazi Jewish families. Polyp number in this syndrome is highly variable but is often in the spectrum consistent with oligopolyposis. (Refer to the Hereditary mixed polyposis syndrome [HMPS] section of this summary for more information.)
NTHL1, POLE, POLD1, and GREM1 pathogenic variant testing is being incorporated into the multigene (panel) tests for CRC susceptibility offered commercially along with APC and MUTYH so that a polyposis panel can be ordered up front for the patients with oligopolyposis. There are minimal data on the optimal surveillance approach for individuals found to have pathogenic germline variants in NTHL1 (biallelic carriers only), POLE, or POLD1, although it is presumed that the risk of CRC is comparable to what is seen in Lynch syndrome, and some guidelines are endorsing similarly early and frequent colonoscopic screening.
Oligopolyposis caused by other polyposis histologies can be distinguished from adenomatous polyposis on simple endoscopic and histologic grounds. For example, individuals with juvenile polyposis syndrome (JPS), PJS, or PTEN hamartoma tumor syndrome (Cowden syndrome) can all manifest oligopolyposis, often inclusive of hamartomatous polyps, as well as other more common polyp histologies (e.g., adenomas).
Serrated polyposis can likewise present in highly variable fashion. The World Health Organization (WHO) criteria for serrated polyposis (≥5 serrated polyps proximal to sigmoid with 2 polyps ≥1 cm, or any number of polyps proximal to sigmoid if there is a relative with serrated polyposis, or ≥20 serrated polyps anywhere in the colon) have never been validated. Rarely, families with serrated polyposis can be identified to harbor pathogenic germline RNF43 variants, but most cases of serrated polyposis cannot be linked to an underlying genetic basis.
Consequently, such patients are increasingly being referred for genetic counseling and for consideration of genetic testing. Occasional cases of MUTYH biallelic pathogenic variants have been found in patients with at least some features of serrated polyposis and serrated polyps can be seen in Lynch syndrome. However, germline evaluation of individuals with serrated polyposis is typically unrevealing.
Two very small case series have described oligopolyposis with varying polyp histologies (e.g., adenomas, serrated, inflammatory, and hamartomatous polyps) in individuals previously treated with chemotherapy and radiation therapy for a prior childhood malignancy.
This phenomenon, termed therapy-associated polyposis (TAP), may be an acquired, nonfamilial phenotype caused by prior antineoplastic therapy, and is on the differential diagnosis when nonfamilial oligopolyposis is identified in individuals previously treated with chemotherapy and/or radiation. Another recent study identified oligopolyposis fulfilling WHO criteria for serrated polyposis syndrome (SPS) in 6% of a cohort of 101 Hodgkin lymphoma survivors treated with prior chemotherapy and/or radiation therapy, suggesting that Hodgkin lymphoma survivors may be a particularly important population in whom TAP can manifest.
Lynch syndrome is the most common inherited CRC syndrome and accounts for approximately 3% of all newly diagnosed cases of CRC. It is an autosomal dominant condition caused by pathogenic variants in the MMR genes MLH1 (mutL homolog 1), MSH2 (mutS homolog 2), MSH6 (mutS homolog 6), and PMS2 (postmeiotic segregation 2), as well as the gene EPCAM (epithelial cellular adhesion molecule, formerly known as TACSTD1), in which deletions in EPCAM cause epigenetic silencing of MSH2. Lynch syndrome is also associated with a predisposition for developing several extracolonic manifestations, including sebaceous adenomas and cancers of the endometrium and ovaries, stomach, small intestine, transitional cell carcinoma of the ureters and renal pelvis, hepatobiliary system, pancreas, and brain. Lynch syndrome–associated cancers exhibit MSI; therefore, tumor testing is a key component in the diagnosis of Lynch syndrome, in addition to family history. Universal tumor testing of all CRCs is now recommended as a strategy to screen for Lynch syndrome and identify those individuals who may subsequently benefit from germline genetic testing. Intensive cancer screening and surveillance strategies, including frequent colonoscopy, along with risk-reducing surgeries, are mainstays in patients with Lynch syndrome.
Between 1913 and 1993, numerous case reports of families with apparent increases in CRC were reported. As series of such reports accumulated, certain characteristic clinical features emerged: early age at onset of CRC; high risk of synchronous (and metachronous) colorectal tumors; preferential involvement of the right colon; improved clinical outcome; and a range of associated extracolonic sites including the endometrium, ovaries, other sites in the GI tract, uroepithelium, brain, and skin (sebaceous tumors). Terms such as cancer family syndrome, and hereditary nonpolyposis colorectal cancer (HNPCC) were used to describe this entity.
The term Lynch syndrome replaced HNPCC and is applied to cases in which the genetic basis can be confidently linked to a germline pathogenic variant in a DNA MMR gene. Moreover, HNPCC is misleading as many patients have polyps and many have tumors other than CRC.
With the increased recognition of families that were considered to have a genetic predisposition to the development of CRC, research for a causative etiology led to the development of the Amsterdam criteria in 1990.
The Amsterdam criteria were originally used for the identification of high-risk families and included fulfillment of all of the following: three or more cases of CRC over two or more generations, with at least one diagnosed before age 50 years, and no evidence of FAP.
In 1987, a chromosomal deletion of a small segment of 5q led to the detection of a genetic linkage between FAP and this genomic region,
from which the APC gene was eventually cloned in 1991.
This led to searches for similar linkage in families suspected of having Lynch syndrome who had multiple cases of CRC inherited in an autosomal dominant fashion and young onset of cancer development. The APC gene was one of several genes (along with DCC and MCC) evaluated in families that fulfilled Amsterdam criteria, but no linkage was found among the Lynch kindreds. In 1993, an extended genome-wide search resulted in the recognition of a candidate chromosome 2 susceptibility locus in large families. Once MSH2, the first Lynch syndrome–associated gene, was sequenced, it was evident from the somatic mutation patterns in the CRC tumors that the MMR family of genes was likely involved. Additional MMR genes were subsequently linked to Lynch syndrome, including MLH1, MSH6, and PMS2. Lynch syndrome now refers to the genetic disorder caused by a germline variant in one of these DNA MMR genes, distinguishing it from other familial clusters of CRC.
In 2009, a germline deletion in the EPCAM gene was identified as another cause of MSH2 inactivation in the absence of a germline pathogenic variant in MSH2. The variant in EPCAM led to hypermethylation of the MSH2 promoter. Thus, EPCAM, which is not a DNA MMR gene, is also implicated in Lynch syndrome and is now routinely tested in at-risk patients along with the DNA MMR genes listed above.
Families with a preponderance of CRC and a possible genetic predisposition were initially categorized as having Lynch syndrome based on family history criteria, as well as personal history of young-onset CRC. With the advent of molecular tumor diagnostic testing and the discovery of the germline alterations associated with Lynch syndrome, the clinical criteria have currently fallen out of favor due to their underperformance. However, their use, or the risk estimates provided by the Lynch syndrome prediction models, may be applicable among individuals without personal history of cancer but with a family history suggestive of Lynch syndrome, or for those individuals with CRC but without available tumor for molecular diagnostic testing. (Refer to the Universal tumor testing to screen for Lynch syndrome and the Clinical risk assessment models that predict the likelihood of an MMR gene pathogenic variant sections of this summary for more information.)
The first criteria for defining Lynch syndrome families were established by the International Collaborative Group meeting in Amsterdam in 1990 and are known as the Amsterdam criteria.
These research criteria were limited to diagnoses of familial CRC. In 1999, the Amsterdam criteria were revised to include some extracolonic cancers, predominantly endometrial cancer.
These criteria provide a general approach to identifying Lynch syndrome families, but they are not considered comprehensive; nearly half of families meeting the Amsterdam criteria do not have detectable pathogenic variants.
These criteria were subsequently used beyond research purposes to identify potential candidates for microsatellite and germline testing. However, the Amsterdam criteria failed to identify a substantial proportion of Lynch syndrome kindreds; families that fulfilled Amsterdam criteria I but did not have evidence of MSI and were without a pathogenic germline variant in a DNA MMR gene, were referred to as familial colorectal cancer type X (FCCX). (Refer to the FCCX section of this summary for more information.)
With the hallmark feature of MSI associated with Lynch syndrome tumors, and the limitations of the Amsterdam criteria related to low sensitivity, the Bethesda guidelines were introduced in 1997. The Bethesda guidelines are a combination of clinical, histopathologic, and family cancer history features that identify cases of CRC that warrant MSI tumor screening. The Bethesda guidelines (with a subsequent revision in 2004) were formulated to target patients in whom evaluation of CRC tumors for MMR deficiency should be considered, and to improve the sensitivity of clinical criteria used to identify individuals who are candidates for mutational DNA analysis.
(Refer to the Genetic and molecular testing for Lynch syndrome section of this summary for more information about testing for MSI and IHC.)
*One criterion must be met for the tumor to be considered for MSI testing.
**Lynch syndrome–associated tumors include colorectal, endometrial, stomach, ovarian, pancreatic, ureter and renal pelvis, biliary tract, and brain tumors; sebaceous gland adenomas and keratoacanthomas in Muir-Torre syndrome; and carcinoma of the small bowel.
Although the Bethesda guidelines were able to identify a higher proportion of Lynch syndrome carriers than the Amsterdam criteria, they still missed approximately 30% of Lynch syndrome families.
Furthermore, the Bethesda guidelines were not consistently used in clinical practice to identify the subset of individuals with CRC who should have MSI tumor testing; the guidelines were deemed cumbersome and difficult to remember by health care providers and the opportunity to refer for genetic evaluation was missed.
With the advent of alternative approaches, including universal testing of all newly diagnosed cases of CRC for MSI (regardless of age at diagnosis or family history of cancer), clinical criteria for Lynch syndrome have been rendered obsolete. While the Bethesda guidelines were intended for individuals with cancer, their performance in individuals unaffected by cancer may still be of use. Given the limited modalities available to assess unaffected individuals for Lynch syndrome, family history and the use of clinical criteria may be appropriate in identifying those who warrant further genetic evaluation and testing.
Because health care providers ineffectively use clinical criteria to select individuals with CRC for genetic referral and evaluation for Lynch syndrome, computer-based clinical prediction models were developed and introduced in 2006 as alternative modalities to provide systematic genetic risk assessment for Lynch syndrome. The risk models include the PREMM (PREdiction Model for gene Mutations) models, MMRpredict, and MMRpro.
Three models (PREMM[1,2,6], MMRpredict, and MMRpro) quantify an individual’s probability of carrying an MMR gene variant in MLH1, MSH2, and MSH6. The PREMM(1,2,6) model was subsequently extended to include prediction of pathogenic PMS2 and EPCAM variants and is the only model to provide prediction of all five genes associated with Lynch syndrome (PREMM5).
While the models were all created for the same purpose, they differ in the way they were developed and the variables used to predict risk. In addition, the populations in which they were validated reveal each model’s specific characteristics that may impact accuracy.
Deciding on which model to use in the risk assessment process depends on both the clinical setting in which it is applied and the patient population that is being evaluated. MMRpro’s predictions account for family size and unaffected relatives, the possibility of including molecular tumor data in the risk analysis, and the option of predicting pathogenic variant carrier status following germline testing. The major limitation in the widespread use of MMRpro in routine practice is the need to input data from the entire pedigree (including individuals without cancer), which is relatively time-consuming. Its best use is likely to be as a genetic counseling tool in a specialized high-risk clinic or research setting, as its accessibility is also limited. PREMM’s major advantages include that it is easy to use, available as an online tool, and has been extensively validated, including in a self-administered setting in a GI clinic.
It includes risk prediction based on personal and family cancer history up to second-degree relatives for a broad spectrum of extracolonic cancers. However, the model does not take into account family size and may overestimate the likelihood of a pathogenic variant in a pedigree that includes multiple elderly family members who are unaffected by CRC or endometrial cancer. Given the ease with which one can use the PREMM model (it has been deemed less time-consuming than MMRpro in validation studies),
it may be used by diverse health care providers whose primary aim is to identify patients who should be referred for genetic evaluation, and is likely to be most useful in the pretesting decision-making process. Lastly, MMRpredict’s use may be limited overall because of its less accurate risk estimates
when used to evaluate families with Lynch syndrome–associated cancers and older individuals affected by CRC; the model was developed using data from young-onset CRC cases (patients diagnosed at age <55 y) and did not include extracolonic malignancies. Furthermore, the model does not incorporate tumor testing results or provide post-hoc risk estimates based on gene sequencing results.
Overall, there is ample evidence that each of the models has superior performance characteristics of sensitivity, specificity, and positive and negative predictive values that support their use when compared with the existing clinical guidelines for diagnosis and evaluation of Lynch syndrome. Because of the diverse clinical settings in which a health care provider has the opportunity to assess an individual for Lynch syndrome, prediction models offer a potentially feasible and useful strategy to systematically identify at-risk individuals, whether or not they are affected with CRC.
In conclusion, the presence of tumor MSI in CRCs, along with a compelling personal and family history of cancer, warrants germline genetic testing for Lynch syndrome, and most clinical practice guidelines provide for such an approach. These guidelines combine genetic counseling and testing strategies with clinical screening and treatment measures. Providers and patients alike can use these guidelines to better understand available options and key decisions. (Refer to Table 13 for more information about practice guidelines for diagnosis and colon surveillance in Lynch syndrome.)
The genetics of both the tumor and the germline have an important role in the development and diagnosis of Lynch syndrome. Tumor DNA in Lynch syndrome–associated tumors exhibits characteristic MSI, and in these cases, there is typically loss of IHC expression for one or more of the proteins associated with the MMR genes. Molecular testing with MSI and/or IHC has been adopted as a universal screen for diagnosis of Lynch syndrome in newly diagnosed patients with CRC and endometrial cancer. IHC testing results can potentially direct gene-specific germline testing. Many genetic testing laboratories offer multigene (panel) tests that simultaneously test for pathogenic variants in all of the Lynch syndrome–associated genes (and often additional genes associated with inherited cancer susceptibility).
The presence of MSI in colorectal tumor specimens is a hallmark feature of Lynch syndrome and can be cause for suspicion of a germline pathogenic MMR gene variant. Microsatellites are short, repetitive sequences of DNA (mononucleotides, dinucleotides, trinucleotides, or tetranucleotides) located throughout the genome, primarily in intronic or intergenic sequences.
The term MSI is used when colorectal, endometrial, or metastatic tumor DNA
shows insertions or deletions in microsatellite regions when compared with normal tissue. MSI indicates probable defects in MMR genes, which may be due to somatic mutations, germline variants, or epigenetic alterations.
In most instances, MSI is associated with absence of protein expression of one or more of the MMR proteins (MSH2, MLH1, MSH6, and PMS2). However, loss of protein expression may not be seen in all tumors with MSI and not all tumors with loss of protein expression on IHC will be microsatellite unstable.
Certain histopathologic features are strongly suggestive of MSI phenotype, including the presence of tumor-infiltrating lymphocytes (refer to Figure 4), Crohn-like reaction, mucinous histology, absence of dirty necrosis, and histologic heterogeneity.
Initial designation of a colorectal adenocarcinoma as microsatellite unstable was based on the detection of a specified percentage of unstable loci from a panel of three dinucleotide and two mononucleotide repeats that were selected at a National Institutes of Health (NIH) Consensus Conference and referred to as the Bethesda panel. If more than 30% of a tumor's markers were unstable, it was scored as MSI-H; if at least one, but fewer than 30% of markers were unstable, the tumor was designated MSI-low (MSI-L). If no loci were unstable, the tumor was designated microsatellite stable (MSS). Most tumors arising in the setting of Lynch syndrome will be MSI-H.
The clinical relevance of MSI-L tumors remains controversial; the probability is very small that these tumors are associated with a germline pathogenic variant in an MMR gene.
The original Bethesda panel has been replaced by a pentaplex panel of five mononucleotide repeats,
which has improved the detection of MSI-H tumors.
(Refer to the Prognostic and therapeutic implications of MSI section of this summary for more information about the treatment implications of MSI testing.)
(Refer to the Universal tumor testing to screen for Lynch syndrome section of this summary for information about the utilization of MSI status in the diagnostic workup of a patient with suspected Lynch syndrome.)
IHC methods are cheaper, easier to understand, and more widely available as a surrogate for MSI and, for these reasons, have replaced polymerase chain reaction (PCR)–based MSI testing in most institutions. IHC is performed in the colorectal or endometrial tumor (or metastatic sites)
for protein expression using monoclonal antibodies for the MLH1, MSH2, MSH6, and PMS2 proteins. Isolated loss of expression of any one of these proteins may suggest which specific MMR gene is altered in a particular patient.
However, certain proteins can form heterodimers (or have other binding partners) and yield loss of two proteins expressed on IHC.
MSI can lead to nucleotide-pairing slippage (looping) in which single nucleotide mispairs are introduced. Heterodimers of MMR proteins are formed to identify the errors and bind the DNA at these sites.
For example, MSH2 protein complexes with MSH6 protein to form MutSα, which has the main ability to repair single base pair mismatches and single base pair loop-out lesions that can occur during the replication of a mononucleotide repeat sequence. In the absence of MSH6 protein, the MSH2 protein will dimerize with the MSH3 protein forming the MutSβ complex, which has the ability to trigger repair of larger loop-out DNA mismatches, but also has some overlapping activity to repair lesions usually repaired by MutSα.
As a result, when the germline pathogenic variant is in the MSH2 gene, the tumor IHC may not express both MSH2 and MSH6, as the latter protein requires binding to MSH2 for stability. In this case, if no pathogenic variant is found in either gene, germline pathogenic variant testing for EPCAM should be considered if it was not already included. Approximately 20% of patients with absence of MSH2 and MSH6 protein expression by IHC and no MSH2 or MSH6 pathogenic variant identified will have germline deletions in EPCAM.
The latter mechanism accounts for approximately 5% of all Lynch syndrome cases.
A deletion in one allele of exon 9 of the EPCAM (TACSTD1) gene, which is immediately upstream of the start site of MSH2 and in the same orientation, can lead to transcriptional read-through and methylation of the MSH2 promoter, and subsequent silencing of MSH2 in any tissue that expresses EPCAM. The presence of EPCAM pathogenic variants showing similar methylation-mediated MSH2 loss has been reported in numerous families.
On the strength of these observations, germline EPCAM testing is performed in patients with loss of MSH2 protein expression on IHC testing of their CRCs but who lack a detectable MSH2 germline pathogenic variant and is included with MSH2 testing in all colon cancer gene panels.
In patients with no variants in any of these genes, tumor sequencing may reveal double somatic MSH2 mutations. (Refer to the and Lynch-like or HNPCC-like syndrome sections of this summary for more information.)
Similarly, the loss of MLH1 (either by germline pathogenic variant or hypermethylation of the MLH1 promoter) results in the absence of expression of both MLH1 and PMS2 proteins in the tumor. The most common abnormal IHC pattern for DNA MMR proteins in colorectal adenocarcinomas is loss of expression of MLH1 and PMS2. PMS2 and MLH1 function as a stable heterodimer known as MutLα. MutLα binds to MutSβ and guides excision repair of the newly synthesized DNA strand.
A functional defect in MLH1 results in degradation of both MLH1 and PMS2, while a defect in PMS2 negatively affects only PMS2 expression. Thus, a loss of MLH1 and PMS2 indicates an alteration in MLH1 (promoter hypermethylation or germline variant), while loss of PMS2 expression indicates a germline PMS2 variant. However, among 88 individuals with PMS2-deficient CRC, PMS2 germline pathogenic variant testing followed by MLH1 germline pathogenic variant testing revealed pathogenic PMS2 variants in 49 individuals (74%) and MLH1 pathogenic variants in 8 individuals (12%).
Eighty-three percent of the alterations in MLH1 were missense variants, but two relatives carried identical MLH1 variants, and one individual, who developed two tumors with retained MLH1 expression, carried an intronic variant that led to skipping of exon 8.
Therefore, in CRCs with solitary loss of PMS2 expression, an MLH1 germline pathogenic variant should be sought if no PMS2 germline variant is found. Tumors with MSI and loss of MSH2 and MSH6 protein expression are generally indicative of an underlying MSH2 germline variant (inferred MSH2 pathogenic variant). Unlike the case with MLH1, MSI with MSH2 loss is rarely associated with somatic hypermethylation of the promoter.
Unlike MLH1 and MSH2 (which both dimerize with other proteins or have other binding partners), germline pathogenic variants in MSH6 and PMS2 result in the isolated loss of those specific proteins by IHC. However, tumors from MSH6 pathogenic variant carriers may not display the MSI phenotype at a frequency as high as MLH1 and MSH2 carriers (despite an inactive DNA MMR system), as there are pathogenic missense variants that do not completely abrogate protein expression yielding false negative results by IHC testing.
In a study that reported tumor testing results among MMR germline carriers enrolled through the Colon Cancer Family Registry, 7 of 24 carriers (28%) with MSH6 pathogenic variants had tumors that displayed normal protein expression on IHC staining. IHC tumor testing was more informative for MLH1 and MSH2 pathogenic variant carriers in which 93% of MLH1 carriers had correlating loss of MLH1 protein expression and 96% of MSH2 carriers had loss of MSH2 protein expression.
In some cases, tumors manifest MSI and/or IHC shows loss of DNA MMR protein expression, but no germline pathogenic variant is identified. This condition is known as Lynch-like (or HNPCC-like) syndrome and the tumor phenotype is predominantly due to biallelic somatic inactivation of DNA MMR genes and not a pathogenic germline alteration. (Refer to the Lynch syndrome–related syndromes section of this summary for more information.)
Loss of Protein Expression | Germline MMR Defect Predicted by IHC Protein Expression Loss | ||||
---|---|---|---|---|---|
MLH1 | MSH2 | MSH6 | PMS2 | EPCAM | |
MLH1/PMS2 | X | ||||
MSH2/MSH6 | X | X | |||
MSH6 | X | ||||
PMS2 | X | X | |||
MLH1 | X | ||||
MSH2 | X | ||||
IHC = immunohistochemistry; MMR = mismatch repair. |
It is important to recognize that hypermethylation of the MLH1 promoter, a somatic event confined to the tumor, can lead to abnormal protein expression of MLH1 on IHC. Approximately 10% to 15% of sporadic CRC cases have a microsatellite unstable tumor phenotype due to MLH1 hypermethylation and are not heritable. These sporadic MSI colon cancers
have a generalized excess of DNA methylation referred to as CIMP.
(Refer to the CIMP and the serrated polyposis pathway section in the Introduction section of this summary for more information.) Because loss of MLH1 protein expression on IHC occurs in both Lynch syndrome and sporadic tumors, its specificity for predicting germline MMR gene variants is lower than for the other MMR proteins, and additional molecular testing is often necessary to clarify the etiology of MLH1 absence.
BRAF pathogenic variants have been detected in 68% of CRC tumors with MLH1 promoter hypermethylation and very rarely, if ever, in CRC from patients with Lynch syndrome.
This suggests that detection of somatic BRAF V600E mutation detection in CRC may be useful in excluding individuals from germline variant testing. As a result, BRAF V600 testing and/or MLH1 hypermethylation assays are increasingly utilized in universal Lynch syndrome–testing algorithms in an attempt to distinguish between an absence of MLH1 protein expression caused by hypermethylation and germline MLH1 pathogenic variants. Making such a distinction is also a more cost-effective approach in excluding individuals from germline testing.
Rarely, patients with MMR gene variants carry such variants in both parental alleles. When two variant alleles are identified, whether homozygous or compound heterozygous, this is termed biallelic mismatch repair deficiency (BMMRD) or constitutional mismatch repair deficiency (CMMRD). The likelihood of BMMRD involving homozygous MMR gene pathogenic variants will inevitably be higher among consanguineous unions. The incidence of consanguinity may be higher in rural and otherwise geographically and/or culturally isolated populations.
Tumor studies yield characteristic abnormalities. In a series of 28 patients with BMMRD,
17 brain tumors showed loss of staining for the MMR protein in the normal stromal cells in addition to neoplastic cells, showing a contradistinction from tumors in patients with Lynch syndrome in which normal staining is retained in nontumor cells. In contrast to this characteristic feature seen with IHC, PCR-based MSI analysis was not reliable, as 20 of 28 tumors were MSS. Of the tumors that were MSI-H, essentially all were colon cancers.
The PMS2 gene is markedly overrepresented in cases of BMMRD. It has been suggested that the presence of homozygosity of variants in the other MMR genes is a prenatally lethal state, while the otherwise milder expression of PMS2 is consistent with survival when present in both parental alleles.
(Refer to the BMMRD section in the Prevalence, clinical manifestations, and cancer risks associated with Lynch syndrome section for more information about the clinical phenotype of BMMRD.)
Clinical Phenotype | Pathogenic Germline Variant in DNA MMR | Somatic Inactivation of DNA MMR | Tumor Phenotype |
---|---|---|---|
Lynch syndrome | Present in one allele | Present in one allele | MSI |
Sporadic CRC with hypermethylation of MLH1 promoter | Absent | +BRAF | MSI |
BMMRD | Present in two alleles | Absent | MSI (tumor and normal tissue) |
Lynch-like | Absent | Present in two alleles | MSI |
FCCX | Absent | Absent | MSS |
BMMRD = biallelic mismatch repair deficiency; FCCX = familial colorectal cancer type X; MMR = mismatch repair; MSI = microsatellite instability; MSS = microsatellite stable. | |||
aAdapted from Carethers et al. |
While somatic hypermethylation of the MLH1 promoter is acquired and not uncommon, examples of MLH1 promoter hypermethylation have been described in the germline and are generally not associated with a stable Mendelian inheritance. This constitutional methylation of MMR genes occurs most often in MLH1 and, to a lesser extent, MSH2 and is termed constitutional .
A constitutional epimutation (also referred to as a primary epimutation) is an acquired alteration in normal tissue that silences an active gene or activates an inactive gene.
Such epimutations occur most often in maternal alleles. In some cases all somatic cells appear involved, while in others there is evidence of mosaicism. Tumors in patients with primary epimutations are generally indistinguishable from those otherwise typical of Lynch syndrome germline variant carriers, including age at onset, tumor spectrum, and presence of abnormal MSI and IHC. Since these are not inherited in a Mendelian fashion, antecedent family history of tumors is minimal, and risk to offspring somewhat unpredictable. Epimutations present in a de novo case seem to typically be "erased" in the process of gametogenesis and to not be passed to the next generation. Very rare cases of inherited MLH1 epimutations have been reported.
Interpreting molecular alterations in tumors and distinguishing the likely primary epimutation cases from those of sporadic MSI poses significant challenges. Most instances of absence of MLH1 expression are caused by the sporadic hypermethylation of the MLH1 promoter. Rare instances of a de novo constitutional epimutation in MLH1
or an inherited germline MLH1 methylation
add some complexity to the interpretation of MSI associated with absence of MLH1 expression. Akin to sporadic MSI, primary epimutation tumors show methylation of the MLH1 promotor and may show BRAF variants as well. As noted above, family history of cancer in such cases tends to be minimal or absent, as in true sporadic MSI. Distinguishing such cases from sporadic cases may call for assaying normal tissue (e.g., blood or normal colon mucosa) for evidence of MLH1 methylation, which will be absent from true sporadic cases and absent from carriers of conventional Lynch syndrome MMR pathogenic variants.
Such MLH1-predominant primary epimutations are to be distinguished from secondary epimutations such as those occurring when MSH2 is methylated as a consequence of inherited variants in the upstream EPCAM gene. (Refer to the section of this summary for more information.)
While many molecular pathology laboratories can assess both MSI and IHC, an approach that uses IHC testing as the initial screen for defective MMR activity has been favored because it is less labor intensive and more cost-effective.
Part of this rationale is that the information provided by IHC may target germline genetic testing toward one specific MMR gene (with the exception of loss of MLH1 expression) as opposed to a comprehensive testing strategy of all Lynch syndrome–related MMR genes that would be directed by the use of MSI alone.
While MSI testing was originally favored in the oncologic evaluation of individuals with CRC for its prognostic and therapeutic implications, screening for Lynch syndrome can be more effectively directed by IHC testing.
Use of MSI and/or IHC testing in all newly diagnosed cases of CRC, regardless of the age at diagnosis or family history of cancer, increases the sensitivity of the initial screen for Lynch syndrome, especially for carriers of MSH6 and PMS2 pathogenic variants. This approach is more sensitive than existing clinical criteria, as many individuals with Lynch syndrome are diagnosed at older ages (>50 y) and have less striking family histories of CRC than previously appreciated. This universal testing of colorectal (and endometrial) tumors using either MSI or IHC testing has been recommended by many professional organizations and is being widely adopted.
Genetic risk assessment and MMR gene variant testing in individuals with newly diagnosed CRC can lead to improved outcomes for the patient and at-risk family members. Dating back to 2009, the Evaluation of Genomic Applications in Practice and Prevention (EGAPP), a project developed by the Office of Public Health Genomics at the Centers for Disease Control and Prevention (CDC), reported that there was sufficient evidence to recommend offering tumor screening for Lynch syndrome to individuals with newly diagnosed CRC to reduce morbidity and mortality in relatives.
At that time, there was insufficient evidence to recommend a specific testing strategy between MSI and IHC.
Several studies have demonstrated the feasibility of universal screening for Lynch syndrome. Initial experience from one institution found that among 1,566 patients screened using MSI and IHC, 44 patients (2.8%) had Lynch syndrome. For each proband, an average of three additional family members were subsequently diagnosed with Lynch syndrome.
A subsequent pooled analysis of 10,206 incident CRC patients tested with MSI/IHC as part of four large studies revealed a pathogenic variant detection rate of 3.1%.
This study compared four strategies for tumor testing for the diagnosis of Lynch syndrome: (1) testing all individuals meeting at least one criterion of the Bethesda guidelines; (2) testing all individuals meeting Jerusalem recommendations;
(3) testing all individuals with CRC aged 70 years or younger, or older than 70 and meeting at least one criterion of the Bethesda guidelines; and (4) universal testing of all individuals with CRC.
Tumor testing with MSI involved panels individualized at each institution and IHC involved testing all four of the DNA MMR genes involved with Lynch syndrome, across all institutions. The strategy of tumor testing in all individuals diagnosed with CRC at age 70 years or younger and testing individuals over age 70 who met one of the revised Bethesda guidelines yielded a sensitivity of 95.1%, a specificity of 95.5%, and a diagnostic yield of 2.1%. This strategy missed 4.9% of Lynch syndrome cases, but 34.8% fewer cases required IHC/MSI testing, and 28.6% fewer cases underwent germline testing than in the universal approach.
The consideration to further stratify the recommendation for molecular tumor testing by age (i.e., 70 y) warrants attention as it influences the cost-effectiveness of universal screening strategy.
Loss of MLH1 and PMS2 due to somatic hypermethylation is not uncommon, and is more frequently detected with increasing age at CRC diagnosis.
Therefore, additional molecular tumor testing including BRAF and MLH1 hypermethylation testing is recommended in cases in which there is loss of MLH1 and PMS2 expression on IHC, thereby decreasing the number of individuals referred for unnecessary germline genetic testing. A testing strategy including MLH1 hypermethylation analyses in individuals aged 70 years or younger with CRC who had loss of MLH1 on IHC was shown to be cost-effective in a population-based study of 1,117 individuals.
Screening individuals with CRC for Lynch syndrome is most often performed in a stepwise fashion based on IHC tumor testing results that evaluate protein expression for the four MMR genes related to Lynch syndrome. One proposed strategy is summarized in Figure 6. This framework does not incorporate a germline testing approach that simultaneously evaluates multiple cancer susceptibility genes (multigene [panel] testing), which may be useful in select patient populations. (Refer to the Multigene [panel] testing section of this summary for more information.)
Clinicians are increasingly utilizing tumor sequencing to advance therapeutic decisions in a more personalized approach, particularly in patients with metastatic disease. The performance of next-generation tumor sequencing (NGS) of CRCs for the detection of Lynch syndrome was compared with existing screening protocols that include MSI testing and IHC staining (with BRAF p.V600E testing) in 419 CRC cases recruited in a multicenter, population-based study.
Twelve participants were identified as Lynch syndrome carriers by germline DNA testing and all were correctly identified by tumor sequencing, while MSI plus BRAF testing and IHC plus BRAF testing missed five and six Lynch syndrome cases, respectively. Tumor sequencing had a higher sensitivity than IHC plus BRAF testing (100% vs. 89.7%; P = .04) and MSI plus BRAF testing (100% vs. 91.4%; P = .07) while specificity was comparable across all strategies (95.3% for tumor sequencing, 94.6% for IHC plus BRAF, and 94.8% for MSI plus BRAF; P = not significant). In a validation cohort of 46 known Lynch syndrome pathogenic variant carriers with CRC, tumor sequencing yielded similar results and correctly identified 100% of carriers. In addition, the authors highlighted potential therapeutic implications by reporting on somatic alterations identified by tumor sequencing in 283 participants. This study suggested that tumor sequencing is a highly effective mode of identifying Lynch syndrome; however, the cost-effectiveness of this strategy remains to be determined.
A 2019 retrospective study using data from a large, community-based, integrated U.S. health care system compared the diagnostic performance of age-restricted screening strategies for Lynch syndrome by reflex MMR IHC of all CRCs versus a universal screening strategy without an upper age limit.
Lynch syndrome identification decreased substantially after age 70 years to age 75 years, with minimal incremental gain after age 80 years. The number of CRCs needed to be screened to identify one Lynch syndrome case was 20 among patients diagnosed with CRC at age 50 years or younger but increased to 208 for those with CRC at age 71 years to age 80 years, and 668 for those diagnosed after age 80 years.
Results are available from a Markov model that incorporated the risks of colorectal, endometrial, and ovarian cancers to estimate the effectiveness and cost-effectiveness of strategies to identify Lynch syndrome among persons aged 70 years or younger with newly diagnosed CRC .
The strategies incorporated in the model were based on clinical criteria, prediction algorithms, and tumor testing or up-front germline pathogenic variant testing followed by directed screening and risk-reducing surgery. IHC followed by BRAF pathogenic variant testing was the preferred strategy in this study. An incremental cost-effectiveness ratio of $36,200 per life-year gained resulted from this strategy. In this model, the number of relatives tested (3–4) per proband was a critical determinant of both effectiveness and cost-effectiveness. These results were similar to earlier analyses conducted by EGAPP which found that the most cost-effective approach was to test all tumors for absence of protein expression of MSH2, MLH1, MSH6, and PMS2 followed by targeted germline testing of MSH2, MLH1, or MSH6 offered depending on which protein was absent. If there was absence of MLH1, testing was offered for BRAF variant-negative tumors.
NCCN 2019 guidelines support universal screening of all CRCs with IHC and/or MSI, and/or a comprehensive tumor NGS panel or germline multigene (panel) testing.
Universal screening in all individuals irrespective of age was associated with a doubling of incremental cost per life-year saved compared with screening only those younger than 70 years.
The authors of this analysis conclude that screening individuals younger than 70 years appears reasonable, while screening all individuals regardless of age might also be acceptable, depending on willingness to pay.
However, it is important to note that the conclusions from this study were contingent upon the number of at-risk relatives who underwent germline testing (through a process known as ) based on the identification of a germline MMR gene variant in the index case of CRC in the family. In their model, to meet the accepted $50,000 cost-effective threshold, testing a minimum of three to four relatives was necessary.
This emphasizes the importance of provider-to-patient communication, family communication, and the need to ensure improved uptake of germline testing in Lynch syndrome families with a known causative gene. (Refer to the Psychosocial Issues in Hereditary Colon Cancer Syndromes section of this summary for more information about family communication and uptake of genetic testing in families with Lynch syndrome.)
Another study addressed the cost-effectiveness of testing for pathogenic variants in the Lynch syndrome–associated genes and evaluated 21 screening strategies, including clinical criteria, use of clinical Lynch syndrome prediction models, and molecular tumor testing.
The model included two steps: (1) measurement of the newly identified number of Lynch syndrome diagnoses; and (2) measurement of the life-years gained as a result of confirming Lynch syndrome in a healthy carrier. Among all of the strategies modeled, screening the proband with a predictive model such as PREMM(1,2,6) followed by IHC for MMR protein expression and germline genetic testing was the best approach, with an incremental cost-effectiveness ratio of $35,143 per life-year gained. Germline genetic testing on all probands was the most effective approach, but at a cost of $996,878 per life-year gained. The authors concluded that the initial step of Lynch syndrome screening should utilize a predictive model in the proband, and that both universal testing and general population screening strategies were not cost-effective screening strategies for Lynch syndrome.
Establishment of an upper age limit for universal tumor testing remains controversial. Some experts have endorsed testing only individuals with CRC who are younger than 70 years (reserving testing in individuals ≥70 y for only those meeting the revised Bethesda criteria; with this strategy, 5% of carriers would be missed).
However, others have advocated against an upper age limit for testing given the potential benefit to younger generations via cascade screening and the opportunity for increased surveillance and other prophylactic interventions in individuals found to carry a known familial pathogenic variant.
Another cost-effectiveness analysis was performed using data from 179 consecutive endometrial cancer patients diagnosed at or before age 70 years and screened with MMR IHC and reflex MLH1 promoter hypermethylation, among whom seven Lynch syndrome carriers (3.9%) were identified.
Only one of the seven Lynch syndrome probands was age 50 years or younger at endometrial cancer diagnosis. The authors calculated that screening women diagnosed with endometrial cancer at age 51 to 70 years resulted in an additional 29.3 life-years gained (on top of the 45.4 life-years gained by screening women diagnosed at age ≤50 y), and the incremental cost-effectiveness ratio for screening all diagnoses at age 70 years or younger versus diagnoses at age 50 years or younger was 5,252 euro per life-year gained. Universal tumor-based screening of all women age 70 years or younger was also cost-effective, compared with strategies using the Bethesda guidelines to guide MMR and MSI testing with an incremental cost-effectiveness ratio of 6,668 euro per life-year gained.
The cost-effectiveness of universal tumor testing in both CRC and endometrial cancer is largely driven by the assumption of cascade screening through which other at-risk family members will be identified, tested, and subsequently pursue their own cancer risk reduction.
The cost of germline genetic testing continues to decrease with advancements in DNA mutational analyses, including simultaneous testing of multiple germline variants associated with malignancy, through multigene (panel) tests. As a result, additional cost-effective analyses using more updated data related to germline testing will need to be conducted. Multigene (panel) testing may become a more favorable and cost-effective approach in the future.
While universal screening continues to be adopted nationally, there is significant variability in the uptake and approach to molecular testing. A 2011 survey of the National Society of Genetic Counselors revealed that more than 25% of respondents had some form of universal screening implemented at their center. Tumor screening methods varied; 34 (64.2%) of 53 centers started with IHC, 11 (20.8%) of 53 centers started with MSI testing, and 8 (15.1%) of 53 centers performed both tests on newly diagnosed colorectal tumors.
A 2012 survey suggested that some form of universal screening was being routinely performed at 71% of the National Cancer Institute (NCI) Comprehensive Cancer Centers, but utilization dropped to 15% among a random sample of community hospital cancer programs.
Because adherence to universal screening for Lynch syndrome may be poor (many patients are not referred for genetic evaluation and testing), a prospective quality improvement study utilizing the Six Sigma conceptual framework was conducted to improve the implementation of universal genetic screening among young patients with CRC.
The main aim of the study was to increase the proportion of tumor studies for deficient MMR among patients with early-onset CRC (aged 18–50 y). The intervention involved patient and provider education, in addition to visual cues provided at point of care. The study demonstrated an improvement of 21.5% in the rate of IHC testing in young adults with CRC over the 12-month postintervention period compared with the preintervention period.
Studies reporting uptake of genetic testing for Lynch syndrome have largely focused on individuals and families who were selected for potential risk of Lynch syndrome based on family history or clinical characteristics. While universal tumor screening is increasingly being adopted to identify newly diagnosed patients who may have a germline variant, few studies have examined the uptake of genetic testing after universal tumor testing. An important implication of universal screening for Lynch syndrome is that it does not result in automatic germline testing in appropriate individuals. In the clinical setting, more follow-up by health care teams to facilitate referral to genetic counseling for patients with abnormal tumor screening results may improve completion of genetic testing.
Higher levels of patient completion of genetic testing after abnormal tumor screening may be associated with having genetic counselors involved in this process to disclose screen-positive results, provide counseling after tumor testing, or facilitate referrals.
Subsequent genetic counseling requires coordination between the pathologist, the referring surgeon or oncologist, and a cancer genetics service. As an illustration, a population-based screening study found that only 54% of patients with an IHC-deficient tumor (that was BRAF pathogenic variant–negative) ultimately consented to and proceeded with germline MMR testing.
One institution found 21 pathogenic variants among 1,100 patients who underwent routine MSI and IHC testing after a diagnosis of CRC. This study found markedly increased uptake of genetic counseling and germline MMR gene testing when both the surgeon and a genetic counselor received a copy of abnormal MSI/IHC results, especially when the genetic counselor played an active role in patient follow-up.
In contrast to tumor testing, which is commonly performed without a patient's prior knowledge, germline genetic testing, such as germline testing for MMR pathogenic variants, generally includes genetic counseling and requires patient permission before it is performed. A cross-sectional survey of U.S. cancer programs (20 NCI–designated Comprehensive Cancer Centers and 49 community hospital cancer programs) found that, of those that performed MSI and/or IHC testing as part of standard pathologic evaluation at the time of colon cancer diagnosis in all or select cases, none required written informed consent before tumor testing.
Given the increased prevalence of endometrial cancer among carriers of MMR pathogenic variants, there is a growing consensus to screen patients with endometrial cancer for Lynch syndrome.
In a study that examined the feasibility and desirability of performing tumor screening of all endometrial cancers, regardless of age at diagnosis or family history of cancer, at least 2.3% (95% CI, 1.3%–4.0%) of newly diagnosed patients had Lynch syndrome.
Eight of thirteen cases diagnosed with Lynch syndrome were aged 50 years or older, eight did not meet published family history criteria for Lynch syndrome, and two would have been missed by MSI testing. Because of the increased prevalence of endometrial cancer and the results of this study, the authors support universal screening of endometrial cancers for Lynch syndrome. (Refer to the IHC section of this summary for more information about performing IHC for MMR protein expression.)
Another smaller study of 242 consecutive endometrial cases demonstrated a 4.5% (11/242) prevalence of MMR-deficient cases lacking somatic MLH1 promoter hypermethylation, including four cases (1.7%) with germline MMR mutations, four cases (1.7%) with two somatic MMR alterations on NGS, and two cases (0.8%) with otherwise unexplained MMR-deficiency.
Such findings demonstrate that universal MMR tumor screening of endometrial cancers will identify individuals with underlying Lynch syndrome and a spectrum of non-Lynch syndrome cases with various forms of MMR-deficiency.
Another study prospectively evaluated universal IHC-based screening of both CRC and endometrial cancer cases, irrespective of age at diagnosis.
In both the tertiary and community settings, 1,290 CRC and 484 endometrial cancer cases were screened between 2011 and 2013. The study additionally calculated PREMM(1,2,6) and PREMM5 scores for all patients in whom a germline pathogenic variant was detected. Abnormal staining was observed in 22% of endometrial cancers and 18.8% of CRCs. After excluding those cases felt to be sporadic because of the presence of BRAF and/or hypermethylation of MLH1, 10.8 % of patients with CRC and 6.6% of patients with endometrial cancer were referred for genetic counselling. Lynch syndrome was diagnosed in 24 individuals (1.4%), 66% of whom had CRC. The overall detection rate of Lynch syndrome was 1.7% in endometrial cancer cases and 1.2% in CRC cases. Among Amsterdam criteria, Bethesda guidelines, PREMM(1,2,6), and PREMM5, the best performing model was PREMM5, which would have detected 82% of cases identified by universal screening.
The cost-effectiveness of tumor testing of women diagnosed with endometrial cancer was examined in a model-based simulation study and included IHC testing in the following scenarios: (1) diagnosis before age 50 years; (2) diagnosis before age 60 years; (3) any age at diagnosis with the presence of an FDR with any Lynch syndrome–associated cancer; and (4) all cases irrespective of diagnosis age and family history. Women fulfilling Amsterdam II criteria or those diagnosed before age 50 years with at least one FDR with any Lynch syndrome–associated cancer were directly referred for genetic counseling and genetic testing without IHC testing. A strategy of IHC testing for MMR protein expression in all patients with endometrial cancer and an FDR with any Lynch syndrome–associated cancer was reported to be cost-effective in the detection of Lynch syndrome.
This strategy had an incremental cost ratio of $9,126 per life-year gained relative to the least-costly strategy, which was genetic testing on all women diagnosed with endometrial cancer before age 50 years with at least one FDR with a Lynch syndrome–related cancer. Life expectancy was highest with the most inclusive testing strategy of IHC testing of all women with endometrial cancer irrespective of age at diagnosis or family history, but had the least favorable incremental cost ratio of $648,494 per life-year gained. NCCN recommends tumor testing with IHC and/or MSI, a comprehensive tumor NGS panel, or germline multigene (panel) testing of all endometrial cancers.
Despite these recommendations, the uptake of universal screening in women newly diagnosed with endometrial cancer is unclear.
(Refer to the PDQ summary on Genetics of Breast and Gynecologic Cancers for more information about endometrial cancer as a component of Lynch syndrome.)
Use of MSI testing across all tumor types has become an important screening tool to select cases that may have a favorable response to immune checkpoint inhibitor therapy. These results may potentially be used to screen for Lynch syndrome in tumors other than CRC. A study evaluated MSI across a wide variety of malignancies and evaluated its use as a potential means to identify Lynch syndrome, regardless of tumor type.
In a study of more than 15,000 patients with more than 50 types of cancers evaluated in a single-center study, data on well-annotated tumor and matched normal DNA sequencing results with paired germline MMR gene testing, were used to determine MSI status. MSI was determined using a software tool that reports the percentage of unstable microsatellites as a score from paired tumor-normal genome sequencing data and allows for comprehensive investigation of MSI sites simultaneously. The approach used has been reported to be more sensitive across cancers not typically screened for MMR-deficiency (dMMR) than MSI testing of five mononucleotide microsatellite foci using PCR.
CRC and endometrial cancer comprised the majority of cancers with MSI-H in this study, but 38% (125 of 326) of MSI-H tumors and more than 90% of those with intermediate-level MSI were other cancer types. Germline testing confirmed a diagnosis of Lynch syndrome in 16.3% and 1.9% of tumors with MSI-H and intermediate-level MSI, respectively, in addition to 0.3% of cases that lacked MSI. Importantly, half of all Lynch syndrome carriers with MSI-H/intermediate tumors had primary cancers other than CRC or endometrial cancer, with many malignancies not associated with Lynch syndrome. Among those individuals with a noncanonical Lynch syndrome cancer, nearly half failed to meet clinical criteria for Lynch syndrome testing on the basis of their cancer diagnosis or family cancer history. Furthermore, intermediate-level MSI and MSS phenotypes were most often observed in cancers not classically related to Lynch syndrome and in individuals with germline PMS2 variants. This study supports other findings related to the variable phenotypic expression of Lynch syndrome on the basis of the altered MMR gene and its broad constellation of associated malignancies that make it difficult to be identified by clinical criteria alone. In addition, the investigators further analyzed a unique gene variant signature in every tumor and correlated results to the observed MSI phenotype and germline MMR status to provide some indirect data on whether a gene variant carrier’s cancer was caused by Lynch syndrome and MMR deficiency or possibly an incidental finding. This is pertinent in evaluating those cancers whose association with Lynch syndrome is unclear and debatable, such as breast and prostate cancer. The authors’ finding that none of the breast cancer patients with Lynch syndrome in this very large cohort had tumors with MSI lends support to the hypothesis that these individuals’ germline MMR gene variants may simply be incidental findings and not etiologic to their cancer diagnosis.
Genetic testing for germline pathogenic variants in MLH1, MSH2, MSH6, PMS2, and EPCAM can help formulate appropriate intervention strategies for the affected variant-positive individual and at-risk family members, many of whom may be unaffected by cancer.
If a pathogenic variant is identified in an affected person, then testing for that same pathogenic variant should be offered to all at-risk family members. At-risk relatives who test negative for the identified pathogenic variant in the family are not at increased risk of CRC or other Lynch syndrome–associated malignancies and can follow surveillance recommendations applicable to the general population. Family members who carry the familial pathogenic variant are referred to surveillance and management guidelines for Lynch syndrome. (Refer to the Management of Lynch syndrome section of this summary for more information.)
If no pathogenic variant is identified in the affected family member, then testing is considered negative for Lynch syndrome in that individual. With advances made in DNA sequencing technologies, it is unlikely that current gene testing is not sensitive enough to detect a pathogenic variant in the genes tested. Advances in testing, including the common use of NGS by most commercial testing laboratories have improved upon the detection of certain alterations such as large deletions or genomic rearrangements as well as the presence of a pseudogene PMSCL in PMS2.
Possible reasons why a pathogenic variant may not be detected include the following:
Failure to detect a pathogenic variant could mean that the family truly is not at genetic risk despite a clinical presentation that suggests a genetic basis (e.g., the patient may have double somatic mutations in an MMR gene). If no variant can be identified in an affected family member, testing should not be offered to at-risk members because results would be uninformative for the relatives. They would remain at increased risk of CRC by virtue of their family history and should continue with recommended intensive screening.
(Refer to the Management of Lynch syndrome section of this summary for more information.)
Germline mutation analysis of MLH1, MSH2 (including EPCAM), MSH6, and PMS2 may be considered in instances in which tumor tissue is not available from individuals to test for MSI and/or MMR protein IHC. This approach has become less expensive with the advent of multigene (panel) testing, which is now offered by several clinical laboratories at a cost that may be comparable to single-gene testing. The cost of multigene testing may also approach the cost of tumor screening and may prove to be a cost-effective approach in individuals affected by CRC. At present, multigene tests are not routinely recommended for universal screening for Lynch syndrome among all newly diagnosed CRC patients, but they may be very useful in select populations, such as those with early-onset CRC
or from familial, high-risk clinic-based populations. It is also important to note that pathogenic variants may be detected in other cancer-associated genes beyond Lynch syndrome. In a study of 1,112 individuals who met NCCN criteria for Lynch syndrome testing and who underwent multigene testing with a 25-gene panel, as expected, 114 individuals (9.0%) were found to have pathogenic variants in MMR genes; however, 71 individuals (5.6%) were found to have a pathogenic variant in non-Lynch syndrome cancer predisposition genes, such as BRCA1, BRCA2, APC, MUTYH (biallelic), and STK11. Lastly, multigene tests yield a high proportion of VUS. In the aforementioned study, a total of 479 patients (38%) had one or more VUS.
Individuals with early-onset CRC have been shown to have a high frequency and wide spectrum of germline pathogenic variants, indicating that panel testing in this population may be beneficial. In a study of 450 patients with early-onset CRC (mean age at diagnosis, 42.5 y) and a family history including at least one FDR with colon, endometrial, breast, ovarian, and/or pancreatic cancer, 75 germline pathogenic or likely pathogenic variants were identified in 72 patients (16%).
The spectrum of variants identified included Lynch syndrome and non-Lynch syndrome–associated genes, including several genes that have not traditionally been associated with CRC (e.g., BRCA1/BRCA2, ATM, CHEK2, PALB2, and CDKN2A). Given the high frequency and variety of hereditary cancer syndromes identified, the authors suggested that multigene testing in this population may be warranted. Similarly, another smaller single-institution analysis of 151 individuals with CRC identified pathogenic germline variants in 9.9% of individuals.
Multigene testing has also been examined in a larger study of 1,058 individuals with CRC who were unselected for age at diagnosis, personal or family history, or MSI/MMR test results.
Germline pathogenic variants in cancer susceptibility genes were identified in 105 individuals (9.9%). While 33 individuals (3.1%) carried pathogenic variants in Lynch syndrome genes, 74 (7.0%) had pathogenic variants in non-Lynch syndrome–associated genes, including APC, MUTYH, BRCA1/, PALB2, CDKN2A, TP53, and CHEK2. These data illustrate the breadth of variants that may be identified in unselected CRC patients; thus, use of a comprehensive multigene test may be warranted.
A 2017 study examined the frequency of pathogenic Lynch syndrome–associated gene variants in individuals undergoing multigene testing at a single commercial United States laboratory between 2012 and 2015, and reported on the characteristics of those carriers identified with Lynch syndrome.
The study reports on the largest cohort of individuals tested through multigene testing to date; data was reported on 34,980 individuals who had undergone various multigene panel tests that included the MMR and EPCAM genes, where the indication for testing was not limited to Lynch syndrome. A total of 618 pathogenic variants were identified in 612 individuals (1.7%) and analyses were conducted on 579 subjects (after exclusion of 33 individuals who had a Lynch syndrome–associated variant and a second MMR variant or other pathogenic alteration in another cancer predisposition gene). The majority of carriers were affected by cancer, including non-Lynch syndrome–associated malignancies, where breast cancer was most frequently reported (124/423, 23.5%). MSH6 variants were most prevalent (29.3%), followed by PMS2 (24.2%), MSH2 (23.7%), MLH1 (21.6%), and EPCAM (1.2%). This finding differs from previous data where MSH2 and MLH1 variants were more prevalent, as individuals were more often selected for Lynch syndrome–specific testing due to a personal and/or family history of CRC.
The study reports on genotype-phenotype correlations on 528 Lynch syndrome carriers, the majority of whom had CRC (186, 35.2%) and endometrial cancer (136, 25.8%), followed by breast cancer (124, 23.5%) and ovarian cancer (74, 14%).
One hundred forty-five carriers presented with breast or ovarian cancer as their sentinel tumor and did not carry a prior diagnosis of CRC or endometrial cancer prior to the time of multigene testing. When examining MMR gene variant distribution among tumor-specific subgroups, a higher frequency of MSH6 and PMS2 variants were detected in carriers with breast cancer only than MLH1 and MSH2, where the latter pathogenic variants were more frequent in subjects with CRC only. For patients with breast cancer only, the frequency of PMS2 gene variants was significantly higher than population estimates, which was not the case for MLH1, MSH2, or MSH6. A comparable retrospective study reported similar findings. Standardized incidence ratios (SIRs) of breast cancer were calculated by comparing observed breast cancer frequencies in a population of 423 women with pathogenic or likely pathogenic variants in MMR genes with those in the general population. The authors reported a statistically significant age-standardized risk of breast cancer for MSH6 carriers (SIR = 2.11; 95% CI, 1.56–2.86) and PMS2 carriers (SIR = 2.92; 95% CI, 2.17–3.92).
A critical limitation of both of these studies was the excess of breast cancer cases in the overall referral population as well as the known high background population prevalence of MSH6 and PMS2 germline pathogenic variants.
Clinical criteria for the identification of Lynch syndrome, including the Amsterdam criteria, revised Bethesda guidelines, or the PREMM(1,2,6) risk prediction model, would have failed to identify 27.3% of Lynch syndrome carriers in this study.
Given the increased prevalence of breast and ovarian cancers, 58.9% met the NCCN guidelines for BRCA1/BRCA2 testing and of these, 36.7% also met NCCN guidelines for Lynch syndrome testing. Lastly, there were limited data on tumor testing results, available only on 18.8% of pathogenic variant carriers, where results were often discordant with the altered gene, which was most often reported in MSH6 and PMS2 carriers. Results of this study support the use of multigene testing for Lynch syndrome and further study of the respective cancer risks, as current testing strategies limit identification of Lynch syndrome carriers and associated malignancies.
Lastly, germline MMR genes have been detected unexpectedly among individuals undergoing multigene testing for cancers not commonly associated with Lynch syndrome, such as breast and prostate cancer. As a result, the cancer spectrum associated with Lynch syndrome may be wider than previously appreciated. (Refer to the Breast cancer and Prostate cancer sections of this summary and the Genetics of Prostate Cancer summary for more information.)
(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.)
As genetic testing becomes routine rather than the exception, questions regarding the cost of testing are inevitable. Historically, a cost-effectiveness ratio of $50,000 per quality-adjusted life-year (QALY) has been utilized as the benchmark for good value for care.
Over time it has been suggested that this threshold is too low and that other thresholds such as $100,000 or $150,000 be utilized.
A 2015 study evaluated the cost-effectiveness of multigene testing for CRC and polyposis syndromes in patients referred to a cancer genetics clinic.
These authors developed a decision model to estimate the immediate and downstream costs for patients referred for evaluation and of CRC surveillance in family members identified as carriers of pathogenic variants. The costs were estimated on the basis of published models from the CDC and from an academic molecular genetics laboratory. They classified the syndromes on the basis of inheritance pattern and penetrance of CRC. Four custom panels were compared with the standard of care. The four panels tested for (1) Lynch syndrome–associated genes only (MLH1, MSH2, MSH6, PMS2, and EPCAM); (2) genes in panel 1 and additional genes associated with autosomal dominant inheritance and high CRC penetrance (APC, BMPR1A, SMAD4, and STK11); (3) genes in panels 1 and 2 and those associated with autosomal recessive inheritance with high CRC penetrance (MUTYH); or (4) all genes in the first three panels and those associated with autosomal dominant conditions with low penetrance (PTEN, TP53, CDH1, GALNT12, POLE, POLD1, GREM1, AKT1, and PIK3CA). The respective costs were as follows: panel 1, $144,235 per QALY; panel 2, $37,467 per QALY; panel 3, $36,500 per QALY; and panel 4, $77,300 per QALY when compared with panel 3. The authors concluded that the use of an NGS multigene test that includes highly penetrant CRC and polyposis syndromes and Lynch syndrome cancer genes was the approach most likely to provide clinically meaningful results in a cost-effective fashion.
The cost of germline genetic testing continues to decrease with advancements in technology since the time this model analysis was conducted; additional studies are needed to continue to assess the cost-effectiveness of this testing approach.
Lynch syndrome is an autosomal dominant syndrome characterized by an early age of onset of CRC, excess synchronous and metachronous colorectal neoplasms, right-sided predominance, and extracolonic tumors, notably endometrial cancer. Lynch syndrome is caused by pathogenic variants in the DNA MMR genes, namely MLH1 (mutL homolog 1) on chromosome 3p21;
MSH2 (mutS homolog 2) on chromosome 2p22-21;
MSH6 on chromosome 2p16;
and PMS2 (postmeiotic segregation 2) on chromosome 7p22.
The function of these genes is to maintain the fidelity of DNA during replication. Lynch syndrome is also associated with pathogenic variants of the EPCAM (epithelial cellular adhesion molecule, formerly known as TACSTD1) gene on chromosome 2p21, which causes epigenetic silencing of MSH2, located immediately downstream of this gene.
Lynch syndrome accounts for about 3% of all newly diagnosed cases of CRC.
In earlier studies, the average age at CRC diagnosis in carriers of Lynch syndrome pathogenic variants was reported as young as 44 to 52 years
versus 71 years in sporadic CRC.
In subsequent studies that corrected for ascertainment bias to determine cancer-related risk estimates and genotype-phenotype correlations, the average age at diagnosis of CRC was reported to be 61 years among carriers of Lynch syndrome–associated pathogenic variants.
Original reports related to overall and gene-specific prevalence estimates in Lynch syndrome relied heavily on retrospective data from familial cancer registries worldwide. Earlier risk estimates of CRC (and endometrial cancer) reported in Lynch syndrome were subject to ascertainment bias and overestimation, given that data were derived largely from familial cancer registries and cases were often ascertained based on young-onset CRC or an increased number of CRC cases among relatives. Correction of these cancer risk estimates has been made possible through modified segregation analyses, where statistical methodology provides more accurate estimates and adjusts for ascertainment bias. Conversely, risk estimates related to extracolonic malignancies, with the exception of endometrial cancer, may be prone to underestimation because many families may have underreported these cancers in relatives, and Lynch syndrome–related tumors may have occurred later in life.
In a large population-based study of 5,744 CRC cases who were recruited irrespective of family cancer history from the United States, Australia, and Canada, it was estimated that 1 in 279 individuals in the population carry an MMR pathogenic variant associated with Lynch syndrome.
In another population-based study of 450 individuals with CRC but limited to young onset with diagnoses occurring before age 50 years, germline pathogenic variants were identified in 72 of 450 individuals (16%), as detected by multigene (panel) testing for inherited cancer susceptibility genes. As expected, the majority of identified variants were in genes known to be associated with CRC, predominantly Lynch syndrome (37 of 72 patients, 51.4%). However, 13 of 72 patients (18.1%) had pathogenic variants in genes not traditionally associated with CRC, including but not limited to BRCA1/BRCA2, which accounted for 8% of the identified variants. Because of the high frequency and wide variety of pathogenic variants identified, the authors suggested consideration of multigene testing for all individuals with early-onset CRC.
The MLH1 and MSH2 genes were originally thought to account for most pathogenic variants of the MMR genes found in Lynch syndrome. However, the prevalence of MSH6 and PMS2 pathogenic variants has been increasing with improved DNA mutational analyses and universal tumor screening of all CRCs.
MSH6 and PMS2 variants may be more common in unselected cases of CRC (and endometrial cancer),
compared with MLH1 and MSH2 variants which were more commonly identified in individuals from high-risk CRC clinics.
A series of papers from the Prospective Lynch Syndrome Database (PLSD) describe the cancer outcomes in patients prospectively followed by (mainly European) registries. Among the key findings was a low risk of CRC in PMS2 carriers, especially among those below age 50 years, leading the authors to conclude that surveillance in PMS2 carriers could safely be scaled back. A later initiation of colonoscopy and perhaps at longer intervals, is gradually being adopted in light of these findings.
The relative risk of extracolonic cancers in PMS2 carriers was no greater or only slightly greater than population expectations, which led the authors to generally recommend against any extracolonic surveillance in PMS2 carriers.
These data in aggregate support a more liberalized approach for screening PMS2 carriers, although current clinical practice guidelines do not reflect this change.
The approach to screening individuals with PMS2 pathogenic variants, and to a lesser extent those with MSH6 pathogenic variants, are matters of ongoing clinical debate.
In early studies, the prevalence of MLH1 pathogenic variants in individuals with Lynch syndrome was reported to be between 41.7%
and 50%,
making MLH1 the most commonly altered MMR gene in Lynch syndrome families. It was not until a report on the population-based prevalence of Lynch syndrome that the MLH1 pathogenic variant was estimated to be 1 in 1,946, ranking third after PMS2 (1 in 714) and MSH6 (1 in 758), as estimated in a large international study of 5,744 CRC cases.
MLH1 pathogenic variants are associated with the entire spectrum of malignancies associated with Lynch syndrome.
The lifetime risk of any Lynch syndrome–associated cancer by age 70 years has been found to range between 59% and 65% in MLH1 pathogenic variant carriers.
The highest risk among carriers of pathogenic MLH1 variants is for CRC, which is estimated to be between 41% and 68%,
and the mean age at diagnosis of CRC was 42.8 years (range, 16–81 y) in one study that included 137 affected individuals.
In a more recent prospective study using pooled European registry data of 944 MLH1 carriers without cancer, the cumulative CRC incidence was 46% at age 70 years, despite colonoscopic surveillance (albeit at various intervals).
The prevalence of MSH2 pathogenic variants in individuals or families with Lynch syndrome has varied across studies. MSH2 pathogenic variants were reported in 38% to 54% of Lynch syndrome families in studies including large cancer registries and among cohorts of early-onset CRC (younger than age 55 y).
The reported prevalence of MSH2 pathogenic variants was 32.8% in 2012 in the database of the International Society for Gastrointestinal Hereditary Tumors (InSiGHT), a large professional organization devoted to the collaborative study of familial GI cancer,
with families readily ascertained based on the presence of extracolonic cancers in MSH2-associated Lynch syndrome. However, the prevalence of MSH2 pathogenic variants was estimated to be 1 in 2,841 in a population-based cohort of 5,744 CRC cases recruited from the United States, Australia, and Canada;
MSH2 was the least prevalent of the MMR gene variants associated with Lynch syndrome.
The risk of any Lynch syndrome–associated cancer by age 70 years has been found to range between 57% to nearly 80% in MSH2 pathogenic variant carriers.
The lifetime risk of colon cancer associated with MSH2 pathogenic variants is estimated to be between 48% and 68%.
In a case series of Lynch syndrome patients, those carrying germline MSH2 pathogenic variants (49 individuals, 45% women) had a lifetime (cutoff age, 60 y) risk of extracolonic cancers of 48% compared with 11% for MLH1 carriers (56 individuals, 50% women).
In a more recent prospective study using pooled European registry data of 616 MSH2 carriers without cancer, the cumulative CRC incidence was 35% at age 70 years, despite colonoscopic surveillance.
The mean age at diagnosis of CRC in MSH2 carriers has been comparable to MLH1 carriers. One study that included 143 affected individuals with MSH2 pathogenic variants found a mean age at CRC diagnosis of 43.9 years (range, 16–90 y). The same study reported a mean age at CRC diagnosis of 42.8 years (range, 16–81 y) in 137 MLH1 pathogenic variant carriers.
Most series have reported a prevalence of germline MSH6 pathogenic variants in approximately 10% of Lynch syndrome families from high-risk clinics and a higher proportion of unselected CRC patients, at approximately 50%.
The reported prevalence of MSH6 pathogenic variants in the InSiGHT database was 18% in 2012.
The wide range of prevalence estimates for pathogenic MSH6 variants was a result of small sample sizes, ascertainment bias, and the later age of CRC onset and less striking family histories in MSH6-associated Lynch syndrome families compared with MLH1- and MSH2-associated Lynch syndrome families.
This is in line with findings from a population-based study of 42 carriers of deleterious MSH6 germline pathogenic variants, 30 (71%) of whom had a family cancer history that did not meet the Amsterdam II criteria.
In a recent, international, population-based study of 5,744 CRC cases, the prevalence of MSH6 pathogenic variants was estimated to be 1 in 758, ranking as the second most prevalent of the MMR genes following PMS2.
The lifetime risk of any Lynch syndrome–associated cancer among MSH6 pathogenic variant carriers is approximately 25%
with CRC lifetime risk estimated to be between 12% and 22%
with MSH6 carriers diagnosed with CRC at a later age than MLH1 and MSH2 carriers. In an earlier study of 146 MSH6 carriers (59 men and 87 women) from 20 families, all of whom had truncating pathogenic variants in MSH6, there was a similar prevalence of CRC by age 70 years among MLH1, MSH2, and MSH6 carriers (P = .0854). However, the mean age at diagnosis for colorectal carcinoma was (a) 55 years for male MSH6 carriers (n = 21; range, 26–84 y) versus 43 years and 44 years in carriers of MLH1 and MSH2 pathogenic variants, respectively; and (b) 57 years for female MSH6 carriers (n = 15; range, 41–81 y) versus 43 years and 44 years in carriers of MLH1 and MSH2 pathogenic variants, respectively.
The largest series of carriers of MSH6 pathogenic variants reported to date includes 113 families from five countries who were ascertained through family cancer clinics and population-based cancer registries.
Compared with the incidence for the general population, MSH6 pathogenic variant carriers had an eightfold increased incidence of CRC (hazard ratio [HR], 7.6; 95% CI, 5.4–10.8), which was independent of sex and age. By age 70 years, 22% (95% CI, 14%–32%) of male carriers of MSH6 pathogenic variants developed CRC compared with 10% (95% CI, 5%–17%) of female carriers. By age 80 years, the CRC prevalence doubled to 44% (95% CI, 28%–62%) of male carriers of MSH6 pathogenic variants diagnosed with CRC compared with 20% (95% CI, 11%–35%) among female carriers.
In a more recent prospective study using pooled European registry data of 305 MSH6 carriers without cancer, the cumulative CRC incidence was 20% at age 70 years despite colonoscopic surveillance.
PMS2 was the last of the genes in the MMR family of genes to be identified. This was because lower penetrance among families made it more difficult to identify
using clinical criteria, and also because of limitations of DNA mutational analysis that result from pseudogene interference.
In earlier studies of individuals with CRC and suspected Lynch syndrome, the prevalence of PMS2 pathogenic variants was variable from 2.2% to 5%,
with an increase to 7.5% as reported in the InSiGHT database in 2012.
From a study examining universal tumor testing results from unselected cases of CRC in Switzerland, IHC evaluation of 1,000 consecutive cases found isolated absence of PMS2 expression in 1.5% of all tumors. If this frequency of PMS2-deficient CRCs were representative of all PMS2-associated Lynch syndrome, PMS2 would be the most common gene associated with Lynch syndrome.
Results from a large, population-based CRC cohort found that the prevalence of PMS2 pathogenic variants was the highest among all MMR variants, in which 1 person in 714 carried a pathogenic PMS2 gene variant.
The lifetime risk of any cancer has been found to range between 25% and 32% for heterozygous PMS2 pathogenic variant carriers.
A meta-analysis of three population-based studies and one clinic-based study estimated that for carriers of PMS2 pathogenic variants, the risk of CRC to age 70 years was 20% among men and 15% among women, and the risk of endometrial cancer was 15%.
Similarly, a European consortium of clinic-based registries, taking care to correct for ascertainment bias, found a cumulative lifetime (to age 70 y) CRC risk of only 19% in men and 11% in women with PMS2 pathogenic variants.
In addition, patients with PMS2 pathogenic variants presented with CRC 7 to 8 years later than did those with MLH1 and MSH2 pathogenic variants.
In a prospective study using pooled European registry data of 77 PMS2 carriers without cancer, the cumulative CRC incidence was 10% at age 70 years despite colonoscopic surveillance.
An analysis of nearly 5,000 patients from 284 PMS2 families from the European consortium, supplemented by data from two more registries, was intended to provide more robust PMS2-associated cancer risk estimates.
The risk of CRC up to age 80 years was 13% (95% CI, 7.9%–22%) for men and 12% (95% CI, 6.7%–21%) for women, compared with general population risk estimates of 6.6% and 4.7%, respectively. Endometrial cancer risk was found to be 13% (95% CI, 7%–24%). No excess risk of other Lynch syndrome–spectrum tumors was identified in these cohorts. The authors concluded that these data justify consideration of delaying initiation of colonoscopy until age 35 to 40 years, and with longer follow-up intervals (2–3 y), although this was not specifically studied. As with the original reports from the European Prospective Lynch Syndrome Database, it was not possible to assess the extent to which such colonoscopies and polypectomies might have reduced the rate of detected CRCs.
The PLSD is a major ongoing initiative to assess cancer risks in Lynch syndrome. Although it lacks specific details regarding screening practices, it includes outcome data from many European programs, classified by age, gender, and MMR gene.
Recognizing limitations in the larger PLSD, a subset with more detailed surveillance data has been provided.
These prospective colonoscopy data from Germany, Holland, and Finland included 2,747 patients of whom 62 had no prior cancer at surveillance initiation. Because of differences in surveillance practices, the colonoscopy interval approximated 1 year in Germany, 2 years in Holland, and 3 years in Finland. The median number of colonoscopies was five and the median per-patient observation time was approximately 8 years. Despite the differences in surveillance intervals, similar adenoma detection rates were found in those patients with a history of cancer (14%) and those without (15.6%). At 10 years of follow-up, rates of first cancer were 8.4% and 14% for metachronous tumors. Factors increasing risk were male gender, prior CRC, presence of MLH1 or MSH2 pathogenic variants, age older than 40 years, and adenoma at index colonoscopy. Notably, no significant difference in CRC detection or in stage at detection was noted between screening intervals of 1, 2, or 3 years.
It is important to note that a more severe phenotype is seen among carriers of biallelic PMS2 pathogenic variants. (Refer to the BMMRD section in the Genetics of Lynch syndrome section of this summary for more information.)
The lifetime risk of CRC and endometrial cancer in carriers of these pathogenic variants is summarized in Table 11.
Gene | Lifetime Risk of Colorectal Cancer (%) | Lifetime Risk of Endometrial Cancer (%) |
---|---|---|
MLH1 | 41–50 | 34–54 |
MSH2 | 35–56 | 21–51 |
MSH6 | 10–22 | 16–49 |
PMS2 | 10 | EPCAM |
A subset of individuals with Lynch syndrome (approximately 1%) have a pathogenic variant in EPCAM, which leads to hypermethylation and inactivation of the MSH2 promoter.
In a European study of 194 EPCAM deletion carriers, the cumulative risk of CRC up to age 70 years was 75% with the average age at onset of 43 years. This is comparable to the risk in MSH2 carriers (up to 68% by age 70 y). However, the risk of endometrial cancer among women with an EPCAM deletion was only 12% in this study, compared with a risk of up to 71% in MSH2 carriers.
The associated phenotype is dependent on the location of the deletion variant in the 3’ end of the EPCAM gene; if the deletion is large and includes parts of the promoter of MSH2, the phenotype will be similar to other MSH2-associated Lynch syndrome families.
When the deletion involves the termination signal of EPCAM but spares all of the MSH2 gene and promoter, the phenotype is mainly confined to CRC.
One study of two families with the same EPCAM deletion limited to the 3’ end of the gene and not extending into the promoter of MSH2 found few extracolonic cancers and no endometrial cancers.
However, a subsequent study demonstrated that women with MSH2 protein expression loss caused by EPCAM variants are also at risk of endometrial cancer.
As described above, patients may carry MMR gene variants in both parental alleles, in a condition known as BMMRD. (Refer to the BMMRD section in the Genetics of Lynch syndrome section of this summary for more information.)
The occurrence of such biallelic variants is associated with a characteristic but not diagnostic clinical phenotype. Clinical features include hematologic malignancies and brain tumors in children. When GI tumors occur, the age of onset is strikingly low, sometimes before age 20 years. Café au lait spots and features otherwise suggesting neurofibromatosis are characteristic. Occasionally, patients present with multiple adenomas.
The frequency of MMR variants does not differ markedly from population to population, with similar frequencies identified in a host of different countries. As with hereditary breast and ovarian cancer (HBOC), there are certain variants that occur at higher frequencies within a particular ethnic group. Notable in HBOC are the commonly recurring Ashkenazi Jewish variants, so common that direct-to-consumer testing is offered for these common variants. (Refer to the Population estimates of the likelihood of having a BRCA1 or BRCA2 pathogenic variant section in the PDQ summary on the Genetics of Breast and Gynecologic Cancers and the Direct-to-Consumer (DTC) Genetic Tests section in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for more information.) The ancientness of apparent founder variants is generally established by haplotype analysis. In some instances, what may appear to be a founder variant is simply a frequently recurring de novo variant.
Among the first population findings regarding the MMR genes of Lynch syndrome was the recognition of two very common MLH1 variants in Finland, accounting for a majority of cases of Lynch syndrome in this country.
Since that time, founder variants have been identified in most populations in which relatively unselected series of patients with CRC have undergone variant testing. Many of the reports originate in Europe. As in Finland, these may be straightforward to identify in the setting of fairly homogeneous ethnicity with low immigration. Founder variants in Europe have been found in the United Kingdom, Sweden, Switzerland, Italy,
Portugal, France, Spain, and Hungary, and are likely present in all ethnic groups. Fewer such reports have come from Asia,
Latin America, the Middle East, and Africa.
In the United States, a deletion in exons 1–6 of the MSH2 gene has been estimated to account for as much as 20% of variants in that gene. This so-called American Founder Mutation has been determined by haplotype analysis to date back about 500 years.
A South American study combining data from Uruguay, Colombia, Brazil, Argentina, and Chile also selected cases of interest according to Amsterdam and Bethesda features, yielding a 60% frequency of MLH1 and 40% frequency of MSH2. MSH6 and PMS2 were not evaluated. Selection bias likely influenced the frequency of variants and perhaps the relative contributions by MLH1 and MSH2. A possible founder variant in Colombia was noted.
Although testing for commonly recurring founder variants in a given ethnic/geographic area has been considered to be a cost-effective first step when a step-wise strategy is employed, it is likely not necessary when the increasingly commonly approach of broad panel testing is undertaken as a basic strategy.
One consideration related to ethnicity is that of increased rates of consanguinity within certain populations and the subsequent risk of BMMRD. (Refer to the Biallelic mismatch repair deficiency [BMMRD] section of this summary for more information.)
In this section, the data exploring the distribution of MMR gene variants amongst differing ethnic groups in the United States are presented. The interpretation of these studies is challenging given the presence of selection and ascertainment bias. In addition, even population-based studies are limited by small sample sizes for many ethnic groups and self-reporting of ethnicity/race.
There are few data suggesting the presence of much variation in Lynch syndrome frequency according to geography or ethnicity. Within a small and/or homogeneous ethnic group the presence of founder variants may seem to increase the prevalence of variants in that particular gene. Slight differences in the proportion of MLH1 and MSH2 variants exist from one population to another. MSH6 and PMS2 have been insufficiently studied at the population level as to enable inferences about their relative frequencies.
The most representative population-based studies in the United States, such as that in Columbus, Ohio, have been overrepresented by whites, in accordance with their greater overall numbers. Consequently, data on minorities such as Hispanics and African Americans suffer from smaller and less rigorously representative samples.
A study conducted in Puerto Rico considered variants in 89 Caribbean Hispanic patients with Lynch syndrome suspected on the grounds of Amsterdam criteria or Bethesda guidelines.
Patients underwent either immediate germline testing or step-wise evaluation beginning with tumor MSI/IHC. Frequencies of variants by gene were 67% for MSH2, 25% for MLH1, and 8% for MSH6. No definite founder variants were evident. Clearly, the selection of participants according to clinical family history criteria would have led to an underreporting of the less penetrant MSH6 and PMS2 genes.
Clinic-based series from California, Texas, and Puerto Rico yielded an overall variant prevalence similar to those described, with somewhat more MLH1 than MSH2, but also including MSH6 and PMS2. Presence of potential founder variants traceable back to Spain and Europe were noted.
The closest population-based information on Lynch syndrome in Hispanics is a Southern California study based on the California Tumor Registry, in which 265 patients were identified.
Of those with MSI-H tumors, 13 (62%) had MMR variants. Frequencies of MMR variants were 46% for MLH1 (6 of 13), 31% for MSH2 (4 of 13), 15% for MSH6 (2 of 13), and 8% for PMS2 (1 of 13).
The problem of small numbers is highlighted by the findings from the more truly population-based studies that have been done in the United States. In a study from Columbus, Ohio, only 8% of the consecutive series patients were African American and the proportion of Hispanics as a subset of whites was not stated.
In another study involving panel testing of nearly all CRC patients treated at Dana-Farber Cancer Institute, less than 5% were African American and less than 3% were Hispanic, underscoring the challenge of extracting meaningful data from small subsets.
The issues in evaluating prevalence of Lynch syndrome and cancer risks associated with MMR variants in African Americans are similar to those in Hispanics: a heterogeneous population that has been understudied. A study of clinic-based data from 13 referral centers in the United States identified 51 families with Lynch syndrome with frequencies of MMR gene variants as follows: 61% MLH1, 21% MSH2, 6% MSH6, and 12% PMS2. Age of cancer onset distribution curves were very similar to those seen in white populations.
As with most of the studies in Hispanics, cases were not identified according to any consistent, programmatic evaluation such as universal tumor testing.
A hallmark feature of Lynch syndrome is that carriers of pathogenic MMR gene variants have an increased risk of development of synchronous and metachronous colorectal neoplasms. In one study of 382 individuals with Lynch syndrome from the Colon Cancer Family Registry, the incidence of metachronous CRCs was 16% at 10 years, 41% at 20 years, and 63% at 30 years after segmental colectomy.
The risk of metachronous CRC decreased in a stepwise fashion by 31% for every 10 cm of the colon that was removed, with none of the 50 individuals who had extensive colectomies diagnosed with metachronous CRC. Another prospective study of 1,273 patients with Lynch syndrome who had prior cancer reported a cumulative incidence of subsequent CRC of 46% for MLH1 carriers, 48% for MSH2 carriers, and 23% for MSH6 carriers. This represents only a slightly greater risk of new cancers than pathogenic variant carriers with no previous cancer diagnosis. Excellent survival was again seen and was regarded as a combination of favorable tumor pathology and the effect of surveillance.
Patients with Lynch syndrome are at an increased risk of other cancers, especially those of the endometrium. The cumulative risk of extracolonic cancer has been estimated to be 20% by age 70 years in 1,018 women in 86 families, compared with 3% in the general population.
There is some evidence that the rate of individual cancers varies from kindred to kindred.
The most common extracolonic malignancy in Lynch syndrome is endometrial adenocarcinoma, which affects at least one female member in about 50% of Lynch syndrome families. In addition, 50% of women with an MMR gene pathogenic variant will present with endometrial cancer as her first malignancy.
The lifetime risk of endometrial cancer has been estimated to be from 44% in carriers of MLH1 pathogenic variants to 71% in carriers of MSH2 pathogenic variants, although some earlier studies may have overestimated risk due to ascertainment bias.
Lifetime risk of endometrial cancer in carriers of MSH6 pathogenic variants in 113 families was estimated to be 26% at age 70 years and 44% at age 80 years;
overall, female carriers of MSH6 pathogenic variants had an endometrial cancer risk that was 25 times higher than women in the general population (HR, 25.5; 95% CI, 16.8–38.7; P < .001).
In another study, the cumulative lifetime risk of uterine cancer was higher in MSH6 carriers (71%) than in carriers of MLH1 (27%) and MSH2 (40%) pathogenic variants (P = .02), with an older mean age at diagnosis of 54 years in carriers of MSH6 pathogenic variants (n = 29; range, 43–65 y) versus 48 years in carriers of MLH1 and 49 years in carriers of MSH2 pathogenic variants.
In carriers of PMS2 pathogenic variants, the endometrial cancer risk at age 70 years has been reported to be 15%.
Prospective data collected in the Colon Cancer Family Registry program yielded 5-year endometrial cancer risks of about 3% and 10-year endometrial cancer risks of about 10% among women with MMR gene pathogenic variants.
A prospective study using pooled European registry data of 1,942 MMR carriers without prior cancer reported a cumulative incidence of endometrial cancer of 34% in MLH1 carriers, 51% in MSH2 carriers, 49% in MSH6 carriers, and 24% in PMS2 carriers.
Women with loss of MSH2 protein expression caused by an EPCAM pathogenic variant are also at risk of endometrial cancer depending upon the location of the variant in EPCAM. One study found a 12% (95% CI, 0%–27%) cumulative risk of endometrial cancer in EPCAM deletion carriers.
A study of 127 women with Lynch syndrome who had endometrial cancer as their index cancer were found to be at significantly increased risk of other cancers. The following elevated risks were reported: CRC, 48% (95% CI, 27.2%–58.3%); kidney, renal pelvis, and ureter cancer, 28% (95% CI, 11.9%–48.6%); urinary bladder cancer, 24.3% (95% CI, 8.56%–42.9%; and breast cancer, 2.51% (95% CI, 1.17%–4.14%).
In a study of 113 families that carried MSH6 pathogenic variants from the Colon Cancer Family Registry, female MSH6 carriers had a 26-fold increased incidence of endometrial cancer (HR, 25.5; 95% CI, 16.8–38.7) compared with the general population. A sixfold increased incidence of other cancers associated with Lynch syndrome (HR, 6.0; 95% CI, 3.4–10.7) was observed compared with the general population, but not among male MSH6 carriers.
Lynch syndrome–associated endometrial cancer is not limited to the endometrioid subtype, and the spectrum of uterine tumors in Lynch syndrome may include clear cell carcinoma, uterine papillary serous carcinoma, and malignant mixed Müllerian tumors.
Also, endometrial cancer most commonly arises from the lower uterine segment. (Refer to the Endometrial cancer screening in Lynch syndrome section of this summary for information about screening methods.)
Multiple studies demonstrate an increased risk of additional malignancies associated with Lynch syndrome, including cancers of the stomach, pancreas, ovary, small intestine, and brain, transitional cell carcinoma of the bladder, ureters, and renal pelvis, and sebaceous adenomas of the skin.
In addition, some studies have suggested an association with breast, prostate, and adrenal cortex cancers.
The strength of the association for many of these malignancies is limited by the majority of studies having a small sample size (and consequently, wide CIs associated with relative risk [RR]), the retrospective nature of the analyses, and referral or ascertainment bias.
The largest prospective study to date is of 446 unaffected carriers of pathogenic variants from the Colon Cancer Family Registry.
The Colon Cancer Family Registry is an international cohort with both population-based and clinic-based recruitment from six centers in North America and Australia. Control subjects were noncarriers from families with a known MMR pathogenic variant. Three subcohorts were used to analyze the risk of CRC (365 carriers, 903 noncarriers), endometrial cancer (215 carriers, 523 noncarriers), and other cancers (446 carriers, 1,029 noncarriers). Participants who were followed for up to 10 years demonstrated an increased SIR for CRC (SIR, 20.48; 95% CI, 11.71–33.27; P < .01), endometrial cancer (SIR, 30.62; 95% CI, 11.24–66.64; P < .001), ovarian cancer (SIR, 18.81; 95% CI, 3.88–54.95; P < .001), gastric cancer (SIR, 9.78; 95% CI, 1.18–35.30; P = .009), renal cancer (SIR, 11.22; 95% CI, 2.31–32.79; P < .001), bladder cancer (SIR, 9.51; 95% CI, 1.15–34.37; P = .009), pancreatic cancer (SIR, 10.68; 95% CI, 2.68–47.70; P = .001), and female breast cancer (SIR, 3.95; 95% CI, 1.59–8.13; P = .001).
A well-described variant of Lynch syndrome whose phenotype includes multiple cutaneous neoplasms (including sebaceous adenomas, sebaceous carcinomas, and keratoacanthomas) and CRC is Muir-Torre syndrome.
Pathogenic variants in the MLH1, MSH2, and MSH6 genes have been found in Muir-Torre families with an increased prevalence described among MSH2 carriers.
A study of 1,914 unrelated MLH1 and MSH2 probands found MSH2 to be more common in individuals with the Muir-Torre syndrome phenotype. Of 15 individuals with sebaceous skin tumors, 13 (87%) had MSH2 pathogenic variants compared with two individuals who had MLH1 pathogenic variants (P = .05).
Evidence of defective DNA MMR activity using IHC or MSI testing was reported in 69 of 163 randomly collected sebaceous neoplasms (42%), suggesting that this is a common mechanism for the development of these lesions, and that testing for defective MMR in sebaceous neoplasms would be an ineffective means to screen for Lynch syndrome or Muir-Torre syndrome.
(Refer to the Sebaceous Carcinoma section in the PDQ summary on Genetics of Skin Cancer for more information about cutaneous neoplasms in Muir-Torre syndrome.)
Cancer Site b | General Population Risk (%) c | Risk in Individuals With Lynch Syndrome (%) d |
---|---|---|
Stomach | <1 | 0.2–13 |
Ovary | 1.3 | 3.4–22 |
Hepatobiliary tract | <1 | 0.02–4 |
Urinary tract | <1 | 0.2–25.5 |
Small bowel | <1 | 0.4–12 |
Brain/CNS | <1 | 1.2–3.7 |
Sebaceous neoplasms | <1 | 9.0 |
Pancreas | 1.6 | 0.4–3.7 |
CNS = central nervous system. | ||
aAdapted from Syngal et al. | ||
bEvolving data suggest a potential association between Lynch syndrome and breast and prostate cancers. (Refer to the Additional cancers potentially associated with Lynch syndrome section of this summary for more information about these cancers.) | ||
cHowlader et al. | ||
dRange of cancer risk estimates vary based on study sample size, subject ascertainment, and statistical methods. |
Additional tumors are being considered as part of the spectrum of Lynch syndrome, but this is controversial. Breast and prostate cancers have been raised as possible Lynch syndrome–associated tumors such that MMR genes are now included on multigene (panel) tests for these cancers.
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.
Prostate cancer was found to be associated with Lynch syndrome in a study of 198 families from two U.S. Lynch syndrome registries in which prostate cancer had not originally been part of the family selection criteria. Prostate cancer risk in relatives of carriers of MMR gene pathogenic variants was 6.3% at age 60 years and 30% at age 80 years, versus a population risk of 2.6% at age 60 years and 18% at age 80 years, with an overall HR of 1.99 (95% CI, 1.31–3.03).
A 2014 meta-analysis supports this association, finding an estimated RR of 3.67 (95% CI, 2.32–6.67) for prostate cancer in men with a known MMR pathogenic variant.
This risk is possibly increased in those with MSH2 pathogenic variants.
Notwithstanding prevalent controversy surrounding routine prostate-specific antigen (PSA) screening, the authors suggested that screening by means of PSA and digital rectal exam beginning at age 40 years in male MMR gene carriers would be “reasonable to consider.”
A study of 692 men with metastatic prostate cancer unselected for family history of cancer or age at diagnosis identified germline MMR pathogenic variants in four men (0.5%).
Currently, molecular and epidemiologic evidence supports prostate cancer as one of the Lynch syndrome cancers. As with breast cancer,
additional studies are needed to define absolute risks and age distribution before surveillance guidelines for prostate cancer can be developed for carriers of MMR pathogenic variants. (Refer to the MMR Genes section in the PDQ summary on Genetics of Prostate Cancer for more information about prostate cancer and Lynch syndrome.)
In a series of 114 ACC cases, of which 94 patients had a detailed family history assessment and Li-Fraumeni syndrome was excluded, three patients had family histories that were suggestive of Lynch syndrome. The prevalence of MMR gene pathogenic variants in 94 families was 3.2%, similar to the proportion of Lynch syndrome among unselected colorectal and endometrial cancer patients. In a retrospective review of 135 MMR gene pathogenic variant–positive Lynch syndrome families from the same program, two probands were found to have had a history of ACC. Of the four ACCs in which MSI testing could be performed, all were MSS. These data suggest that if Lynch syndrome is otherwise suspected in an ACC index case, an initial evaluation of the ACC using MSI or IHC testing may be misleading.
Several additional cancers have been found to be associated with Lynch syndrome in some studies, but further investigation is warranted. Table 12 compares the risk of these cancers in the general population with that of individuals with Lynch syndrome.
Several aspects of the biologic behavior of CRC and its precursor lesion, the adenomatous polyp, in individuals with Lynch syndrome support a different approach to CRC screening in this population as compared with those recommendations for average-risk people in the general population. At present, the recommendations for cancer screening and surveillance in Lynch syndrome take into account the differences in cancer risks as compared with those in the general population due to the causative germline deficiency in the MMR system. The following biological differences form the basis of the currently implemented screening strategies in Lynch syndrome:
CRCs in Lynch syndrome occur earlier in life than do sporadic cancers; however the age of onset varies based on which of the MMR genes is altered. (Refer to the Prevalence, clinical manifestations, and cancer risks associated with Lynch syndrome section of this summary for more information about gene-specific age of onset of CRC.)
Carriers of Lynch syndrome pathogenic variants have an increased risk of developing colon adenomas and the onset of adenomas appears to occur at a younger age than in pathogenic variant–negative individuals from the same families.
The risk of a carrier of MMR pathogenic variants developing adenomas has been reported to be 3.6 times higher than the risk in noncarriers.
By age 60 years, 70% of the carriers developed adenomas, compared with 20% of noncarriers. Most of the adenomas in carriers had absence of MMR protein expression and were more likely to have dysplastic features, compared with adenomas from control subjects.
In one study, the mean age at diagnosis of adenoma in carriers was 43.3 years (range, 23–63.2 y), and the mean age at diagnosis of carcinoma was 45.8 years (range, 25.2–57.6 y).
A larger proportion of Lynch syndrome CRCs (60%–70%) occur in the right colon, suggesting that sigmoidoscopy alone is not an appropriate screening strategy and that a colonoscopy provides a more complete structural examination of the colon. Evidence-based reviews of surveillance colonoscopy in Lynch syndrome have been reported.
The incidence of CRC throughout life is substantially higher in patients with Lynch syndrome, suggesting that the most-sensitive test available should be used. (Refer to Table 13 for available colon surveillance recommendations.)
The progression from normal mucosa to adenoma to cancer is accelerated,
suggesting that screening should be performed at shorter intervals (every 1–2 years) and with colonoscopy.
It has been demonstrated that carriers of MMR gene pathogenic variants develop detectable adenomas at an earlier age than do noncarriers.
It is not known whether this reflects a greater prevalence of adenomas or the presence of larger adenomas with better detection in Lynch syndrome.
The risk of CRC in Lynch syndrome has been studied and updated in a Finnish screening trial, which spans from the early 1980s to present.
Over the course of this trial, the design of the longitudinal study has evolved. In the earliest period, information about each individual's variant status was unknown and study participants were eligible based on fulfillment of clinical criteria; the study consisted of some people with a previous cancer or adenoma diagnosis and others without such history who were undergoing asymptomatic screening while the comparison group was composed of individuals from those same families who refused screening. Many of these people (68%) had screening with x-ray contrast/barium enema. Colonoscopy was the approach used for carriers of MMR pathogenic variants when this information was obtainable and the interval between exams was shortened from 5 years to 3 years to 2 years, based on results from the study over time.
A 15-year controlled screening trial conducted in this series demonstrated a reduction in the incidence of CRC, CRC-specific mortality, and overall mortality with colonoscopy in individuals from Lynch syndrome families.
Colonic screening was provided at 3-year intervals in 133 individuals from Lynch syndrome families and 119 controls from these families had no screening. Among those screened, 8 individuals (6%) developed CRC compared with 19 control subjects (16%), for a risk reduction of 62% with screening. Furthermore, all CRCs in the screened group were local, causing no deaths, while there were 9 deaths caused by CRC in the control group. There was also a benefit in overall mortality in the screened group with 10 deaths in the screened group and 26 deaths in the control group (P = .003).
The series subsequently limited its attention to subjects without prior diagnosis of adenoma or cancer. The eligible 420 carriers of pathogenic variants had a mean age of 36 years and underwent an average of 2.1 colonoscopies, with a median follow-up of 6.7 years. Adenomas were detected in 28% of subjects. Cumulative risk of one or more adenomas by age 60 years was 68.5% in men and 48.3% in women. Notably, risk of detecting cancer in those free of cancer at baseline exam, and thus regarded as interval cancers, by age 60 years was 34.6% in men and 22.1% in women. The combined cumulative risk of adenoma or cancer by age 60 years was 81.8% in men and 62.9% in women. For both adenomas and carcinomas, about one-half were located proximal to the splenic flexure. While the rates for CRC despite colonoscopy surveillance appear high, the recommended short intervals were not regularly adhered to in this nonrandomized series. These authors recommended surveillance at 2-year intervals. This is in line with most consensus guidelines (refer to Table 13), in which the appropriate colonoscopy screening interval remains every 1 to 2 years. Analysis of colonoscopic surveillance data in 242 carriers of pathogenic variants 10 years after testing shows 95% compliance in surveillance procedures for CRC and endometrial cancer. Although not all CRCs were prevented, mortality was comparable with variant-negative relatives. However, this may be attributable to the modest sample size of the study.
Given that colonoscopy is the accepted measure for colon cancer surveillance, preliminary data suggest that the use of chromoendoscopy, such as with indigo carmine, may increase the detection of diminutive, histologically advanced adenomas.
When an adenoma is detected, the question of whether to test the adenoma for MSI/IHC is raised. One study of patients with prior CRC and known MMR pathogenic variants found eight of 12 adenomas to have both MSI and IHC protein loss.
However, the study authors emphasized that normal MSI/IHC testing in an adenoma does not exclude Lynch syndrome. Abnormal MSI/IHC are uncommon in the smallest adenomas, and more prevalent in adenomas larger than 8 mm, which also suggests that the MMR defect is acquired in the growing adenoma.
Because of the variability of gene-specific CRC risks, experts in the field have proposed gene-specific screening and surveillance recommendations. For example, a European consortium
made a clinical recommendation for delaying the onset of colorectal and endometrial cancer screening to age 30 years, in line with their recommendation for later initiation of screening for carriers of MSH6 pathogenic variants. Additionally, a 2015 review by an ad hoc American virtual workgroup involved in the care of Lynch syndrome patients and families concluded that despite multiple studies indicating reduced penetrance in monoallelic PMS2 carriers, they could not recommend any changes to Lynch syndrome cancer surveillance guidelines for this group.
While initial data may support different strategies for the initiation and surveillance of CRC and other extracolonic cancers by specific MMR gene alteration,
concerns related to (a) the adherence of recommendations overall by the medical community and by affected individuals
and (b) limitations related to specific screening modalities
have prevented the implementation of gene-specific guidelines until additional data are available.
Organization | Age Screening Initiated | Screening Interval | Recommended Screening Modality | Comments |
---|---|---|---|---|
NCCN (2019) | 20–25 y or 2–5 y before youngest case of CRC in family if before age 25 y | 1–2 y | Colonoscopy | For MSH6 carriers, consider a later age for colonoscopy initiation such as at age 30 y or 10 y younger than the age of any relative with CRC. Due to limited data for PMS2 gene carriers, the panel is not able to make a specific recommendation regarding later age of onset for colonoscopy. |
U.S. Multi-Society Task Force on Colorectal Cancer (2014)b | 20–25 y or 2–5 y before youngest case of CRC in family if before age 25 y | 1–2 y (annual for carriers of MMR pathogenic variants) | Colonoscopy | For MSH6 and PMS2 carriers, consider starting screening at ages 30 y and 35 y, respectively, unless an early-onset cancer occurs in the family. Recommendations for individuals with BMMRD are also available. |
Mallorca group (2013) | 20–25 y | 1–2 y | Colonoscopy | |
ESMO (2013)c | 20–25 y or 5 y before youngest case of CRC in family; no upper limit established | 1–2 y | Colonoscopy | |
BMMRD = biallelic mismatch repair deficiency; CRC = colorectal cancer; ESMO = European Society for Medical Oncology; IHC = immunohistochemistry; MMR = mismatch repair; MSI = microsatellite instability; NA = not addressed; NCCN = National Comprehensive Cancer Network. | ||||
aThis table summarizes available guidelines from 2010 and later. Other organizations, including the American Cancer Society, have published guidelines before 2010. | ||||
bU.S. Multi-Society Task Force on Colorectal Cancer includes the following organizations: American Academy of Family Practice, American College of Gastroenterology, American College of Physicians-American Society of Internal Medicine, American College of Radiology, American Gastroenterological Association, American Society of Colorectal Surgeons, and American Society for Gastrointestinal Endoscopy. | ||||
cThe American Society of Clinical Oncology and the Japanese Society of Medical Oncology have endorsed the ESMO guidelines as presented in the table. |
Note: A separate PDQ summary on Endometrial Cancer Screening in the general population is also available.
Cancer of the endometrium is the most common extracolonic cancer observed in Lynch syndrome families, affecting at least one female in about 50% of Lynch syndrome families. (Refer to the Endometrial cancer section of this summary for more information about gene-specific risks of endometrial cancer in carriers of MMR pathogenic variants.)
In the general population, the diagnosis of endometrial cancer is generally made when women present with symptoms including abnormal or postmenopausal bleeding. Endometrial sampling is performed to provide a histologic specimen for diagnosis. Eighty percent of women with endometrial cancer present with stage I disease and there are no data to suggest that the clinical presentation in women with Lynch syndrome differs from that in the general population.
Given their substantial increased risk of endometrial cancer, endometrial screening for women with Lynch syndrome has been suggested. Proposed modalities for screening include transvaginal ultrasound (TVUS) and/or endometrial biopsy. TVUS continues to be widely recommended without data to support its use; current NCCN guidelines suggest that there is no clear evidence to support endometrial cancer screening for Lynch syndrome.
Two studies have examined the use of TVUS in endometrial screening for women with Lynch syndrome.
In one study of 292 women from Lynch syndrome families or "Lynch syndrome-like/HNPCC-like" families, no cases of endometrial cancer were detected by TVUS. In addition, two interval cancers developed in symptomatic women.
In a second study, 41 women with Lynch syndrome were enrolled in a TVUS screening program. Of 179 TVUS procedures performed, there were 17 abnormal scans. Three of the 17 women had complex atypical hyperplasia on endometrial sampling, while 14 had normal endometrial sampling. However, TVUS failed to identify one patient who presented 8 months after a normal TVUS with abnormal vaginal bleeding, and was found to have stage IB endometrial cancer.
Both of these studies concluded that TVUS is neither sensitive nor specific.
A study of 175 women with Lynch syndrome, which included both endometrial sampling and TVUS, showed that endometrial sampling improved sensitivity compared with TVUS. Endometrial sampling found 11 of the 14 cases of endometrial cancer. Two of the three other cases were interval cancers that developed in symptomatic women and one case was an occult endometrial cancer found at the time of hysterectomy. Endometrial sampling also identified 14 additional cases of endometrial hyperplasia. Among the group of 14 women with endometrial cancer, ten also had TVUS screening with endometrial sampling. Four of the ten had abnormal TVUS, but six had normal TVUS.
While this cohort study demonstrated that endometrial sampling may have benefits over TVUS for endometrial screening, there are no data that predict that screening with any other modality has benefits for endometrial cancer survival in women with Lynch syndrome.
Some studies suggest that women with a clinical or genetic diagnosis of Lynch syndrome do not universally adopt intensive gynecologic screening.
(Refer to the Gynecologic cancer screening in Lynch syndrome section in the Psychosocial Issues in Hereditary Colon Cancer Syndromes section of this summary for more information.)
Estimates of the cumulative lifetime risk of ovarian cancer in Lynch syndrome patients range from 3.4% to 22%.
However, no studies on the effectiveness of ovarian screening are currently available for women in Lynch syndrome families. TVUS used for endometrial cancer screening has been extended to include ovarian cancer screening in clinical practice for those women who do not undergo risk-reducing surgery for gynecological cancer prevention. However, NCCN asserts that data do not support routine ovarian cancer screening for Lynch syndrome due to a lack of sensitivity and specificity of available screening modalities.
Level of evidence: None assigned
An effective strategy for the prevention of endometrial and ovarian cancers in Lynch syndrome families is risk-reducing surgery. A retrospective study of 315 women with pathogenic MMR gene variants compared the rate of endometrial and ovarian cancer among the women who did and did not have hysterectomy and oophorectomy. In women followed for endometrial cancer, the mean follow-up periods were 13.3 years in the surgical group and 7.4 years in the nonsurgical group; in women followed for ovarian cancer, the mean follow-up periods were 11.2 years in the surgical group and 10.6 years in the nonsurgical groups. For those women in the surgical group, no cancers were diagnosed, compared with a 33% rate of endometrial cancer and a 5.5% rate of ovarian cancer in the nonsurgical group.
Cost-effectiveness–analysis modeling of risk-reducing surgeries (prophylactic hysterectomy and bilateral salpingo-oophorectomy) versus nonsurgical screening in a theoretical population of carriers aged 30 years with MMR gene variants associated with Lynch syndrome revealed that prophylactic surgery was cost-effective with lower cost and yielded higher QALY.
A subsequent modeling study evaluated multiple screening and surgical strategies and found that annual screening initiated at age 30 years followed by risk-reducing surgery at age 40 years was the most effective strategy.
The decision to screen for other Lynch syndrome–associated cancers is done on an individual basis and relies on the cancers reported among FDRs and second-degree relatives with Lynch syndrome.
The lifetime risk of gastric cancer is approximately 8% for male Lynch syndrome carriers and 5% for female Lynch syndrome carriers.
Recent epidemiologic data report a decreasing trend in the diagnosis of gastric cancer than was previously reported, which was as high as 13%. The histologic characterization of most Lynch syndrome–associated gastric cancer is of the intestinal type and may thereby be detected using screening esophagogastroduodenoscopy (EGD).
Although there are no clear data to support surveillance for gastric, duodenal, and more distal small bowel cancers, EGD with visualization of the duodenum at the time of colonoscopy can be used in individuals with Lynch syndrome with a baseline examination performed at age 40 years. Evaluation and treatment of H. pylori infection is recommended when found. Despite limited data on appropriate surveillance intervals, there is general consensus that surveillance be performed every 3 to 5 years, particularly if there is a family history of gastric, duodenal, or more distal small bowel cancer or for those of Asian descent.
There are variable reports on the lifetime risk of small bowel cancer associated with Lynch syndrome, ranging from less than 1% to 12%.
Most small bowel malignancies are confined to the duodenum and the ileum, which are within endoscopic reach using EGD and colonoscopy (with dedicated ileal intubation), respectively. Other modalities to assess for small bowel lesions include CT enterography and capsule endoscopy but cost-effectiveness analyses do not support use of these evaluations for routine screening in Lynch syndrome.
Urinary tract malignancies include those of the transitional cell type of the renal pelvis and ureters, and the bladder. The associated lifetime risk of these malignancies is variable, ranging from less than 1% to as high as 25%, with higher estimates related to pooling the cancers found in different locations within the urinary tract and including the bladder.
Studies that have evaluated urinary cytology as a potential screening modality revealed that it was associated with low sensitivity and a high false-positive rate and ultimately leads to additional evaluation that is often invasive (i.e., cystoscopy). There are currently no effective modalities used for routine screening in asymptomatic individuals with Lynch syndrome.
An elevated risk of pancreatic cancer among Lynch syndrome carriers has been supported by two cohort studies that adjust for ascertainment bias. One study reported a cumulative risk of pancreatic cancer of 3.7% by age 70 years and an 8.6-fold increase compared with the general population.
Another prospective study using data from the Colon Cancer Family Registry reported an SIR of 10.7 with cumulative risk of 0.95%.
Results of these studies have supported an expert consensus that recommended screening for pancreatic cancer in individuals with Lynch syndrome and an FDR with pancreatic cancer, similar to other high-risk populations with comparable risk.
Of note, screening for cancers of the urinary tract, bladder, hepatobiliary system, and pancreas is not recommended beyond that for the general population; however, NCCN suggests the consideration of urothelial cancer surveillance for individuals with a family history of urothelial cancer or individuals with MSH2 pathogenic variants (especially males).
The Colorectal Adenoma/Carcinoma Prevention Programme (CAPP2) was a double-blind, placebo-controlled, randomized trial to determine the role of aspirin in preventing CRC in patients with Lynch syndrome who were in surveillance programs at a number of international centers.
The study randomly assigned 861 participants to receive aspirin (600 mg/day), aspirin placebo, resistant starch (30 g/day), or starch placebo for up to 4 years. At a mean follow-up of 55.7 months (range, 1–128 months), 53 primary CRCs developed in 48 participants (18 of 427 in the aspirin group and 30 of 434 in the aspirin placebo group). Seventy-six patients who refused randomization to the aspirin groups (because of an aspirin sensitivity or a history of peptic ulcer disease) were randomly assigned to receive resistant starch or resistant starch placebo. The intent-to-treat analysis yielded an HR for CRC of 0.63 (95% CI, 0.35–1.13; P = .12). However, five of the patients who developed CRC developed two primary colon cancers. A Poisson regression was performed to account for the effect of the multiple primary CRCs and yielded a protective effect for aspirin (incidence rate ratio [IRR], 0.56; 95% CI, 0.32–0.99; P = .05). For participants who completed at least 2 years of treatment, the per-protocol analysis yielded an HR of 0.41 (95% CI, 0.19–0.86; P = .02) and an IRR of 0.37 (0.18–0.78; P = .008). An analysis of all Lynch syndrome cancers (endometrial, ovarian, pancreatic, small bowel, gallbladder, ureter, stomach, kidney, and brain) revealed a protective effect of aspirin versus placebo (HR, 0.65; 95% CI, 0.42–1.00; P = .05). There were no significant differences in adverse events between the aspirin and placebo groups, and no serious adverse effects were noted with any treatment. The authors concluded that 600 mg of aspirin per day for a mean of 25 months substantially reduced cancer incidence in Lynch syndrome patients. CAPP2 failed to show any effect from daily resistant starch intake. A limitation of the trial is that the frequency of surveillance studies at the various centers was not reported as being standardized. Earlier CAPP2 trial results for 746 Lynch syndrome patients enrolled in the study were published in 2008
and failed to show a significant preventive effect on incident colonic adenomas or carcinomas (relative risk, 1.0; 95% CI, 0.7–1.4) with a shorter mean follow-up of 29 months (range, 7–74 months). A 2015 survey of 1,858 participants in the Colon Cancer Family Registry suggested that aspirin and ibuprofen might be chemopreventive for carriers of MMR gene pathogenic variants.
The CAPP3 trial, which is evaluating the effect of lower doses of aspirin (blinded 100 mg, 300 mg, and 600 mg enteric-coated aspirin), began in 2013 and is expected to enroll approximately 3,000 carriers of pathogenic variants by about 2021.
Despite level 1 evidence, experts believe that the evidence regarding aspirin use for the chemoprevention of Lynch syndrome is not sufficiently robust or mature to recommend its standard use.
One of the hallmark features of Lynch syndrome is the presence of synchronous and metachronous CRCs. The incidence of metachronous CRCs has been reported to be 16% at 10 years, 41% at 20 years, and 63% at 30 years after segmental colectomy.
Because of the increased incidence of synchronous and metachronous neoplasms, the recommended surgical treatment for a patient with Lynch syndrome with neoplastic colonic lesions is generally an extended colectomy (total or subtotal). Nevertheless, treatment has to be individualized and has often included segmental colectomy. Mathematical models suggest that there are minimal benefits of extended procedures in individuals older than 67 years, compared with the benefits seen in younger individuals with early-onset cancer. In one Markov decision analysis model, the survival advantage for a young individual with early-onset CRC undergoing an extended procedure could be up to 4 years longer than that seen in the same individual undergoing a segmental resection.
The recommendation for an extended procedure must be balanced with the comorbidities of the patient, the clinical stage of the disease, the wishes of the patient, and surgical expertise. No prospective or retrospective study has shown a survival advantage for patients with Lynch syndrome who underwent an extended resection versus a segmental procedure.
Two studies have shown that patients who undergo extended procedures have fewer metachronous CRCs and additional surgical procedures related to CRC than do patients who undergo segmental resections.
Balancing functional results of an extended procedure versus a segmental procedure is of paramount importance. Although the majority of patients adapt well after an abdominal colectomy, some patients will require antidiarrheal medication. A decision model compared QALYs for a patient aged 30 years undergoing an abdominal colectomy versus a segmental colectomy.
In this model, there was not much difference between the extended and segmental procedure, with QALYs being 0.3 years more in patients undergoing a segmental procedure than in those undergoing an extended procedure.
When considering surgical options, it is important to recognize that a subtotal or total colectomy will not eliminate the rectal cancer risk. The lifetime risk of developing cancer in the rectal remnant after an abdominal colectomy has been reported to be 12% at 12 years post-colectomy.
In addition to the general complications of surgery are the potential risks of urinary and sexual dysfunction and diarrhea after an extended colectomy; these risks increase as the anastomosis becomes more distal. Therefore, the choice of surgery must be made on an individual basis by the surgeon and the patient.
In patients with Lynch syndrome and rectal cancer, similar surgical options (extended vs. segmental resection) and considerations must be given. Extended procedures include restorative proctocolectomy and IPAA if the sphincter can be saved, or proctocolectomy with loop ileostomy if the sphincter cannot be saved. The risk of metachronous colon cancer after segmental resection for an index rectal cancer has been reported to be between 15% and 27%.
Two retrospectives studies reported a 15% and 18% incidence of metachronous colon cancer after segmental rectal cancer–resection in patients with Lynch syndrome.
In one of the studies, the combined risk of metachronous high-risk adenomas and cancers was 51% at a median follow-up of 101.7 months after proctectomy.
There are no data about fertility after surgery in Lynch syndrome patients. In female FAP patients, no difference in fecundity after abdominal colectomy and IRA has been reported, whereas there is a 54% decrease in fecundity in patients who undergo restorative proctocolectomy with IPAA compared with the general population.
Another study in which a questionnaire was sent to FAP patients reported a similar prevalence of fertility problems among patients who had undergone IRA, IPAA, and proctocolectomy with end ileostomy. In that study, it was reported that earlier age at the time of surgery was associated with more fertility problems.
Most clinicians who treat patients with Lynch syndrome will favor an extended procedure at the time of CRC diagnosis. However, as stated above, the choice of surgery must be made on an individual basis by the surgeon and the patient.
As discussed in previous sections, MSI is not only a molecular feature of Lynch syndrome, but is also present in 10% to 15% of sporadic cases of CRC (largely due to MLH1 hypermethylation or biallelic somatic mutations in an MMR gene). Although MSI testing was initially utilized to screen patients who might harbor pathogenic MMR gene variants, it has been increasingly recognized that MSI has important prognostic and therapeutic implications. The utility of MSI testing beyond identifying Lynch syndrome has made the case for universal MSI screening more compelling, and has contributed to its widespread adoption. Several studies have suggested that stage-specific survival is better for MSI-H CRC compared with MSS cancers. Additionally, the chemotherapeutic agent fluorouracil (5-FU) appears ineffective in the adjuvant treatment of resected MSI-H CRC, in contrast to MSS CRC in which this agent is widely utilized for this purpose. Finally, immunomodulation with agents such as checkpoint inhibitors appears effective in the treatment of advanced MSI-H CRC based on early phase 1 and phase 2 studies, while these agents, at least when utilized as monotherapy, show little activity in MSS CRC.
Although MSI-H tumors account for 15% of all sporadic CRC, they appear to be more frequent in stage II compared with stage III CRC, and are even less common in metastatic disease, being present in only 3% to 4% of metastatic cases. This stage distinction alludes to the possibility of a better prognosis associated with underlying MSI-H status.
Several studies subsequently confirmed the improved survival of stage II MSI-H CRC compared with MSS cases. A meta-analysis of 32 studies of 7,642 cases, including 1,277 with MSI-H, showed a combined HR estimate for overall survival (OS) associated with MSI of 0.65 (95% CI, 0.59–0.71; heterogeneity P = .16; I2 [a measure of the percentage of variation across studies that is due to heterogeneity rather than chance] = 20%).
However, while data were limited, tumors with MSI derived no benefit from adjuvant 5-FU (HR, 1.24; 95% CI, 0.72–2.14). Subsequent data from several large randomized clinical trials confirmed the favorable prognosis associated with MSI-H. These included the QUick And Simple And Reliable (QUASAR) trial, which explored the benefit of adjuvant 5-FU–based chemotherapy compared with surgery alone in 1,900 patients with resected stage II CRC. In this study, MSI-H tumors were associated with a recurrence risk of half that of MSS tumors (risk ratio [RR], 0.53; 95% CI, 0.40–0.70).
Similar results were seen in the Pan European Trial Adjuvant Colon Cancer (PETACC)-3 trial, a randomized trial of 5-FU with or without irinotecan in resected stage II or stage III CRC.
MSI-H status was associated with an OS odds ratio (OR) of 0.39 (95% CI, 0.24–0.65) and this advantage was seen in both stage II and stage III disease.
Consistent with other prior data, clinicopathologic analysis of 85 Lynch syndrome–associated CRCs and 67 sporadic dMMR CRCs demonstrated a significantly superior survival among patients with Lynch syndrome, as well as younger ages at diagnosis and higher numbers of tumor-infiltrating lymphocytes (TILs).
Exome sequencing and neoantigen data from a subset of 16 CRC tumors (eight Lynch syndrome CRCs and eight sporadic dMMR CRCs) from this cohort suggest that somatic mutational burden and neoantigen load is significantly higher among Lynch syndrome–associated CRCs than sporadic dMMR CRCs; this was speculated to be the source of the improved survival outcomes and increased TILs.
Given the predilection for MSI-H tumors to involve the right side of the colon, there is a paucity of data on the outcome and prognosis of MSI-H tumors involving the rectum. One study suggested only 2% of rectal cancers are MSI-H.
A study of 62 patients with MSI-H rectal cancers from a single institution were followed for a median of 6.8 years. The 5-year rectal cancer–specific survival was 100% for stage I and stage II, 85.1% for stage III, and 60.0% for stage IV disease, suggesting the favorable prognosis associated with MSI-H may also apply to cancers involving the rectum.
The authors additionally reported a favorable 26% pathologic complete response rate with 5-FU combined with radiation therapy, suggesting that 5-FU given with radiation for the locoregional treatment of rectal cancer may still be effective in the setting of MSI-H tumors. The substantial rate of pathologic complete responses demonstrated in this study also reinforces the need for adequate biopsies to assess MSI status prior to commencing treatment.
The finding of MSI in a CRC has been shown in several studies to predict the lack of benefit of adjuvant chemotherapy with 5-FU in resected stage II or stage III colon cancer.
This has been a controversial area historically. It was known that loss of DNA MMR activity in cultured colon cancer cells conferred resistance to DNA-damaging agents (the common mechanism of cytotoxic chemotherapy) through loss of the signal to arrest the cell cycle in response to DNA damage that cannot be repaired.
This led to the prediction that DNA dMMR tumors may not be fully sensitive to alkylating agents, 5-FU, and platinum-containing drugs.
Unexpectedly, in 2000, a paper was published suggesting that patients with Dukes C (stage III) CRC with MSI had a substantial survival benefit when given 5-FU–based adjuvant chemotherapy.
However, the patients in this analysis had not been randomized to therapy; they were selected for adjuvant chemotherapy based upon clinical status, and inadvertently, the median age in the treatment group was 13 years younger than the controls.
In 2003, however, the outcomes in a randomized controlled prospective trial of adjuvant chemotherapy in 570 colon cancer patients demonstrated no benefit from adjuvant 5-FU in the group with MSI. Moreover, there were nonsignificant trends towards increased mortality when colon cancers with MSI were treated: twofold for stage III cancers and threefold for stage II cancers.
Subsequently, ten studies confirmed this, as all failed to show benefit when CRC patients were given 5-FU–based chemotherapy.
In contrast, a meta-analysis of randomized trials of 5-FU versus observation suggested a potential benefit of 5-FU in patients with MSI stage III disease. An exploratory subset analysis suggested benefit only in those patients with Lynch syndrome–related MSI. An analysis of stage II patients was not undertaken in this study.
Preclinical data suggests the addition of oxaliplatin to 5-FU can overcome the resistance to 5-FU monotherapy seen in MSI-H tumors.
A retrospective analysis of 433 MSI-H stage II and stage III CRC cases (both sporadic and secondary to Lynch syndrome) suggested a benefit in disease-free survival (DFS) with FOLFOX (5-FU and oxaliplatin) compared with surgery alone.
There was a trend towards improved DFS utilizing FOLFOX in the subset of patients with MSI due to Lynch syndrome, however, the result was not statistically significant. Additional studies have demonstrated similar survival outcomes irrespective of MSI status with adjuvant chemotherapy including FOLFOX.
Tumors that develop via the MSI pathway have more somatic mutations than tumors that develop via other pathways. This could imply that dMMR tumors may have more potential antigens (termed neoantigens) and may be more responsive to immune system manipulation than proficient MMR (pMMR) tumors. Microscopically, MSI-H tumors often exhibit abundant tumor-infiltrating lymphocytes, sometimes resulting in a Crohn-like reaction. This histologic feature has long suggested the possibility of increased tumor immune surveillance in MSI-H cancers, and is one of the main hypotheses for the better stage-specific survival seen in MSI-H compared with MSS cancers.
To test the hypothesis of efficacy of immunomodulation in MSI-H tumors, a phase 2 trial of programmed cell death-1 (PD-1) inhibition was carried out in a small cohort of patients with MSI-H or MSS cancers. Patients with metastatic disease that had failed various chemotherapy regimens were treated with pembrolizumab, an anti–PD-1 immune checkpoint inhibitor.
In this small phase 2 study, 32 patients with CRC (11 were dMMR, 21 were pMMR, and 9 others had noncolorectal dMMR tumors) were treated with intravenous pembrolizumab every 14 days. The immune-related response among evaluable patients was 40% (4 of 10) for dMMR CRC tumors, 0% (0 of 18) for pMMR CRC tumors, and 71% (5 of 7) for non-CRC dMMR tumors. The immune-related 20-week progression-free survival was 78% (7 of 9) in patients with dMMR CRC tumors, 11% (2 of 18) in patients with pMMR CRC tumors, and 67% (4 of 6) in patients with non-CRC dMMR tumors. dMMR tumors had a mean of 24-fold more somatic mutations than pMMR tumors. Additionally, in this study somatic mutation load was associated with prolonged PFS. The authors concluded that MMR status predicted clinical benefit to immune checkpoint blockade with pembrolizumab.
A single-arm phase 2 study (CheckMate 142) of another PD-1 inhibitor, nivolumab, was performed in 74 patients with MSI-H/dMMR CRC that had progressed on prior cytotoxic chemotherapy (including 5-FU, irinotecan, and oxaliplatin).
Overall, 31% of patients (23 of 74) experienced an objective response to therapy, and 69% (51 of 74) had disease control for at least 12 weeks. Among patients who responded to nivolumab, the median duration of response was not reached at the time of study analysis (median follow up of 12 months). There was no significant difference in the response rates among individuals with Lynch syndrome–associated metastatic MSI-H/dMMR CRC versus non-Lynch metastatic MSI-H/dMMR CRC in this study. Twenty percent of study participants experienced grade 3 or greater toxicities, most commonly elevations in amylase and/or lipase, and there were no deaths that were attributed to nivolumab.
Based on these data, pembrolizumab 200 mg given intravenously every 3 weeks was approved by the FDA in May 2017 for the treatment of any MSI-H/dMMR metastatic cancer that is refractory to standard therapy and nivolumab 240 mg given intravenously every 2 weeks was granted accelerated approval by the FDA in August 2017 for the treatment of MSI-H/dMMR CRC that is refractory to cytotoxic chemotherapy.
In another arm of CheckMate 142, 119 individuals with metastatic dMMR CRC were treated with nivolumab plus ipilimumab.
The objective response rate was 55% with a 12-week disease control rate of 80%, a 12-month PFS of 71%, and a medial duration of response that was not reached. Grade 3 and grade 4 toxicities occurred in 32% of participants (most commonly increased liver function tests) and 13% of all participants discontinued therapy due to toxicity. This was a nonrandomized study, and thus questions remain as to whether the combination of immune checkpoint blockade is superior to PD-1 inhibition alone, especially given the apparent increase in toxicity with combination therapy. On the basis of these data, in July 2018 the FDA granted accelerated approval to nivolumab plus ipilimumab therapy for the treatment of dMMR/MSI-H metastatic CRC that has progressed through prior chemotherapy with a fluoropyrimidine, oxaliplatin, and irinotecan.
An alternative approach to immunotherapy in MSI-H CRC involves the use of tumor-directed vaccines. The most promising approaches thus far involve the use of tumor-related neoantigens as epitopes to increase tumor-specific T-cell immunity. Studies are currently under way in the adjuvant treatment of resected stage III CRC (NCT01461148), in patients with metastatic disease (NCT01885702), and in the prevention of CRC in patients with Lynch syndrome (NCT01885702).
Lynch-like syndrome may account for up to 70% of cases in which Lynch syndrome is suspected but germline testing fails to identify a pathogenic MMR gene variant.
Similar to the tumor phenotype seen in Lynch syndrome, CRCs manifest MSI and IHC loss of a DNA MMR protein. However, the MMR-deficient CRCs are due to biallelic somatic inactivation of DNA MMR genes,
in which a somatic mutation in one allele of the MMR gene along with loss of heterozygosity of the other allele is most probable versus the presence of two somatic sequence mutations. (Refer to Table 10 for more information about the tumor phenotype of Lynch-like syndrome.)
Possible explanations for the cause of Lynch-like syndrome include the following: (1) the possibility that some germline DNA variants are not detected by current testing; (2) affected individuals may have germline pathogenic variants in genes other than DNA MMR genes currently known to be associated with Lynch syndrome; or (3) there are other mechanisms that inactivate DNA MMR beyond those related to alterations in the germline.
There is growing evidence that the CRC risk among probands and families with Lynch-like syndrome are lower, with an SIR of 2.12, than in Lynch syndrome, with an SIR of 6.04.
Preliminary estimates reveal a lower risk of extracolonic cancers with a SIR of 1.69 in Lynch-like syndrome versus 2.81 in Lynch syndrome. Another retrospective study of 14 patients with early-onset (<50 y) CRC and dMMR reported that 43% of patients had Lynch syndrome and 57% had Lynch-like syndrome.
In the absence of large-scale studies with longitudinal follow-up, in addition to data pertaining to the rates of neoplastic progression in Lynch-like syndrome, intensive cancer screening recommendations are currently similar to those in Lynch syndrome guidelines.
The term familial colorectal cancer type X or FCCX was coined to refer to families who meet Amsterdam criteria but lack MSI/IHC abnormalities.
Approximately 50% of families that fulfill Amsterdam criteria, lack pathogenic MMR gene variants and thereby are characterized as FCCX families. Research is ongoing to determine a genetic etiology for FCCX, but for the most part it remains unknown and is thought to be a heterogeneous condition. However, differentiating between Lynch syndrome and FCCX has important implications regarding cancer risk assessment and screening recommendations for affected individuals and at-risk relatives. While the risk of CRC is increased to twice that in the general population, it is less than that in Lynch syndrome (>sixfold increase) and there is no significant risk of extracolonic malignancy. Cancer screening recommendations are therefore modified and CRC surveillance is recommended every 5 years.
The epidemiology of CRC with regard to age at diagnosis is shifting with individuals increasingly being diagnosed before age 50 years.
(Refer to the PDQ summary on Colorectal Cancer Prevention for more information about CRC incidence trends in the general population.) One study that examined the prevalence of highly penetrant pathogenic variants in 450 individuals with young-onset CRC (mean age at diagnosis, 42.5 y) and a family history including at least one FDR with colon, endometrial, breast, ovarian, and/or pancreatic cancer identified 75 germline pathogenic or likely pathogenic variants in 72 patients (16%).
The spectrum of variants identified included Lynch syndrome and non-Lynch syndrome–associated genes, including several genes that have not traditionally been associated with CRC (e.g., BRCA1/BRCA2, ATM, CHEK2, PALB2, and CDKN2A). Given the high frequency and variety of hereditary cancer syndromes identified, the authors suggest that multigene (panel) testing in this population may be warranted.
In the absence of additional family or personal history suggestive of Lynch syndrome, isolated cases of CRC diagnosed before age 36 years are uncommonly associated with MMR gene pathogenic variants. One study found MMR pathogenic variants in only 6.5% of such individuals,
whereas another study of CRC patients younger than 50 years with no more than one FDR with CRC found abnormal MSI in 21% of tumors and overrepresentation of defects in the PMS2 and MSH6 genes.
Therefore, isolated cases of very early-onset CRC should be offered tumor screening with MSI/IHC rather than proceeding directly to germline pathogenic variant analysis.
Performance of endoscopic therapies for adenomas in FAP and Lynch syndrome, and decision-making regarding surgical referral and planning, require accurate estimates of the presence of adenomas. In both AFAP and Lynch syndrome the presence of very subtle adenomas poses special challenges—microadenomas in the case of AFAP and flat, though sometimes large, adenomas in Lynch syndrome.
The need for sensitive means to endoscopically detect subtle polyps has increased with the recognition of flat adenomas and sessile serrated polyps in otherwise average-risk subjects, very attenuated adenoma phenotypes in AFAP, and subtle flat adenomas in Lynch syndrome. Modern high-resolution endoscopes improve adenoma detection yield, but the use of various vital dyes, especially indigo carmine dye-spray, has further improved detection. Several studies have shown that the improved mucosal contrast achieved with the use of indigo carmine can improve the adenoma detection rate. Whether family history is significant or not, careful clinical evaluation consisting of dye-spray colonoscopy (indigo carmine or methylene blue),
with or without magnification, or possibly newer imaging techniques such as narrow-band imaging,
may reveal the characteristic right-sided clustering of more numerous microadenomas. Upper GI endoscopy may be informative if duodenal adenomas or fundic gland polyps with surface dysplasia are found. Such findings will increase the likelihood of variant detection if APC or MUTYH testing is pursued.
In various large series of average-risk populations, subtle flat lesions were detected in about 5% to 10% of cases, including adenomas with high-grade dysplasia and invasive adenocarcinoma.
Some of these studies involved tandem procedures—white-light exam followed by randomization to “intensive” (> 20-minute pull-back from cecum) inspection versus chromoendoscopy—with significantly more adenomas detected in the chromoendoscopy group.
However, in several randomized trials, no significant difference in yield was seen.
In a randomized trial of subjects with Lynch syndrome,
standard colonoscopy, with polypectomy as indicated, was followed by either indigo carmine chromoendoscopy or repeat “intensive” white-light colonoscopy (a design very nearly identical to the average-risk screening group noted above). In this series, no significant difference in adenoma yield between the chromoendoscopy and intensive white-light groups was detected. However, these patients were younger and in many cases had undergone several previous exams that might have resulted in polyp clearing.
In a German study,
one series of Lynch syndrome patients underwent white-light exam followed by chromoendoscopy, while a second series underwent colonoscopy with narrow-band imaging followed by chromoendoscopy. Significant differences in flat polyp detection favored chromoendoscopy in both series, although some of the detected lesions were hyperplastic. In a French series of Lynch syndrome subjects that also employed white-light exam followed by chromoendoscopy, significantly more adenomas were detected with chromoendoscopy.
Fewer evaluations of chromoendoscopy have been performed in AFAP than in Lynch syndrome. One study examined four patients with presumed AFAP and fewer than 20 adenomas upon white-light examination.
All had more than 1,000 diminutive adenomas found on chromoendoscopy, in agreement with pathology evaluation after colectomy.
A similar role for chromoendoscopy has been suggested to evaluate the duodenum in FAP. One study from Holland that used indigo carmine dye-spray to detect duodenal adenomas showed an increase in the number and size of adenomas, including some large ones. Overall Spigelman score was not significantly affected.
Patients with PJS and JPS are at greater risk of disease-related complications in the small bowel (e.g., bleeding, obstruction, intussusception, or cancer). FAP patients, although at great risk of duodenal neoplasia, have a relatively low risk of jejunoileal involvement. The RR of small bowel malignancy is very high in Lynch syndrome, but absolute risk is less than 10%. Although the risks of small bowel neoplasia are high enough to warrant consideration of surveillance in each disease, the technical challenges of doing so have been daunting. Because of the technical challenges and relatively low prevalences, there is virtually no evidence base for small-bowel screening in Lynch syndrome.
Historically, the relative endoscopic inaccessibility of the mid and distal small bowel required radiographic measures for its evaluation, including the barium small bowel series or a variant called tube enteroclysis, in which a nasogastroduodenal tube is placed so that all of the contrast goes into the small intestine for more precise imaging. None of these measures were sensitive for small lesions. Any therapeutic undertaking required laparotomy. This entailed resection in most cases, although intraoperative endoscopy, with or without enterotomy for scope access, has been available for many years. Peroral enteroscopy (aided by stiffening overtubes with two balloons, one balloon, or spiral ribs) has been employed to overcome the technical problem of excessive looping, enabling deep jejunal access with therapeutic (polypectomy) potential.
Most data relate that PJS with double-balloon enteroscopy is the preferred method for endoscopy of the small bowel.
This may involve only peroral enteroscopy, although subsequent retrograde enteroscopy has been described for more complete evaluation of the total small bowel. Because these procedures are time-consuming and involve some risk of complication, deep enteroscopy is usually preceded by more noninvasive imaging, including traditional barium exams, capsule endoscopy, and CT or magnetic resonance enterography.
In FAP, data from capsule endoscopy
show a 50% to 100% prevalence of jejunal and/or ileal polyps in patients with Spigelman stage III or stage IV duodenal involvement but virtually no such polyps in Spigelman stage I or stage II disease. All polyps were smaller than 10 mm and were not biopsied or removed. Consequently, their clinical significance remains uncertain but is likely limited, given the infrequency of jejunoileal cancer reports in FAP.
Capsule endoscopy in the small series of PJS patients described above
showed the presence of a similar frequency (50%–100%) of polyps, but the prevalent polyps were much larger than in FAP, were more likely to become symptomatic, and warranted endoscopic or surgical excision. Capsule studies were suggested as an appropriate replacement for radiographic studies because of the sensitivity of capsule endoscopy.
Genetic studies have demonstrated a common autosomal dominant inheritance pattern for colon tumors, adenomas, and cancers in familial CRC families,
with a gene frequency of 0.19 for adenomas and colorectal adenocarcinomas.
A subset of families with MSI-negative familial colorectal neoplasia was found to link to chromosome 9q22.2-31.2.
A more recent study has linked three potential loci in familial CRC families on chromosomes 11, 14, and 22.
For more than a decade, little progress has been made on these putative familial cancer loci.
Families meeting Amsterdam-I criteria for Lynch syndrome who do not show evidence of defective MMR by MSI testing do not appear to have the same risk of colorectal or other cancers as those families with classic Lynch syndrome and clear evidence of defective MMR. These Amsterdam-I criteria families with intact MMR systems have been described as FCCX,
and it has been suggested that these families be classified as a distinct group.
The genetic etiology of FCCX remains unclear. Utilizing whole-genome linkage analysis and exome sequencing, a truncating variant in ribosomal protein S20 (RPS20), a ribosomal protein gene, was identified in four individuals with CRC from an FCCX family.
The variant cosegregated with CRC in the family, with a logarithm of the odds score of 3. Additionally, the variant was not identified in 292 controls. No LOH was observed in tumor samples, and in vitro analyses of mature RNA formation confirmed a model of haploinsufficiency for RPS20. No germline variants in RPS20 were found in 25 additional FCCX families studied, suggesting RPS20 variants are an infrequent cause of FCCX. The same group had previously identified variants in the bone morphogenetic protein receptor type 1A (BMPR1A) gene in affected individuals from 2 of 18 families with FCCX.
Additional studies are necessary to definitively confirm or refute a role for RPS20 or BMPR1A in FCCX.
Subsequent to these initial studies, several other putative FCCX genes have been found in familial, non-Lynch syndrome clusters of CRC including the polypeptide N-acetylgalactosaminyltransferase 12 (GALNT12) gene,
BUB1 and BUB3,
the SEMA4A gene,
RINT1,
FAN1,
and combined effects of pathogenic variants in HNRNPA0 and WIF1 in one large kindred.
The list of possible candidate genes will continue to grow, complicating any facile approach to handling these families.
Age of CRC onset in Lynch syndrome ranges from 44 years (registry series) to a mean of 52 years (population-based series).
There are no corresponding population-based data for FCCX because FCCX by definition requires at least one early-onset case, is almost certainly very heterogeneous, and is not likely to lend itself to any population-based figures in the foreseeable future. Studies that have directly compared age of onset between FCCX and Lynch syndrome have suggested that the age of onset is slightly older in FCCX,
and the lifetime risk of CRC is substantially lower. The SIR for CRC among families with intact MMR (FCCX families) was 2.3 (95% CI, 1.7–3.0) in one large study, compared with 6.1 (95% CI, 5.7–7.2) in families with defective MMR (Lynch syndrome families).
The risk of extracolonic tumors was also not found to be elevated in the FCCX families, suggesting that enhanced surveillance for CRC would be sufficient. Although further studies are required, tumors arising within FCCX families also appear to have a different pathologic phenotype, with fewer tumor-infiltrating lymphocytes than those in families with Lynch syndrome.
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.
The age-adjusted risk of CRC was increased in carriers of pathogenic variants in both studies (SIR, 5.7–10.3).
In addition, one study found that 93% of individuals with PTEN pathogenic variants who had undergone at least one colonoscopy had polyps.
The most common histology was hyperplastic, although adenomas and sessile serrated polyps were also observed. The increased risk of CRC among carriers of PTEN pathogenic variants has led to the recommendation of surveillance colonoscopy in these patients.
However, both the age at which to begin (30–40 y) and the subsequent frequency of colonoscopies (biennial to every 3–5 y) vary considerably and are based on expert opinion.
Cancer | Age-Adjusted SIR (95% CI) | Age-Related Penetrance Estimates |
---|---|---|
Breast | 25.4 (19.8–32.0) | 85% starting around age 30 y b |
Colorectal | 10.3 (5.6–17.4) | 9% starting around age 40 y |
Endometrial | 42.9 (28.1–62.8) | 28% starting around age 25 y |
Kidney | 30.6 (17.8–49.4) | 34% starting around age 40 y |
Melanoma | 8.5 (4.1–15.6) | 6% with earliest age of onset at 3 y |
Thyroid | 51.1 (38.1–67.1) | 35% at birth and throughout life |
CI = confidence interval; SIR = standardized incidence ratio. | ||
aAdapted from Tan et al. | ||
bOther historical studies have suggested a lower lifetime risk of breast cancer, in the range of 25%–50%. (Refer to the PTEN hamartoma tumor syndromes [including Cowden syndrome] section in the PDQ summary on Genetics of Breast and Gynecologic Cancers for more information.) |
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 15 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 (%) b |
---|---|---|
Any cancer | 60–70 | 37–93 |
GI cancer c,d | 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 e | 65 | 10 |
Testes e | 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.
JPS is a genetically heterogeneous, rare, childhood- to early adult-onset, autosomal dominant disease that presents characteristically as hamartomatous polyposis throughout the GI tract, although colorectal polyps predominate.
JPS can present with diarrhea, GI tract hemorrhage, protein-losing enteropathy, and prolapsing polyps.
JPS is defined by the presence of a specific type of hamartomatous polyp called a juvenile polyp, often in the setting of a family history of JPS. The diagnosis of a juvenile polyp is based on its histologic appearance, rather than age at onset. Solitary juvenile polyps of the colon or rectum are seen sporadically in infants and young children and do not imply a diagnosis of JPS. A clinical diagnosis of JPS is met by individuals fulfilling one or more of the following criteria:
JPS is caused by germline pathogenic variants in the SMAD4 gene, also known as MADH4/DPC4, at chromosome 18q21
in approximately 15% to 60% of cases,
and by pathogenic variants in the gene encoding the bone morphogenic protein receptor 1A (BMPR1A) residing on chromosome band 10q22 in approximately 25% to 40% of cases.
Because pathogenic variants in SMAD4 and BMPR1A are known to account for juvenile polyposis, clinicians have referred young patients with fewer than five polyps for genetic testing. A study conducted on 77 patients with a total of 84 polyps found that the yield of genetic testing in patients with a limited number of polyps is minimal; of the germline variants detected, none were classified as definitely pathogenic or likely pathogenic.
Genotype/phenotype correlations suggest SMAD4 variants may be associated with a greater risk of severe gastric polyposis
and features of hereditary hemorrhagic telangiectasia (HHT) (refer to the features of HHT below).
The lifetime risk of CRC in JPS has been reported to be 39%.
There appears to be an increased risk of gastric cancer, albeit much lower than the risk of CRC.
Cardiac valvular abnormalities were present in 12% of individuals with JPS who were followed through a single-institution–based polyposis registry,
and all those with identifiable pathogenic variants had SMAD4 variants.
JPS patients with SMAD4 pathogenic variants may also have signs and symptoms of HHT, such as arteriovenous malformations, mucocutaneous telangiectasias, digital clubbing, osteoarthropathy, hepatic arteriovenous malformations, and cerebellar cavernous hemangioma, suggesting that the two syndromes overlap.
When a patient is found clinically to have features of both JPS and HHT, the pathogenic variant will be in the SMAD4 gene. Most patients with isolated HHT will have a pathogenic variant in the activin receptor-like kinase 1 (ALK1) gene or in the endoglin (ENG) gene, but SMAD4 pathogenic variants have also been reported, although they are quite rare (approximately 1%–2% of patients with HHT).
One series suggested a slightly higher incidence of SMAD4 pathogenic variants in unselected patients with HHT. In this study, 3 of 30 patients (10%) with HHT without a clinical diagnosis of JPS were found to have germline variants in SMAD4.
Conversely, features of HHT were noted in 21% to 22% of carriers of SMAD4 pathogenic variants in two studies of individuals with a clinical diagnosis of JPS.
In a study of 21 carriers of SMAD4 pathogenic variants from nine JPS families, 81% (17 of 21) of patients had HHT manifestations.
The high prevalence in this study may have been a result of the inclusion of several relatives from a single family and the inclusion of several families with the same pathogenic variant.
Surveillance for HHT has been suggested in JPS patients with germline SMAD4 pathogenic variants.
On the other hand, patients with HHT without germline variants in ALK1 or ENG may be considered for SMAD4 germline genetic testing; the GI tract should be evaluated if a SMAD4 germline pathogenic variant is confirmed.
(Refer to Table 17, Published Recommendations for Diagnosis and Surveillance of JPS, for more information.)
A severe form of JPS, in which polyposis develops in the first few years of life, is referred to as JPS of infancy. JPS of infancy is often caused by microdeletions of chromosome 10q22-23, a region that includes BMPR1A and PTEN. (Refer to the PTEN hamartoma tumor syndromes [including Cowden syndrome] section of this summary for more information about PTEN.) The phenotype often includes features such as macrocephaly and developmental delay, possibly as a result of loss of PTEN function.
Recurrent GI bleeding, diarrhea, exudative enteropathy, in addition to associated developmental delay, are associated with a very high rate of morbidity and mortality in these infants, thereby limiting the heritability of such cases.
JPS is caused by germline pathogenic variants in the SMAD4 gene in approximately 15% to 60% of cases, and to pathogenic variants in BMPR1A in approximately 25% to 40% of cases.
The large variability in variant frequency likely reflects the relatively small number of patients reported in individual studies. A subset of individuals meeting clinical criteria for JPS will not have an identified pathogenic variant in either SMAD4 or BMPR1A.
SMAD4 encodes a protein that is a component of the transforming growth factor (TGF)-beta signaling pathway, which mediates growth inhibitory signals from the cell surface to the nucleus. Germline pathogenic variants in SMAD4 predispose individuals to forming juvenile polyps and cancer,
and germline variants have been found in 6 of 11 exons. Most variants are unique, but several recurrent pathogenic variants have been identified in multiple independent families.
Patients with SMAD4 pathogenic variants are also at high risk for developing extracolonic GI cancers such as gastric cancers, often in the context of gastric polyposis.
BMPR1A is a serine-threonine kinase type I receptor of the TGF-beta superfamily that, when activated, leads to phosphorylation of SMAD4. The BMPR1A gene was first identified by linkage analysis in families with JPS who did not have identifiable pathogenic variants in SMAD4. Variants in BMPR1A include nonsense, frameshift, missense, and splice-site variants.
Large genomic deletions detected by MLPA have been reported in both BMPR1A and SMAD4 in patients with JPS.
Rare JPS families have demonstrated variants in the ENG and PTEN genes, but these have not been confirmed in other studies.
Several studies initially suggested that a subset of families with hereditary breast and colon cancers may have a cancer family syndrome caused by a pathogenic variant in the CHEK2 gene.
However, subsequent studies have suggested that CHEK2 variants are associated with only a modest increase in CRC risk (i.e., low penetrance). One large study showed that truncating variants in CHEK2 were not significantly associated with CRC; however, a specific missense pathogenic variant (I157T) was associated with modest increased risk (OR, 1.5; 95% CI, 1.2–3.0) of CRC.
Similar results were obtained in another study conducted in Poland.
In this study, 463 probands from Lynch syndrome and Lynch syndrome–related families and 5,496 controls were genotyped for four CHEK2 pathogenic variants, including I157T. The missense I157T allele was associated with Lynch syndrome–related cancer only for MMR variant-negative cases (OR, 2.1; 95% CI, 1.4–3.1). There was no association found with the truncating variants. Further studies are needed to confirm this finding and to determine whether they are related to FCCX.
(Refer to the section in the PDQ summary on Genetics of Breast and Gynecologic Cancers for more information.)
HMPS is a rare cancer family syndrome characterized by the development of a variety of colon polyp types, including serrated adenomas, atypical juvenile polyps and adenomas, and colon adenocarcinoma. Although initially mapped to a locus between 6q16-q21, the HMPS locus is now believed to map to 15q13-q14.
While there is considerable phenotypic overlap between JPS and HMPS, one large family has been linked to a locus on chromosome 15, raising the possibility that this may be a distinct disorder. Linkage analysis of Ashkenazi Jewish families with HMPS revealed shared haplotypes on chromosome 15q13.3.
An unusual heterozygous 40kb single-copy duplication was discovered upstream of gremlin 1 (GREM1) that segregated perfectly with individuals and family members with HMPS and not with unaffected controls.
The presence of this duplication in HMPS individuals was associated with increased expression of GREM1 transcript levels in the normal intestinal epithelium.
GREM1 is a bone morphogenetic protein (BMP) antagonist and thus theoretically would promote the stem cell phenotype in the intestine. Germline variants leading to defective BMP signaling also underlie JPS, thus drawing a potential link between HMPS and JPS.
Although exceedingly rare, GREM1 pathogenic variants have been described in several additional families of Ashkenazi Jewish ancestry, with varying clinical presentations. Although polyposis appears to be a unifying feature in most families, there is a high degree of variability with respect to polyp number, histology, and age of onset. In addition, extracolonic malignancies have been described in several pathogenic variant carriers, although the small number of affected individuals limits the ability to definitively demonstrate a causal link to the GREM1 pathogenic variant. On the basis of relatively limited data, it is reasonable to consider GREM1-variant analysis in Ashkenazi Jewish families presenting with unexplained polyposis and/or familial CRC.
In such families, comprehensive variant analysis that includes testing for duplications in noncoding regions of GREM1 is necessary.
Isolated and multiple hyperplastic polyps (HPs) (typically white, flat, and small) are common in the general population, and their presence does not suggest an underlying genetic disorder. Historically, the clinical diagnosis of SPS, as defined by WHO, must satisfy one of the following criteria:
Although the vast majority of cases of SPS lack a family history of HPs, approximately half of the SPS cases have a positive family history of CRC.
Several studies show that the prevalence of colorectal adenocarcinoma in patients with formally defined criteria for SPS is 50% or more.
One study, using a variation of the WHO criteria for SPS (SPS was defined as at least five histologically diagnosed HPs and/or sessile serrated adenomas (SSAs) proximal to the sigmoid colon, of which two are greater than 10 mm in diameter, or more than 20 HPs and/or SSAs distributed throughout the colon), found an RR for CRC in 347 FDRs (41% male) from 57 pedigrees of 5.4 (95% CI, 3.7–7.8).
The WHO criteria are based on expert opinion; and, there is no known susceptibility gene or genomic region that has been reproducibly linked to this disorder, so genetic diagnosis is not possible. Two studies have reported potentially causative germline variants in SPS individuals.
In a study of 38 patients with more than 20 HPs, a large (>1 cm) HP, or HPs in the proximal colon, molecular alterations were sought in the base-excision repair genes MBD4 and MUTYH.
One patient was found to have biallelic MUTYH pathogenic variants, and thus was diagnosed with MUTYH-associated polyposis. No pathogenic variants were detected in MBD4 among 27 patients tested. However, six patients had single nucleotide polymorphisms of uncertain significance. Only two patients had a known family history of SPS, and ten of the 38 patients developed CRC. This series presumably included patients with sporadic HPs mixed in with other patients who may have SPS.
In a cohort of 40 SPS patients, defined as having more than five HPs or more than three HPs, two of which were larger than 1 cm in diameter, one patient was found to have a germline variant in the EPHB2 gene (D861N).
The patient had serrated adenomas and more than 100 HPs in her colon at age 58 years, and her mother died of colon cancer at age 36 years. EPHB2 germline variants were not found in 100 additional patients with a personal history of CRC or in 200 population-matched healthy control patients.
Far more is known about the somatic molecular genetic alterations found in the colonic tumors occurring in SPS patients. In a study of patients with either more than 20 HPs per colon, more than four HPs larger than 1 cm in diameter, or multiple (5–10) HPs per colon, a specific somatic BRAF mutation (V600E) was found in polyp tissue.
Fifty percent of HPs (20 of 40) from these patients demonstrated the V600E BRAF pathogenic variant. The HPs from these patients also demonstrated significantly higher CpG island methylation phenotypes (CIMP-high), and fewer KRAS variants than left-sided sporadic HPs. In a previous study from this group, HPs from patients with SPS showed a loss of chromosome 1p in 21% (16 of 76) versus 0% in HPs from patients with large HPs (>1 cm), or only five to ten HPs.
Many of the genetic and histological alterations found in HPs of patients with SPS are common with the CIMP pathway of colorectal adenocarcinoma. Sporadic serrated polyps are the precursors to CRCs of the CIMP pathway. (Refer to the CIMP and the serrated polyposis pathway section in the Introduction section of this summary for more information.)
Individuals with PJS and JPS are at increased risk of CRC and extracolonic cancers. Because these syndromes are rare, there have been no evidence-based surveillance recommendations. Because of the markedly increased risk of colorectal and other cancers in these syndromes, a number of guidelines have been published based on retrospective and case series (i.e., based exclusively on expert opinion).
Clinical judgment must be used in making screening recommendations based on published guidelines.
Organization | STK11 Gene Testing Recommended a | Age Colon Screening Initiated | Frequency | Method | Extracolonic Screening Recommendations | Comment |
---|---|---|---|---|---|---|
Johns Hopkins (2006) | Yes, at age 8 y | 18 y | 2–3 y | C | Breast, gynecologic (cervix, ovaries, uterus), pancreas, small intestine, stomach, testes | |
Johns Hopkins (2007) | Yes, age not specified | Late teens or at onset of symptoms | 3 y | C | Breast, gynecologic (cervix, ovaries, uterus), pancreas, small intestine, stomach, testes | Genetic testing in the late teens or at onset of symptoms. |
ACPGBI (2007) | 18 y | 3 y | C or FS + BE | No mention of extracolonic screening | No recommendation for genetic testing; need to consider STK11/LKB1 testing. | |
Cleveland Clinic (2007) | 18 y | 3 y | C | Breast, gynecologic (cervix, ovaries), pancreas, small intestine, stomach, testes | ||
Erasmus University Medical Center (2010) | 25–30 y | C | Breast, gynecologic (cervix, ovaries, uterus), pancreas, small intestine, stomach | |||
NCCN (2019) | No specific recommendation | Late teens | 2–3 y | C | Breast (women), gynecologic (cervix, ovaries, uterus), lungb, pancreas, small intestine, stomach, testes | Refer to specialized team. |
ACPGBI = Association of Coloproctology of Great Britain and Ireland; BE = barium enema; C = colonoscopy; FS = flexible sigmoidoscopy; NCCN = National Comprehensive Cancer Network. | ||||||
aSTK11 testing includes sequencing followed by analysis for deletions (e.g., multiplex ligation-dependent probe amplification), if no variant found by sequencing. | ||||||
bLung cancer risk is increased, but there are no recommendations beyond smoking cessation and heightened awareness of symptoms. | ||||||
(Refer to the Other High-Penetrance Syndromes Associated With Breast and/or Gynecologic Cancer section in the PDQ summary on the Genetics of Breast and Gynecologic Cancers for more information about PJS and the risk of breast and ovarian cancer.) |
Organization/ Author | SMAD4 / BMPR1A Testing Recommendeda | Age Screening Initiated | Frequency | Method | Comment |
---|---|---|---|---|---|
ACPGBI (2007) | 15–18 y b | 1–2 y | C or FS + BE | Surveillance for gene carriers and affected until age 70 y and discussion of prophylactic surgery. | |
Cleveland Clinic (2007) | 15 y | 3 y | C, EGD | Some families with SMAD4 pathogenic variant also have HHT; these individuals may need to be screened for HHT. | |
Johns Hopkins (2007) | Yes, genetic testing preferred over C | 15 y or at onset of symptoms | Yearly until polyp free then every 2–3 y | C | Prophylactic surgery if >50–100 polyps, unable to manage endoscopically, severe GI bleeding, JPS with adenomatous changes, strong family history of CRC. |
St. Mark's (2012) | Yes, genetic testing at age 4 y | 12 y | 1–3 y based on severity | C, EGD | Consider HHT workup. |
NCCN (2019) | Yes | 15 y | 2–3 y or 1 y if polyps are found | C | Refer to specialized team. In families without an identified pathogenic variant, consider substituting endoscopy every 5 y beginning at age 20 y and every 10 y beginning at age 40 y in patients in whom no polyps are found. |
ACPGBI = Association of Coloproctology of Great Britain and Ireland; BE = barium enema; C = colonoscopy; CRC = colorectal cancer; EGD = esophagogastroduodenoscopy; FS = flexible sigmoidoscopy; GI = gastrointestinal; HHT = hereditary hemorrhagic telangiectasia; NCCN = National Comprehensive Cancer Network. | |||||
aSMAD4/BMPR1A testing includes sequencing followed by analysis for deletions (e.g., multiplex ligation-dependent probe amplification), if no variant found by sequencing. | |||||
bYounger, if patient has presented with symptoms. |
癌症遗传咨询和检测中的心理社会研究侧重于在不同疾病风险水平的人群中进行检测的兴趣、心理结果、人际和家庭影响以及文化和社区反应。这项研究还确定了鼓励或阻碍监测和其他健康行为的行为因素。心理社会研究得出的数据可以指导临床医生与患者的交流,可能包括以下内容:
本总结这一部分将着重于林奇综合征、家族性腺瘤性息肉病(FAP)和黑色素斑-胃肠多发性息肉综合征(PJS)的遗传咨询和检测的心理社会学方面的问题,包括有关这些综合征的医学筛查、降低风险的手术和化学预防问题。
早期评估遗传咨询和检测的研究主要集中在选定的高危研究人群,包括结直肠癌(CRC)患者和未受累的家庭成员,其在很大程度上是根据家族史确定高危CRC患者。参与者主要来自临床和家族结肠癌登记处。大多数研究招募癌症先证病例、典型CRC患者,专门为错配修复(MMR)变体提供遗传咨询和种系检测;这些通常作为免费服务提供。
对具有致病性变体的索引病例的亲属也提供类似的咨询和检测。一项总结这些早期研究的综述报告的检测应用率范围较大,从14%到75%,包括在索引病例和接受检测的高危亲属中的应用。
综述表明,进行遗传检测的主要原因包括希望了解儿童风险,了解早期检测和筛查的需要,以及减少不确定性。检测减少的原因包括花费、保险歧视问题、对本人或家人的潜在负面情绪影响、低预期获益和时间缺少。
尽管这些早期遗传检测应用的研究提供了患者可能有意愿或无意愿进行检测原因的初步见解,但提供遗传咨询和检测的过程不同于已纳入当前临床实践治疗的过程。临床实践较少单独依赖于家族史来确定可能从检测中获益的患者,而是使用MSI和/或IHC作为林奇综合征的初步筛查,对新诊断患者的CRC和子宫内膜癌肿瘤进行普遍的分子诊断检测。(有关更多信息,请参考本总结的“林奇综合征筛查的通用肿瘤检测”一节。)
尽管通用MSI/IHC筛查越来越多应用于确定可能存在种系变体的新确诊患者,但一项重要的提示是,并非所有适合种系检测的患者都遵循推荐的遗传咨询和检测服务。来自单个机构的两份报告发现,发现IHC结果异常后,20%CRC和13%子宫内膜癌索引病例进行了林奇综合征种系变体检测。
这些研究并没有提出继续进行遗传咨询和检测的理由。然而,有人认为,在MSI/IHC结果异常后患者遗传检测的完成度较高可能促使遗传咨询师参与这一过程以揭示筛查阳性结果、提供MSI/IHC检测后咨询,或促进转诊有关。
在凯撒医疗西北部医疗保健系统中的145例CRC患者中开展的研究中,这些患者在知晓MSI结果前接受了调查,大多数患者对MSI/IHC筛查持积极态度。
大多数患者(84.8%)认可MSI/IHC筛查的6个或更多获益;然而,89.4%也认可少于四个潜在障碍,主要是额外的检测和监测的费用。有较强癌症家族史的患者更容易遇到较少MSI/IHC筛查障碍。患者还经历了与筛查有关的轻微痛苦,77.2%的参与者得分为零(表示没有痛苦)。
有关家族史和癌症风险的教育以及鼓励让医护人员提供检测可能促使遗传咨询和检测的应用。对符合癌症遗传风险评估和咨询高风险标准的新诊断 CRC 患者进行的一项小型 (n = 19) 定性研究确定了患者可能未按照建议寻求咨询的潜在原因。这些原因包括不完全了解家族癌症史,没有意识到家族史与个人癌症诊断的相关性;无具体、直接的医生建议进行遗传咨询;以及在处理新发癌症诊断时把心理咨询作为一个较低的优先事项。
在一项随机试验中对91名个体进行随访调查,以促进林奇综合征高危人群的结肠镜筛查,只有24%的人报告曾与其医生讨论过遗传检测,而进行检测的最常见障碍是缺乏医疗保健提供者的建议。
临床实践中越来越多地采用对新确诊林奇综合征肿瘤的通用筛查。然而,这一过程的临床获益和成本效益归因于级联筛查的开展,或在发现有种系变体的癌症索引病例的高危亲属中进行预测性检测。对林奇综合征索引病例的一级亲属遗传检测应用频率和预测因子进行系统评价。
在被纳入综述的四项研究中,报告了其在一级亲属中的应用率,结果显示,34%到52%的一级亲属已经接受了检测。与亲属接受检测相关的因素包括年龄(<50岁)、女性、父母身份、就业状况、教育水平、参与医学研究、心理因素(无抑郁症状)和受癌症影响的亲属人数。
在林奇综合征登记处登记的三代芬兰家庭中进行的一项大规模回顾性遗传检测研究还发现,在有致病性变体的高危亲属中,预测性检测的接受率不完全,并且每代的接受率都在下降。
在1184名有林奇综合征变体的先证者中,67%、43%和24%的一级、二级、三级高危亲属分别进行了预测性检测。在539名第一代林奇综合征变体携带者中,其62%的高危成年子女接受了检测。在多因素分析中,年龄较大、家族特异性变体(MLH1和MSH2与MSH6)、独生子女或有致病性变体的兄弟姐妹、父母坚持结肠镜检查与预测性检测应用相关。这项研究表明,在林奇综合征相关变体个体的高危亲属中,家庭层面的因素,如预测性检测和筛查行为,可能会影响预测性检测。
已发表的关于在林奇综合征家族中增加级联筛查的干预措施的报道有限。澳大利亚的一篇文章比较了两种告知高危亲属遗传性癌症致病性变体,包括林奇综合征的方法。
在该研究中,来自33个接受过遗传检测的家族的索引病例向其临床医生提供了知情同意,允许他们向高危亲属发送详细的信件,建议他们确定家族中遗传性癌症的易感性。信中还包括一项建议,建议与医生或遗传学专家讨论这些信息,且提供了关于遗传学评估包括内容的信息。在随访的前2年内,40%的一级和二级亲属已经进行了预测性遗传检测,其被认定为非携带者,或接受了评估但拒绝了遗传检测。作者将这些发现与41个家族队列进行了比较,这41个家族性变体是在临床医生写信之前发现的,其中变体阳性索引病例只被要求告知亲属遗传检测是可行的。在较早期的队列研究中,23%的高危亲属寻求澄清其遗传风险状况的服务,这与接受临床医生来信组相比显著较少(P=.001)。收到这些信件并没有产生侵犯隐私权或自主权的问题。
有关伦理问题(包括警示职责)的信息,请参阅本PDQ总结中有关癌症遗传学风险评估与咨询部分中的伦理、法律和社会影响部分。
多项研究已经考察了患者在接受林奇综合征遗传咨询和检测之前、期间和之后的心理状态。一些研究只纳入了没有任何林奇综合症相关癌症病史的人,
以及其他研究纳入了CRC患者和未受癌症影响但有患林奇综合征致病性变体风险的人。
对接受林奇综合征遗传咨询和检测的患者心理社会特征进行的横断面评估表明,大多数参与者心理功能的平均预测得分在正常范围内,
尽管一项研究中,比较了受累和未受累的个体,受累的个体与林奇综合征有关的痛苦和担忧更多。
多项纵向研究已经对林奇综合征遗传咨询和检测前与检测结果公布后一年中多个时间段的心理结果进行了评估。一项研究基于个人癌症史、性别和年龄(50岁以下与50岁以上)对在遗传咨询会议预测之前和之后2周的焦虑变化进行了探讨。两个年龄组中受累和未受累的女性参与者,和50岁以上年龄组中受累的男性随着时间的推移焦虑显著降低。未受累的50岁以下男性保持较低的焦虑水平;然而,受累的50岁以下男性在接受预测咨询时的焦虑水平没有降低。
一项在评估受累和未受累的患者在接受咨询后(在检测结果公布之前)8周的心理困扰的研究发现,一般焦虑、癌症担忧和痛苦显著减少。
总的来说,在致病性变体状态揭晓后的一段时间内(例如,2周到1个月)的研究结果表明,相对于预测前,错配修复(MMR)致病性变体携带者可能会遭受更多的一般痛苦、癌症特异性痛苦、或癌症担忧。携带者在获得检测结果后通常比未携带家族中先前确定的致病性变体(非携带者)的个体遭受更大的痛苦。
然而,在大多数情况下,在获知检测结果后的一年中,携带者的痛苦水平逐渐降低且与在获知结果后1年时的预测值水平无差异。
这些研究的结果还表明,在结果获知后长达1年,非携带者的痛苦减少或无变化。
一项纳入未受累的患者和CRC患者的研究发现,将携带者与在检测后至1年任何时间点收到信息不足的结果或显示为未知意义的变体的结果的患者相比,两者间痛苦水平无异,且与预测痛苦水平相似。
对林奇综合征遗传咨询和检测后的长期心理社会学结果进行探讨的研究较少。
在对遗传检测前后心理痛苦进行评估的纵向研究发现,致病性变体携带者和非携带者的长期痛苦水平(检测后3年或7年测量)与基线时的痛苦水平相似。
有一个例外:一项研究中非携带者的癌症特异性痛苦评分
在检测后持续下降,显著低于其基线评分,且携带者在检测后1年的评分与3年后观察到的趋势类似。在一项研究中,携带者在检测后7年更可能担忧CRC风险;然而,报告为担忧(即“在某种程度上担忧”或“非常担忧”)CRC的非携带者比报告为不担忧的非携带者更可能怀疑检测结果的真实性。
当被问及对进行检测的决定是否满意时,大多数携带者和非携带者在检测后7年内都非常满意,并表示愿意再次进行检测。
一些研究的结果表明,在检测结果揭晓后可能存在心理痛苦较高风险的患者亚组,包括那些在接受检测前其一般或癌症特异性痛苦的评分值相对较高的患者。
一项通过采血进行林奇综合征检测的CRC患者中开展的研究发现,在女性、年轻人、非白人、那些受正规教育程度较低、对社会支持来源越来越不满意的人群中,抑郁症状和/或焦虑症状的水平较高。
在同一人群中发现了一个表现出较高心理痛苦程度和较低生活质量及社会支持的患者亚组;此外,该亚组更可能担忧发现他们是林奇综合征致病性变体的携带者,以及担忧是否能应对了解其检测结果。
在一份随访报告中,评估了 CRC 患者和有林奇综合征致病性变体风险的亲属在检测结果披露后的心理结果,一个具有相同心理社会特征的亚组,在结果揭晓后一年内,经历了更高水平的一般性痛苦和以及基因检测特异性痛苦,而不管其检测结果如何。与白人和受过高等教育的人相比,非白人和受教育程度较低的人在任何时候的抑郁和焦虑评分都较高。
其他研究也发现,既往有严重或轻微抑郁症病史,癌症特异性痛苦的预测水平较高,一级亲属患癌症的人数较多,情绪低落反应较大,预示在检测结果揭晓后的1到6个月内抑郁程度较高。
然而,在这方面还需要进一步的研究,但个案研究表明,重要的是确定可能有精神痛苦风险的人,并在整个遗传咨询和遗传检测过程中提供心理支持和随访。
研究还考察了林奇综合征遗传咨询和检测对癌症风险理解的影响。一项研究报告称,几乎所有致病性变体的携带者和非携带者都能准确地在检测后1年回忆起检测结果。在揭晓结果后的1个月和1年内,更多的非携带者比携带者正确地识别出他们患CRC的风险。与正确识别其风险水平的携带者相比,错误识别其CRC风险的致病性变体携带者更可能具有较低的检测前主观风险感知水平。
另一项研究报告称,在发现携带者和非携带者的变体状态后,对结直肠癌和子宫内膜癌风险的预估的准确性有所提高。
林奇综合征的遗传咨询和检测的获益包括患者有机会了解癌症早期检测和预防的选择,包括筛查和降低风险的手术。研究表明,许多有林奇综合征风险的人可能在遗传咨询和检测前进行过一些CRC筛查,但大多数人不太可能坚持遵循林奇综合征筛查的建议。在18岁或以上的无结直肠癌病史,并且参与了基于美国的提供林奇综合征遗传咨询和检测的人中,52%至62%的人报告在遗传检测前曾做过结肠镜检查。
在比利时和澳大利亚参与类似研究的癌症未受累者中,分别有51%和68%在研究开始前做过结肠镜检查。
遗传检测前接受结肠镜检查的相关因素包括收入较高和年龄较大、
发展为CRC的感知风险较高、
受教育程度较高,且被告知CRC风险增加。
在一项对符合林奇综合征临床标准的受癌症影响的患者和未受癌症影响患者的研究中,92%的患者报告在遗传检测前至少做过一次结肠镜和/或乙状结肠镜检查。
另一项对未受累患者进行遗传风险评估和可能考虑林奇综合征、FAP或APC I1307K遗传检测的研究报告称,77%的患者至少接受过一次筛查(结肠镜检查、乙状结肠镜检查或钡灌肠检查)。
三项研究确定了遗传检测前癌症未受累者是否坚持林奇综合征结肠镜检查的建议,并报告坚持率为10%,
28%,
和47%。
多项纵向研究检查了癌症未受累者在接受已知林奇综合征致病性变体检测后结肠镜筛查使用情况。
这些研究比较了林奇综合征遗传检测前进行结肠镜检查与获知结果后1年内进行结肠镜检查的情况。一项研究报告称,与非携带者和拒绝接受遗传检测者相比,林奇综合征致病性变体携带者更有可能进行结肠镜检查(73%vs16%vs22%),以及在结果揭晓后1年内携带者结肠镜检查增加(36%vs73%)。
另外两项研究报告显示,在结果揭晓后1年,携带者的结肠镜检率(71%和53%)与检测前相比没有显著差异,
尽管同期非携带者结肠镜检率有所下降。获知结果后1年进行结肠镜检查的因素包括携带林奇综合征-易感致病性变体、
年龄增加、
以及对CRC更强的控制意愿。这些结果表明,在遗传检测结果获知后的1年内,结肠镜检率在致病性变体携带者中增加或保持,而在非携带者中减少。一项纵向研究的数据包括134例MMR致病性变体携带者,这些携带者在接受与林奇综合征相关的癌症诊断后6个月内未接受结肠镜检查,其报告临床显著抑郁症状的可能性是接受遗传检测的6倍,这通过流行病学研究中心抑郁量表(CES-D)测量所得(比值比[OR],6.06;95%CI,2.09–17.59)。在遗传检测前测得的更高水平的CRC担忧也与临床显著的抑郁症状相关(OR,1.53;95%CI,1.19-1.97)。
两项研究基于变体状态,对林奇综合征遗传检测后发布筛查指南的依从性水平进行了探讨。一项研究报告称,在致病性变体携带者中,结肠镜检查的依从率为100%。
另一项研究发现,35%的携带者和13%的非携带者不符合已公布的适用CRC筛查指南;
在这两组中,约一半患者的筛查频率高于公布的指南建议,一半的筛查频率较低。
上述纵向研究对在收到遗传检测结果后的相对较短时间(1年)内的结直肠筛查行为进行了探讨,而对长期筛查行为的了解较少。一项纵向研究(N=73)在基因检测结果揭晓后3年对未受癌症影响者的心理学和行为学结果进行了探讨,发现所有携带者(N=19)在获知结果后1至3年内至少接受了一次结肠镜检查。
一项对检测后7年内发现类似结果的纵向研究还表明,所有携带者都接受了结肠镜检查;大多数(83%)患者每3年或按照建议更频繁地接受一次结肠镜检查,11%的患者报告了更长的筛查间隔。
在这项研究中,那些报告筛查间隔比推荐时间长的人也报告更有可能害怕很快死亡。此外,16%的非携带者报告在检测后7年内进行了结肠镜检查;那些对检测结果真实性表示怀疑的人更有可能进行了结肠镜检查。
在一项研究中,94%的携带者表示打算在未来进行一年或两年一次的结肠镜检查;在非携带者中,64%的人不打算在未来进行结肠镜检查或不确定,33%的人打算每隔5-6年或更短时间进行一次结肠镜检查。
在荷兰开展的一项横断面研究,探讨了在风险评估和咨询后2年至18年期间,患有结直肠癌、子宫内膜癌或临床或遗传诊断为林奇综合征的患者进行乙状结肠镜或结肠镜检查的情况。
根据从医疗记录中获得的数据,86%的林奇综合征致病性变体携带者、68%的未经检测或林奇综合征遗传检测结果信息不足的携带者和73%的林奇综合征临床确诊携带者被认为应遵循筛查的建议。参与者还回答了有关筛查依从性的问题,16%的参与者报告说,他们进行筛查的频率低于推荐的频率。对于整个参与群体而言,更大的筛查感知障碍与通过病历审查确定的筛查不依从性相关,及筛查程序尴尬与自我报告不依从有关。另一项同样在荷兰进行的横断面研究调查了未受癌症影响的林奇综合征变体携带者(n=42)在了解其致病性变体状态(6个月-8.5年)后的结直肠筛查行为。31%的被调查者报告称其在林奇综合征遗传检测前每年进行一次结肠镜检查,88%的被调查者报告称他们在遗传诊断后进行过结肠镜检查(P<0.001)。
部分人对林奇综合征筛查行为知之甚少,这些人可能有种系致病性变异的风险,但其未接受遗传咨询和/或遗传检测来了解其风险状况。在澳大利亚结直肠癌家族登记处的林奇综合征种系致病性变体携带者亲属中,26例未接受遗传咨询和/或检测的患者完成了一次访谈,以评估其在未来10年内发展成CRC的感知风险,及对结肠镜检查状况的自行上报。
其平均感知风险为30.5%,超过了通过MMRpro软件计算的4%的平均预测风险。
73%的人(n=19)报告曾进行过结肠镜检查(1例是出于诊断原因);35%的人在过去2年内进行过结肠镜检查,并被认为应遵循建议。感知风险与最后一次结肠镜检查后的年数(Pearson r,0.49;范围,0.02-0.79)略有正相关,但与其他筛查或个人特征无关。作者的结论是,对于未接受过遗传咨询和/或检测的林奇综合征致病性变体携带者的亲属,单凭感知风险可能不足以预测是否进行结肠镜检查。
多项小型研究已经探讨了子宫内膜癌和卵巢癌患者进行林奇综合征有关的筛查的应用(见表18)。这些研究存在一些局限性,包括样本量小、随访时间短、回顾性设计、依赖自我报告作为数据来源,不包括接受过林奇综合征基因检测的患者。一些研究将不符合接受筛查最低年龄标准的患者纳入筛查应用分析。在对已知家族致病性变体检测结果为阴性后接受的筛查进行评估的研究中,仅很少研究对筛查适应症进行了评估,如既往确定为异常的随访检测。最后,一些研究将积极接受另一种癌症治疗的患者纳入应用分析,这可能会影响提供者的筛查建议。因此,表18仅限于接受过林奇综合征遗传检测、较大样本量、较长随访和分析纳入适当筛查年龄的个体的研究。
研究引文 | 研究人群 | 在遗传咨询和检测前妇科筛查的应用 | 在收到遗传检测结果后妇科筛查的应用 | 随访时长 | 备注 |
---|---|---|---|---|---|
Claes et al. (2005)1,a | 携带者 (n = 7) | 未报告 | TVUS | 1年 | 1名非携带者报告因先前子宫内膜问题接受了TVUS,而3名非携带者报告因预防性原因接受了TVUS。 |
– 携带者 86% (6/7) | |||||
非携带者(n = 16) | |||||
– 非携带者 27% (4/15) | |||||
Collins et al. (2007)1,a | 携带者 (n = 13) | 未报告 | TVUS | 3年 | 4名携带者中有2名在3年的随访评估中行RRH/RRSO。 |
– 携带者 69% (9/13) | |||||
– 非携带者 6% (2/32) | |||||
非携带者(n = 32) | ES | ||||
– 携带者54% (7/13) | |||||
–非携带者 3% (1/32) | |||||
Yurgelun et al. (2012): Cohort 12,a | 77例有林奇综合征相关的EC风险;45例携带者;19例未进行遗传检测,但有林奇综合征相关家族史 | 75%(58/77)参与EC筛查或接受降低EC风险的干预措施;42例行年度TVUS和/或ES;16例行RRH | 未报告 | 不适用(N/A) | |
Yurgelun et al. (2012): Cohort 21,a | 有林奇综合征临床风险的40例女性 | 65%(26/40)坚持EC筛查或接受降低风险的干预措施;6例接受RRH;13例接受年度ES和/或TVU;6例未达到推荐筛查年龄 | 携带者:100%(n=16)坚持EC筛查或降低风险的干预措施;4例接受预防性RRH;5例接受RRH;5例接受EC筛查(TVUS和/或ES);2例未达到推荐筛查年龄 | 1年 | |
携带者(n=16) | |||||
非携带者(n=9);14个不确定的结果;1个意义不明的变体 | 非携带者:11%(1/9)接受EC筛查;11%(1/9)接受RRH筛查 | ||||
EC=子宫内膜癌;ES=子宫内膜采样;RRH=降低风险的经腹全子宫切除术;RRSO=降低风险的输卵管卵巢切除术;TVUS=经阴道超声。 | |||||
非携带者=已知家族致病性变体阴性。 | |||||
1前瞻性研究设计。 | |||||
2回顾性研究设计。 | |||||
a作为数据源的自我报告。 |
总的来说,这些研究纳入了相对较少的女性,并且表明在基因咨询和检测之前,林奇综合征相关妇科癌症的筛查率很低。然而,在参与遗传教育和咨询以及收到林奇综合征致病性变体检测结果后,在携带者中接受妇科癌症筛查的人数普遍增加,而非携带者则减少进行筛查。
关于使用降低风险的结肠切除术治疗林奇综合征还没有达成共识,关于降低风险的结肠切除术治疗林奇综合征的决策和心理后遗症还知之甚少。
在收到阳性检测结果的人群中,有更大比例的人表示有兴趣在获知结果后进行降低风险的结肠切除术。
这项研究还表明,林奇综合征患者对行风险降低手术的考虑可能会促使人们参与遗传检测。在收到结果之前,46%的人表示其正在考虑降低风险的结肠切除术,69%的女性正在考虑降低风险的经腹全子宫切除术(RRH)和降低风险的双侧输卵管卵巢切除术(RRSO);然而,这项研究没有评估人们在收到检测结果后是否真的进行了降低风险的手术。在接受林奇综合征遗传咨询和检测之前,在一项纵向研究中,未受癌症影响的患者有MMR变体风险,其中5%报告称其将考虑结肠切除术,5%的女性表示,如果致病性变体结果阳性,其将进行RRH和RRSO。获知结果后3年,无参与者接受过降低风险的结肠切除术。
两例在遗传检测前接受过RRH的女性在检测后1年内接受了RRSO,
然而,研究中没有其他致病性变体的女性携带者报告在获知检测结果后3年内进行了这两种手术。
一项研究调查林奇综合征患者接受范围较广(结肠次全切除)或范围较小的(部分切除或半结肠切除)切除术后,探讨横断面生活质量和功能结局,总体生活质量结果相当,尽管切除范围越大的患者,其排便次数和相关功能障碍也越频繁。
关于遗传性CRC易感性遗传检测的家族交流,特别是关于这种检测结果的家族交流较复杂。人们普遍认为,在家族内部交流遗传风险信息在很大程度上是家族成员自己的责任。一些研究已经探讨了那些接受过林奇综合征遗传咨询和检测的家族的交流模式。研究的重点是,患者个人是否向其家族成员透露了有关林奇综合征遗传检测的信息,患者将这些信息告知的对象,以及可能促进或阻碍这类交流的家族特征或问题。这些研究调查了健康护理专业人员告知患者林奇综合征易感后,家族内的交流和信息揭晓过程,研究样本相对较小。
研究结果表明,人们通常愿意在其家庭中分享有关林奇综合症致病性变体存在的信息。
分享遗传风险信息的动机包括:希望提高家族对个人风险的认识、促进健康的选择和预测性遗传检测、希望获得情感支持以及认为有道德义务和责任帮助家族中的其他人。
多项研究的结果表明,大多数研究参与者认为,林奇综合征的遗传风险信息在家族内部是公开共享的;然而,这种交流更可能发生在一级亲属(如兄弟姐妹、子女)中,而不是与更多的远亲。
芬兰的一项研究招募了40岁或40岁以上、已知携带MMR致病性变体的父母,以完成一份调查问卷,调查父母如何与成年和未成年子女分享遗传风险知识。研究还发现了交流过程中存在的挑战。
在248名家长中,87%报告称其已经向子女告知了结果。不告知的原因与之前的研究一致(子女年龄小,社会关系疏远或在讨论这个话题时感到困难)。
几乎所有的父母都告诉成年子女其遗传风险和进行遗传检测的可能性,但近三分之一的父母不确定其子女是如何使用这些信息的。家长们认为讨论孩子的癌症风险是交流过程中最困难的方面。在被告知的191名长子女中,69%已经接受了遗传检测。三分之一的家长建议,卫生专业人员应参与信息告知,告知时应在遗传学诊所进行家庭预约。
在告知二级和三级亲属方面,患者可能倾向于采用级联方法;例如,假设一旦向亲属提供有关其家庭患林奇综合症风险的信息,他或她将负责通知其一级亲属。
在告知亲属的子女,特别是未成年子女方面,这种级联的交流方式显然是可取的,而且一致意见认为,不首先通过家族关系等级,就向二级或三级亲属告知这种信息是不适当的。
在一项研究中,接受测试并被发现携带林奇综合征(Lynch syndrome)易感致病性变体的人比收到真正阴性或不具信息性结果的人更可能将其基因检测结果告知至少一名二级或三级亲属。
虽然有关遗传风险的交流通常被视为一个开放的过程,但研究中也报告了一些交流障碍。不告知亲属的原因包括缺乏亲密关系和与患者缺乏联系;事实上,情感上的而非关系上的亲密关系是决定风险交流程度的更重要因素。不想让亲属担忧检测结果信息,以及认为亲属不理解这些信息的含义的认知也被认为是交流障碍。
如果认为有风险的个体太小而无法接受信息(即儿童),如果遗传性癌症风险的信息先前在家庭中造成了冲突,则告知的可能性似乎较小,
或者如果亲属对检测信息不感兴趣。
之前的冲突抑制了关于遗传性癌症风险的讨论,特别是如果这种讨论涉及到坏消息的披露。
对于这些研究的大多数参与者来说,对其家族中的癌症模式归因于林奇综合征-易感致病性变体并不奇怪,
因为有人怀疑家族性癌症的遗传原因,或者有过关于癌症的家族讨论。家族中林奇综合征-易感致病性变体的确定被认为是私事,但不一定是秘密,
许多人与非本家族的人讨论了家族的致病性变体状态。了解林奇综合征-易感致病性变体的检测并不羞愧,尽管个人对这一信息对保险歧视的潜在影响表示担忧。
此外,虽然可能有人愿意告知家族中存在致病性变体的信息,但一项研究表明,在告知个人结果方面,有一种倾向为维持更多隐私,将个人结果与家族风险信息区分开来。
在少数情况下,个人报告称,其亲属在收到有关家庭林奇综合症风险的消息后表示愤怒、震惊或其他负面情绪反应;
然而,大多数人在告知亲属方面几乎没有任何困难。
有人认为,那些对与癌症相关的讨论更自在、更开放的家庭可能接受性更强,更可能接受有关遗传风险的消息。
在某些情况下,先证者报告称其特别有义务告知家庭成员遗传性癌症的风险
他们经常是鼓励家庭成员接受遗传咨询和检测家族致病性变体的最有力倡导者。
在传播遗传性癌症风险信息方面,也出现了一些性别和家庭角色的差异。一项研究报告说,女性先证者比男性先证者更愿意讨论遗传信息,男性先证者在家庭交流过程中更需要专业支持。
另一项研究表明,母亲在传达健康风险信息方面可能是家庭网络中特别有影响力的成员。
与先证者和其他已经接受过遗传检测的高危人群相比,致病性变体阴性的人、选择不进行检测的人以及高危人群配偶报告称他们没有亲身参与风险交流过程。
各种通信方式(例如,面谈、电话或书面联系)通常可用于告知家族的遗传风险信息。
在一项研究中,诸如遗传咨询总结信或林奇综合征小册子等交流辅助工具被视为交流过程中的有益辅助工具,但并不被认为是交流成功的核心或必要因素。
研究表明,医护人员建议告知亲属遗传性癌症风险可能会鼓励人们就林奇综合征进行交流
以及卫生保健专业人员的支持可能有助于克服向家族成员传播此类信息的障碍。
到目前为止,许多关于家族交流的文献都将关注点置于检测结果的告知上;然而,目前正在探讨家族交流的其他要素。一项研究评估了老年家族成员对林奇综合征患者及其家族成员(来自33个家族的206名受访者)提供各类支持(如工具性、情感性、危机帮助和必要时的可靠性)的作用。
受访者完成了关于其家族社交网络(血缘和非血缘亲属以及家族以外的其他人)和家族内部交流模式的访谈。受访者中位年龄与其家族社交网络成员的中位年龄没有差异(43岁)。研究发现,23%的家族社交网络成员进行CRC筛查(其他类型的支持,如社会支持,被更频繁报告)。那些鼓励筛查的人是老年人、女性和重要的其他人或血缘上的家族成员,而不是非家族成员。鉴于许多家族社交网络的成员并不住在同一个家庭,研究指出了整个家族在鼓励和支持筛查方面的重要性。
FAP遗传检测的应用可能高于林奇综合征检测。一项在美国有FAP风险的无症状患者中进行的研究发现,82%的成年人和95%的未成年人接受了遗传检测。
据报道,英国的应用率接近100%。
对于APC遗传检测率应用较高的一个可能解释是,APC遗传检测比每年的内镜检查更具成本效益,
且可以免除每年必须在青春期前进行的筛查的负担。有机会消除对降低风险的手术的担忧是FAP遗传检测的另一个可能的获益。进行APC遗传检测的决定可视为医疗管理决策;
对FAP和其他遗传性癌症综合征,潜在心理社会因素可能影响检测结果的研究并不充分。APC致病性变体的高外显率、较早发病和明确的表型也可能影响对这种情况进行遗传检测的决定,这可能是因为对该疾病有更多认识及多个受累的家族成员有更多的经验。
FAP遗传检测目前提供给其父母受累的儿童,通常在10到12岁,推荐在此年龄进行内镜筛查。由于在18岁之前诊断FAP是预防CRC的最佳方法,而且由于在确定个人是否为APC致病性变体携带者时,筛查和可能的手术是必要的,因此在这种情况下,对未成年人进行遗传检测是合理的。(了解有关儿童遗传检测的伦理、心理和遗传咨询问题的更详细讨论,请参阅PDQ癌症遗传风险评估和咨询总结中的儿童检测部分。)
在荷兰对患有FAP的家族成员进行的一项调查中,三分之一(34%)的人认为,向12岁以前的儿童提供APC遗传检测最为合适,而38%的人更愿意向12岁至16岁的儿童提供APC遗传检测,该年龄的儿童将能够更好地了解DNA检测过程。只有4%的人认为儿童根本不应该接受DNA检测。
来自28位被确诊为FAP的美国父母的定性访谈数据结果显示,61%的人倾向于在他们的高危子女(10-17岁)中进行APC变体的遗传检测;71%的人认为其子女应该收到其检测结果。父母选择对孩子进行检测的主要原因包括早期检测和管理、减少父母的焦虑和不确定性,并帮助作出有关监测的决策。未进行检测的原因集中在认知度和成本问题。
临床观察表明,家庭成员受到FAP影响的儿童非常清楚降低风险的手术可能性,并将检测结果作为决定是否需要进行FAP手术的因素。
就儿童的年龄、发育问题和对FAP的心理担忧而言,重要的是要考虑向儿童告知基因检测结果的时机。携带APC致病性变体的儿童已经表达了对同伴如何看待他们的担忧,并可能会从帮助他人制定保持自尊的解释中获益。
对FAP遗传检测后的心理结果进行评估的研究表明,部分患者,特别是致病性变体携带者,可能面临更大的痛苦。在一项对先前接受过APC遗传检测的成年人进行的横断面研究中,那些携带致病性变体的成年人表现出比非携带者更高的焦虑水平,并且更有可能表现出临床显著的焦虑水平。
在这项研究中,较低的乐观度和较低的自尊性与其较高的焦虑度有关,
对于携带者,FAP相关的焦虑、FAP的严重性、对遗传检测准确性的信任度与较多的焦虑相关。
然而,在先前的一项研究中,比较了接受过FAP、亨廷顿病和遗传性乳腺癌/卵巢癌综合征遗传检测的成年人,在揭晓阳性或阴性检测结果后的1周内FAP特异性痛苦有所升高,总体上低于其他综合征。
在一项澳大利亚横断面研究中,研究对象为18岁至35岁确诊为FAP(N=88)的年轻人,其最常报告以下与FAP相关的问题,他们认为需要中等到高水平的支持或帮助:担忧子女患FAP的风险,对癌症发展的恐惧,以及对FAP影响的不确定性。
75%的人表示他们会考虑进行FAP的产前检测;61%的人会考虑PGT,61%的人更希望他们的孩子在出生时或10岁之前接受遗传检测。有一小部分受访者(16%)报告称遇到了与FAP相关的歧视,主要指照顾其医疗或自理需要(例如,筛查的下班时间、频繁上厕所的需要和身体限制)可能会在同事和管理者中产生消极态度。
在荷兰对FAP家族进行的另一项大规模横断面研究纳入了16至84岁的人,他们或者是FAP确诊,或者有50%的风险存在APC致病性变体,或者被证明是APC非携带者。
在接受过APC检测的人中,48%的人在这项研究之前至少做过5年或更长时间的APC检测。在诊断为FAP的患者中,76%的患者接受过预防性结肠切除术,78%的患者处于术后至少5年。这项研究评估了广泛性心理痛苦,特别是与FAP相关的痛苦,以及与癌症相关的担忧的患病率。5项心理健康指数(SF-36的一个子量表,用来评估广泛性痛苦)的平均得分与荷兰普通人群相当。根据事件影响量表(IES)的测量结果,20%的被调查者具有中度到高度FAP特异性痛苦,其中23%的被诊断为FAP,11%的有FAP风险,17%的非携带者报告得分在该范围内。5%的IES报告分数显示严重的和与临床有关的痛苦;其中,大多数(78%)确诊为FAP。总的来说,癌症担忧量表的平均得分与另一项针对林奇综合征家庭的研究结果相当。确诊为FAP的人更容易产生较为频繁的癌症相关担忧,最常见的担忧是可能需要进行额外的手术(26%)和自己(17%)或家族成员(14%)可能会发展成癌症。在多因素分析中,与FAP特异性痛苦水平较高相关的因素包括发展为癌症、与家人或朋友更频繁地讨论FAP以及没有子女。与癌症相关的焦虑水平较高的因素包括性别为女性、较差的家庭功能、与家人或朋友就FAP进行较多实际和期望的讨论、较高的癌症风险感知、较差的一般健康认知,以及曾经是癌症家族成员的照顾者。作者指出,大多数与癌症和FAP相关的因素是心理社会因素,而不是临床或人口统计学因素。
在荷兰进行的另一项横断面研究发现,在FAP患者中,37%的人表示疾病影响了其生育意愿(即想要更少的孩子或不想要孩子)。33%的人表示他们会由于FAP而考虑产前诊断(PND);30%的人会考虑胚胎植入前遗传学检测(PGT)。较高的负罪感和对终止妊娠更积极的态度与对PND和PGT更感兴趣相关。
在另一项美国研究中,愿意考虑产前检查的预测因素包括有一个受影响的子女和一级亲属继发于FAP死亡。
在FAP遗传检测中,接受检测的儿童的心理脆弱性尤其令人关注。研究结果表明,大多数儿童在APC检测后没有出现临床意义上的心理痛苦。然而,在涉及成年人的研究中,成人亚组可能更容易遭受更大的痛苦,并将从持续的心理支持中获益。一项对接受FAP遗传检测的儿童进行的研究发现,遗传咨询和检测结果揭晓后,他们的情绪和行为仍保持正常。家庭状况的各个方面,包括母亲或兄弟姐妹的疾病与亚临床抑郁症状的增加有关。
在对48名接受FAP检测的儿童进行的长期随访研究中,大多数儿童没有遭受心理痛苦;但是,有一小部分接受检测的儿童表现出临床上显著的检测后痛苦。
另一项研究发现,尽管APC致病性变体阳性儿童在得知结果后对发生疾病的感知风险增加,但在得知结果后一年内,焦虑和抑郁水平保持不变。
本研究中致病性变体阴性的儿童在同一时间段内遭受的焦虑较少,且自尊有所提高。
对FAP筛查的心理方面知之甚少。一项对少数有FAP家族史的人(17-53岁)进行的研究发现,在那些无症状的人中,所有人都报告在参与研究之前接受了至少一次内镜检查。
只有33%(6例患者中的2例)报告在推荐的时间间隔内继续筛查。在接受结肠切除术的患者中,92%(12例患者中的1例)坚持推荐的结直肠监测。在一项对150例临床或遗传诊断为典型FAP或衰减FAP(AFAP)的患者及其高危亲属的横断面研究中,52%的FAP患者和46%的FAP高危亲属接受了推荐的内镜检查。
在确诊或AFAP高危人群中,分别有58%和33%接受过筛查。与在建议的时间间隔内接受筛查的人相比,未接受筛查的人不太可能遵循医疗保健提供者的筛查建议,较可能缺乏医疗保险或筛查保险报销,更可能认为其患CRC的风险不会增加。只有42%的研究人群曾接受过遗传咨询。一小部分参与者(14%-19%)将筛查描述为“必要的恶行”,表示不喜欢肠道准备,或经历过疼痛和不适。19%的人报告说,这些问题可能会对未来的内镜检查造成困难。19%的人报告说,改进的技术和麻醉的使用提高了对筛查程序的耐受性。
当FAP高危者发展成多发性息肉时,结肠次全切除术或全结肠直肠切除术是降低CRC风险的唯一有效方法。大多数FAP患者可以避免永久性造口,保留肛门和/或直肠,一定程度上可以排便。(更多关于FAP手术管理程序的信息,请参阅本总结的FAP干预措施部分。)这些干预措施的生活质量结果的证据不断积累,总结参见表19。
人群 | 随访时长 | 手术类型 | 排便次数 | 控制大便 | 身体意象 | 性功能 | 备注 |
---|---|---|---|---|---|---|---|
279例接受结肠切除术后FAP受累的个体(135例女性和144例男性);对照组为来自荷兰普通人群的1771例个体 | IRA平均: 12年 (SD, 7.5年) | IRA: n = 161 | 未评估 | 未评估 | EORTC QLQ-CR38 a | EORTC QLQ-CR38 a | 所有子量表的SF-36b评分(荷兰语版)均显著低于一般人群(IRA:P<0.001;IPAA:P<0.001)。 |
IRA: 87.5 (SD, 21.9) | IRA: 38.9 (SD, 26.6) | ||||||
IPAA平均值: 6.8年(SD, 4.9年) | IPAA: n = 118 | IPAA: 84.4 (SD, 22.7) | IPAA: 42.2 (SD, 26.3) | ||||
88例澳大利亚患者(63例女性和25名男性),年龄18-35岁,其中57例是结肠切除术后患者,14例是未行手术的FAP患者 | 未报告 | IRA: n = 33 | 未评估 | 未评估 | SF-36 b | SF-36 b | |
IPAA: n = 21 | IRA: 89.9 (SD, 16.1) | IRA: 86.2 (SD, 21.6) | |||||
回肠造口术: n = 1 | IPAA: 72.1 (SD, 23) | IPAA: 77.5 (SD, 26.2) | |||||
未知手术类型:n=2 | 未行手术:94.1(SD,9.4) | 未行手术:91(SD,19) | |||||
525例患者(283例女性和242例男性),其中296例是结肠切除术后患者,45例未行手术的FAP患者,50例未行手术的FAP高危患者,134例非携带者 | Range: 0–1年to >10年 | IRA: n = 136 | 未评估 | 未评估 | EORTC QLQ-CR38 a | EORTC QLQ-CR38 a | 41%FAP患者报告职业中断: |
行结肠切除术后: 85.4 (SD, 20.5) | 行结肠切除术后: 42.2 (SD, 23.2) | 部分或完全丧失能力: n = 73 (59%) | |||||
IPAA: n = 112 | 未行FAP的患者: 91.9 (SD, 16.1) | 行结肠切除术后: 42.2 (SD, 23.2) | 进行较少: n = 30 (24%) | ||||
回肠造口术: n = 42 | 高危: 94.0 (SD, 13.1) | 高危: 47.6 (SD, 23.7) | 进行更多 n = 5 (4%) | ||||
其他: n = 6 | 非携带者: 92.3 (SD, 13.1) | 非携带者r: 45.7 (SD, 21.2) | 不同时期或多或少进行:n=16(13%) | ||||
209名年龄在18-75岁结肠切除术后的瑞典FAP患者(116名女性和93名男性) | 上次手术后的平均时间:14年(SD,10;范围,1-50年) | IRA: n = 71 | 未评估 | 白天: 71% (n = 149) | 未评估 | 未评估 | 评估的21种腹部症状的平均数量为7(SD,4.61;范围,1-18)。女性报告的症状比男性多,但在症状困扰的程度上,性别之间没有差异。较高的症状数是身心健康较差的独立预测因子。 |
IPAA: n = 82 | |||||||
回肠造口术: n = 39 | 夜间: 61% (n = 128) | ||||||
可控性回肠造口术: n = 14 | |||||||
其他: n = 3 | |||||||
28例年龄在14岁或14岁以下接受结肠切除术的患者(10例女性和18例男性) | 12年(SD, 8.4; 范围, 1–37年) | IRA: n = 7 | 白天: | 白天: | 罗森伯格自尊量表 : 25.53/30 c | 未评估 | 10/28报告了结肠切除术后的癌症相关担忧,有一种趋势,即年轻(<18岁)与更多的癌症相关担忧相关。 |
IRA: 3.8 (SD, 1.5) | IRA: 71.4% (n = 7) | ||||||
IPAA: 5.3 (SD, 2.4) | IPAA: 85.7% (n = 21) | ||||||
IPAA: n = 21 | 夜间: | 夜间: | |||||
IRA: 1.3 (SD, 0.6) | IRA: 50.0% (n = 7) | ||||||
IPAA: 1.3 (SD, 0.5) | IPAA: 61.9% (n = 21) | ||||||
EORTC QLQ=欧洲癌症研究和治疗组织制定的结直肠癌生活质量问卷;IPAA=回肠储袋肛管吻合术;IRA=回肠直肠吻合术;SD=标准差;SF-36=简明(36)健康状况调查问卷。 | |||||||
aEORTC QLQ-C38评分范围为0-100。功能量表:0=最低功能水平,100=最高/健康功能水平。症状量表:0=最低症状水平,100=最高症状水平。 | |||||||
bSF-36评分范围为0-100,0表示可能的最低健康状况,100表示可能的最佳健康状况。 | |||||||
c在同一年龄组的正常范围内。 |
对FAP的风险降低的手术的研究发现,一般的生活质量指标都在正常范围内,而且大多数报告对其身体意象无负面影响。然而,这些研究表明,降低FAP风险的手术可能对至少部分患者的生活质量产生负面影响。
目前正在进行化学预防试验,以评估各种疗法对林奇综合征和FAP高危人群的疗效。
在被邀请参加一项为期5年的试验以评估维生素和纤维对腺瘤性息肉发展的影响的FAP患者样本中,55%的人同意参加。
参与者更倾向于较年轻,最近被确诊为FAP,并且居住在离试验中心较远的地方,但在任何其他心理社会变量上与非参与者没有差异。
致病性变体可能遗传给子女,可能引发受遗传性CRC综合征影响的家族的担忧,以至于一定程度上部分携带者可能避免生育。这些担忧也可能促使人们考虑使用产前诊断(PND)方法来帮助降低遗传风险。PND是一个包含广义的术语,指为评估胎儿是否存在遗传性疾病而进行的任何医疗操作。方法包括羊水穿刺和绒毛采样。
两种手术均有流产的低风险。
此外,发现胎儿是癌症易感性变体的携带者,可能会给夫妇带来继续妊娠或终止妊娠的艰难决定,可能需要额外的专业咨询和支持。
这些检测的替代方法是胚胎植入前基因检测(PGT),一种用于在子宫植入前检测受精胚胎是否存在遗传性疾病的操作。
利用遗传检测获得的信息,准父母可以决定是否植入。PGT可用于检测遗传性癌症易感基因的致病性变体,包括APC。
从目前发表的有限研究来看,人们似乎对ART用于FAP、林奇综合征和PJS中很感兴趣。
然而,实际应用率尚未报告。
研究人群 | N c | 对ART的兴趣或意图 | 备注 |
---|---|---|---|
FAP受累的患者 | 20 | 95%(19/20)的患者会考虑FAP的产前GT;90%(18/20)的患者会考虑PGT;75%(15/20)的患者会考虑羊膜穿刺或绒毛采样 | |
FAP受累的患者 | 341 | 33%(16/64)的患者由于FAP会考虑进行PND;30%(76/256)考虑PGT;15%(52/341)的患者由于FAP会考虑终止妊娠 | 分别有24%和25%的患者没有回答对PND和PGT的态度。 |
与FAP相关的APC致病性变体患者 | 65 d | 25%(16/64)的患者了解PGT;78%(50/64)的患者认为应该提供PGT;55%(31/56)的患者将考虑PGT | |
接受林奇综合征遗传检测的患者 | 48 e | 21%(10/48)的患者考虑PND和/或PGT;19%(9/48)仅考虑PND;2%(1/48)仅考虑PGT | 在GT结果公布后1年,9名携带者中有2人报告称他们正在考虑PGT用于未来的妊娠。 |
林奇综合征致病性变体的携带者 | 43 f | 19%(8/42)的携带者了解PGT;69%(29/42)的携带者认为应该提供PGT;41%(16/39)的携带者会考虑PGT | |
PJS受累的患者 | 52 | 15%(8/52)表示,如果PND确定胎儿患有PJS,则终止妊娠是可接受的;52%(27/52)表示,若为PJS患者,则可接受PGT | 10(19%)例患者,其中9例是女性,报告称其由于PJS决定不进行生育。 |
GT=遗传检测;PGT=胚胎植入前基因检测;PND=产前诊断。 | |||
a采用横截面设计进行的研究,研究点在美国和荷兰。 | |||
b在林奇综合征的临床遗传检测之前以及在遗传检测结果揭晓后的3个月和1年,参与者受邀完成问卷调查。 | |||
c表示18岁以上的参与者人数,除非另有规定。 | |||
d指具有APC致病性变体的患者总数。并非所有人对每个问题进行了回答或并非所有人都有资格回答每个问题。 | |||
e表示在基因检测前回答问卷的130名个体中,正在考虑将来生育的人数。 | |||
f携带林奇综合征致病性变体的患者总数。并非所有人对每个问题进行了回答或并非所有人都有资格回答每个问题。 |
Psychosocial research in cancer genetic counseling and testing focuses on the interest in testing among populations at varying levels of disease risk, psychological outcomes, interpersonal and familial effects, and cultural and community reactions. This research also identifies behavioral factors that encourage or impede surveillance and other health behaviors. Data resulting from psychosocial research can guide clinician interactions with patients and may include the following:
This section of the summary will focus on psychosocial aspects of genetic counseling and testing for Lynch syndrome, familial adenomatous polyposis (FAP), and Peutz-Jeghers syndrome (PJS), including issues surrounding medical screening, risk-reducing surgery, and chemoprevention for these syndromes.
Early studies that evaluated the uptake of genetic counseling and testing focused on selected, high-risk research populations, including colorectal cancer (CRC) patients and unaffected family members identified at high risk of CRC largely based on family history. The participants were recruited mainly from clinical settings and familial colon cancer registries. Most studies recruited index cancer cases, typically CRCs, specifically to offer genetic counseling and germline testing for mismatch repair (MMR) variants; these were frequently offered as free services.
Counseling and testing were similarly offered to relatives of index cases with pathogenic variants. A review that summarized these early studies reported a wide range of testing uptake rates, from 14% to 75%, and included uptake among both index cases and at-risk relatives who were offered testing.
The review indicated that the primary reasons for undergoing genetic testing included a desire to learn about children’s risk and to learn about early detection and screening needs, as well as a reduction in uncertainty. Reasons for declining testing included cost, insurance discrimination concerns, potential adverse emotional effects for oneself or one’s family, low anticipated benefit, and lack of time.
While these early studies of genetic testing uptake offered preliminary insight regarding why individuals may or may not be motivated to have testing, the process for offering genetic counseling and testing differed from what has evolved into current clinical practice. Clinical practice relies less solely on family history to identify individuals who may benefit from testing, and instead utilizes universal molecular diagnostic testing of CRC and endometrial cancer tumors in newly diagnosed patients using MSI and/or IHC as an initial screen for Lynch syndrome. (Refer to the Universal tumor testing to screen for Lynch syndrome section of this summary for more information.)
While universal MSI/IHC screening is increasingly being adopted to identify newly diagnosed patients who may have a germline variant, an important implication is that not all individuals who may be appropriate for germline testing follow through with recommended genetic counseling and testing services. Two reports from a single institution found that 20% and 13% of CRC and endometrial cancer index cases, respectively, with abnormal IHC results followed through with germline variant testing for Lynch syndrome.
These studies did not solicit reasons for follow through with genetic counseling and testing. However, it has been suggested that higher levels of patient completion of genetic testing after abnormal MSI/IHC results may be associated with having genetic counselors involved in this process to disclose screen-positive results, provide counseling after MSI/IHC testing, or facilitate referrals.
In a study of 145 patients with CRC in the Kaiser Permanente Northwest health care system who were surveyed before receiving their MSI results, most patients had a positive attitude toward MSI/IHC screening.
The majority (84.8%) endorsed six or more benefits of MSI/IHC screening; however, 89.4% also endorsed fewer than four potential barriers, primarily the cost of additional testing and surveillance. Patients with stronger family histories of cancer were more likely to cite fewer barriers of MSI/IHC screening. Patients also experienced minimal distress associated with the screening, with 77.2% of participants having a score of zero (indicating no distress).
Education regarding family history and cancer risk and encouragement to have testing from health care providers may facilitate uptake of genetic counseling and testing. A small (n = 19) qualitative study of newly diagnosed patients with CRC who met high-risk criteria for referral to cancer genetics risk assessment and counseling identified potential reasons why patients may not seek counseling as recommended. These reasons included incomplete knowledge of family cancer history and not realizing the relevance of family history to their personal cancer diagnosis; lack of a specific, direct physician’s recommendation for counseling; and viewing counseling as a lower priority than coping with the immediate demands of a new cancer diagnosis.
In a follow-up survey of 91 individuals in a randomized trial to promote colonoscopy screening in those at risk for Lynch syndrome, only 24% reported ever having discussed genetic testing with their physicians, and the most common barrier to undergoing testing was lack of advice to do so by a health care provider.
There is increasing adoption of universal screening of newly diagnosed tumors for Lynch syndrome in clinical practice. However, the clinical benefit and cost-effectiveness of this process have been attributed to uptake of cascade screening, or predictive testing among at-risk relatives of index cancer cases who are found to have a pathogenic germline variant. A systematic review evaluated the frequency and predictors of genetic testing uptake by first-degree relatives (FDRs) of index cases with Lynch syndrome.
Among four studies that were included in the review and reported uptake rates among FDRs, results showed that 34% to 52% of FDRs had undergone testing. Factors associated with testing uptake in relatives included age (<50 y), female sex, parenthood, employment status, level of education, participation in medical research, psychological factors (lack of depressive symptoms), and the number of relatives affected with cancer.
A large retrospective study of genetic testing uptake across three generations of Finnish families enrolled in a Lynch syndrome registry also found an incomplete uptake of predictive testing among at-risk relatives of individuals with pathogenic variants, and a decreasing uptake rate by generation.
Among 1,184 probands with a Lynch syndrome variant, 67%, 43%, and 24% of at-risk adult first-, second-, and third-generation relatives, respectively, had predictive testing. Among 539 first-generation Lynch syndrome variant carriers, 62% of their at-risk adult children underwent testing. In multivariate analysis, older age, family-specific variant (MLH1 and MSH2 vs. MSH6), being an only child or having a sibling with a pathogenic variant, and having a parent who adhered to colonoscopy surveillance were associated with predictive testing uptake. This study suggested that family-level factors such as predictive testing and screening behavior may influence predictive testing among at-risk relatives of individuals with Lynch syndrome–associated variants.
Published reports of interventions to increase uptake of cascade screening in Lynch syndrome families are limited. An Australian paper compared two approaches for informing at-risk relatives about pathogenic variants for hereditary cancers, including Lynch syndrome.
In this study, index cases from 33 kindreds who had undergone genetic testing provided consent for their clinicians to send detailed letters to at-risk relatives advising them about the identification of an inherited cancer predisposition in the family. Letters also included a recommendation to discuss the information with a physician or genetics specialist, and provided information about what a genetics evaluation comprised. Within the first 2 years of follow-up, 40% of first- and second-degree relatives had had predictive genetic testing, were determined to be presumed noncarriers, or had undergone evaluation but declined genetic testing. The authors compared these findings with a cohort of 41 kindreds seen prior to the initiation of the clinician-generated letters, of whom variant-positive index cases had only been asked to advise relatives that genetic testing was available. In the earlier cohort, 23% of at-risk relatives had sought services to clarify their genetic risk status, which was significantly fewer compared with the group receiving clinician-generated letters (P = .001). Receipt of the letters did not generate concerns about a breach of privacy or autonomy.
Refer to the Ethical, Legal, and Social Implications section in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for information about ethical concerns, including duty to warn.
Studies have examined the psychological status of individuals before, during, and after genetic counseling and testing for Lynch syndrome. Some studies have included only persons with no personal history of any Lynch syndrome–associated cancers,
and others have included both CRC patients and cancer-unaffected persons who are at risk of having a Lynch syndrome pathogenic variant.
Cross-sectional evaluations of the psychosocial characteristics of individuals undergoing Lynch syndrome genetic counseling and testing have indicated that mean pretest scores of psychological functioning for most participants are within normal limits,
although one study comparing affected and unaffected individuals showed that affected individuals had greater distress and worry associated with Lynch syndrome.
Several longitudinal studies have evaluated psychological outcomes before genetic counseling and testing for Lynch syndrome and at multiple time periods in the year after disclosure of test results. One study examined changes in anxiety based on personal cancer history, gender, and age (younger than 50 y vs. older than 50 y) before and 2 weeks after a pretest genetic-counseling session. Affected and unaffected female participants in both age groups and affected men older than 50 years showed significant decreases in anxiety over time. Unaffected men younger than 50 years maintained low levels of anxiety; however, affected men younger than 50 years showed no reductions in the anxiety levels reported at the time of pretest counseling.
A study that evaluated psychological distress 8 weeks postcounseling (before disclosure of test results) among both affected and unaffected individuals found a significant reduction in general anxiety, cancer worry, and distress.
In general, findings from studies within the time period immediately after disclosure of pathogenic variant status (e.g., 2 weeks to 1 month) suggested that carriers of mismatch repair (MMR) pathogenic variants may experience increased general distress, cancer-specific distress, or cancer worries relative to their pretest measurements. Carriers often experienced significantly higher distress after disclosure of test results than do individuals who do not carry a pathogenic variant previously identified in the family (noncarrier).
However, in most cases, carriers’ distress levels subsided during the course of the year after disclosure and did not differ from pretest distress levels at 1 year postdisclosure.
Findings from these studies also indicated that noncarriers experienced a reduction or no change in distress up to 1 year after results disclosure.
A study that included unaffected individuals and CRC patients found that distress levels among patients did not differ between carriers and individuals who received results that were uninformative or showed a variant of unknown significance at any point up to 1 year posttest and were similar compared with pretest distress levels.
A limited number of studies have examined longer-term psychosocial outcomes after Lynch syndrome genetic counseling and testing.
Longitudinal studies that evaluated psychological distress before and after genetic testing found that long-term distress levels (measured at 3 or 7 years posttesting) among carriers and noncarriers of pathogenic variants were similar to distress levels at baseline.
with one exception: noncarriers’ cancer-specific distress scores in one study
showed a sustained decrease posttesting and were significantly lower than their baseline scores and with carriers’ scores at 1 year posttesting, with a similar trend observed at 3 years posttesting. In one study, carriers were more likely to be worried about CRC risk at 7 years posttesting; however, noncarriers who reported worry about CRC (i.e., “worried to some extent” or “very worried”) were more likely to doubt the validity of their test result than were noncarriers who reported no worry.
When asked about their satisfaction with the decision to have testing, the majority of carriers and noncarriers were extremely satisfied up to 7 years posttesting and indicated they would be willing to undergo testing again.
Findings from some studies suggested that there may be subgroups of individuals at higher risk of psychological distress after disclosure of test results, including those who present with relatively higher scores on measures of general or cancer-specific distress before undergoing testing.
A study of CRC patients who had donated blood for Lynch syndrome testing found that higher levels of depressive symptoms and/or anxiety were found among women, younger persons, nonwhites, and those with less formal education and fewer and less satisfactory sources of social support.
A subgroup of individuals who showed higher levels of psychological distress and lower quality of life and social support were identified from the same population; in addition, this subgroup was more likely to worry about finding out that they were carriers of Lynch syndrome pathogenic variants and being able to cope with learning their test results.
In a follow-up report that evaluated psychological outcomes after the disclosure of test results among CRC patients and relatives at risk of having a Lynch syndrome pathogenic variant, a subgroup with the same psychosocial characteristics experienced higher levels of general distress and distress specific to the experience of having genetic testing within the year after disclosure, regardless of variant status. Nonwhites and those with lower education had higher levels of depression and anxiety scores at all times compared with whites and those with higher education, respectively.
Other studies have also found that a prior history of major or minor depression, higher pretest levels of cancer-specific distress, having a greater number of cancer-affected first-degree relatives, greater grief reactions, and greater emotional illness–related representations predicted higher levels of distress from 1 to 6 months after disclosure of test results.
While further research is needed in this area, case studies indicate that it is important to identify persons who may be at risk of experiencing psychiatric distress and to provide psychological support and follow-up throughout the genetic counseling and genetic testing process.
Studies also have examined the effect of Lynch syndrome genetic counseling and testing on cancer risk comprehension. One study reported that nearly all carriers and noncarriers of pathogenic variants could accurately recall the test result 1 year after disclosure. More noncarriers than carriers correctly identified their risk of developing CRC at both 1 month and 1 year after result disclosure. Carriers of pathogenic variants who incorrectly identified their CRC risk were more likely to have had lower levels of pretest subjective risk perception compared with those who correctly identified their level of risk.
Another study reported that accuracy of estimating colorectal and endometrial cancer risk improved after disclosure of variant status in carriers and noncarriers.
Benefits of genetic counseling and testing for Lynch syndrome include the opportunity for individuals to learn about options for the early detection and prevention of cancer, including screening and risk-reducing surgery. Studies suggest that many persons at risk of Lynch syndrome may have had some CRC screening before genetic counseling and testing, but most are not likely to adhere to Lynch syndrome screening recommendations. Among persons aged 18 years or older who did not have a personal history of CRC and who participated in U.S.-based research protocols offering genetic counseling and testing for Lynch syndrome, between 52% and 62% reported ever having had a colonoscopy before genetic testing.
Among cancer-unaffected persons who participated in similar research in Belgium and Australia, 51% and 68%, respectively, had ever had a colonoscopy before study entry.
Factors associated with ever having a colonoscopy before genetic testing included higher income and older age,
higher perceived risk of developing CRC,
higher education level, and being informed of increased risk of CRC.
In a study of cancer-affected and cancer-unaffected persons who fulfilled clinical criteria for Lynch syndrome, 92% reported having had a colonoscopy and/or flexible sigmoidoscopy at least once before genetic testing.
Another study of unaffected individuals presenting for genetic risk assessment and possible consideration of Lynch syndrome, FAP, or APC I1307K genetic testing reported that 77% had undergone at least one screening exam (either colonoscopy, flexible sigmoidoscopy, or barium enema).
Three studies determined whether cancer-unaffected persons adhered to Lynch syndrome colonoscopy screening recommendations before genetic testing, and reported adherence rates of 10%,
28%,
and 47%.
Several longitudinal studies examined the use of screening colonoscopy by cancer-unaffected persons after undergoing testing for a known Lynch syndrome pathogenic variant.
These studies compared colonoscopy use before Lynch syndrome genetic testing with colonoscopy use within 1 year after disclosure of test results. One study reported that carriers of Lynch syndrome pathogenic variants were more likely to have a colonoscopy than were noncarriers and those who declined testing (73% vs. 16% vs. 22%) and that colonoscopy use increased among carriers (36% vs. 73%) in the year after disclosure of results.
Two other studies reported that carriers’ colonoscopy rates at 1 year after disclosure of results (71% and 53%) were not significantly different from rates before testing,
although noncarriers’ colonoscopy rates decreased in the same time period. Factors associated with colonoscopy use at 1 year after disclosure of results included carrying a Lynch syndrome–predisposing pathogenic variant,
older age,
and greater perceived control over CRC. These findings suggest that colonoscopy rates increase or are maintained among carriers of pathogenic variants within the year after disclosure of results and that rates decrease among noncarriers. Data from a longitudinal study including 134 carriers of MMR pathogenic variants with and without a prior Lynch syndrome–related cancer diagnosis found that those who did not undergo colonoscopy for surveillance within 6 months after receiving genetic test results were six times more likely to report clinically significant depressive symptoms as measured by the Center for Epidemiological Studies-Depression (CES-D) scale (odds ratio [OR], 6.06; 95% confidence interval [CI], 2.09–17.59). Higher levels of CRC worry measured before genetic testing also were associated with clinically significant depressive symptoms (OR, 1.53; 95% CI, 1.19–1.97).
Two studies examined the level of adherence to published screening guidelines after Lynch syndrome genetic testing, based on variant status. One study reported a colonoscopy adherence rate of 100% among carriers of pathogenic variants.
Another study found that 35% of carriers and 13% of noncarriers did not adhere to published guidelines for appropriate CRC screening;
in both groups, about one-half screened more frequently than published guidelines recommend, and one-half screened less frequently.
The longitudinal studies described above examined colorectal screening behavior within a relatively short period of time (1 year) after receiving genetic test results, and less is known about longer-term use of screening behaviors. A longitudinal study (N = 73) that examined psychological and behavioral outcomes among cancer-unaffected persons at 3 years after disclosure of genetic test results found that all carriers (n = 19) had undergone at least one colonoscopy between 1 and 3 years postdisclosure.
A longitudinal study of similar outcomes up to 7 years posttesting also found that all carriers had undergone colonoscopy; most (83%) underwent the procedure every 3 years or more frequently as recommended, and 11% reported longer screening intervals.
In this study, those who reported longer screening intervals than recommended also were more likely to report a fear of dying soon. Also, 16% of noncarriers reported undergoing colonoscopy within the 7 years posttesting; those who indicated doubts about the validity of their test result were more likely to have had a colonoscopy.
Ninety-four percent of carriers in one study stated an intention to have annual or biannual colonoscopy in the future; among noncarriers, 64% did not intend to have colonoscopy in the future or were unsure, and 33% intended to have colonoscopy at 5- to 6-year intervals or less frequently.
A cross-sectional study conducted in the Netherlands examined the use of flexible sigmoidoscopy or colonoscopy among persons with CRC, endometrial cancer, or a clinical or genetic diagnosis of Lynch syndrome during a time that ranged from 2 years to 18 years after risk assessment and counseling.
Eighty-six percent of carriers of Lynch syndrome pathogenic variants, 68% of those who did not test or who had an uninformative Lynch syndrome genetic test result, and 73% of those with a clinical Lynch syndrome diagnosis were considered adherent with screening recommendations, based on data obtained from medical records. Participants also answered questions regarding screening adherence, and 16% of the overall sample reported that they had undergone screening less frequently than recommended. For the overall sample, greater perceived barriers to screening were associated with screening nonadherence as determined through medical record review, and embarrassment with screening procedures was associated with self-reported nonadherence. A second cross-sectional study, also conducted in the Netherlands, surveyed cancer-unaffected carriers of Lynch syndrome variants (n = 42) regarding their colorectal screening behaviors after learning their pathogenic variant status (range, 6 mo–8.5 y). Thirty-one percent of respondents reported that they had undergone annual colonoscopy before Lynch syndrome genetic testing, and 88% reported that they had undergone colonoscopy since their genetic diagnosis (P < .001).
Less is known about Lynch syndrome screening behaviors in persons who may be at risk of having a germline pathogenic variant but who do not undergo genetic counseling and/or genetic testing to learn about their risk status. Among relatives of carriers of a Lynch syndrome germline pathogenic variant from the Australian Colorectal Cancer Family Registry, 26 who had not undergone genetic counseling and/or testing completed an interview to assess their perceived risk of developing CRC in the next 10 years and to self-report their colonoscopy status.
Their mean perceived risk was 30.5%, which exceeded the mean predicted risk of 4% as calculated by MMRpro software.
Seventy-three percent (n = 19) reported having ever undergone a colonoscopy (one for diagnostic reasons); 35% had undergone colonoscopy within the past 2 years and were considered adherent to recommendations. Perceived risk was slightly and positively correlated with years since last colonoscopy (Pearson's r, 0.49; range, 0.02–0.79) but otherwise was not associated with other screening or personal characteristics. The authors concluded that perceived risk alone may not be a sufficient predictor of colonoscopy use in relatives of carriers of Lynch syndrome pathogenic variants who have not undergone genetic counseling and/or testing.
Several small studies have examined the use of screening for endometrial and ovarian cancers associated with Lynch syndrome (refer to Table 18). There are several limitations to these studies, including small sample sizes, short follow-up, retrospective design, reliance on self-report as the data source, and some not including patients who had undergone Lynch syndrome genetic testing. Several studies have included individuals in the screening uptake analysis who do not meet the minimum age criteria for undergoing screening. Of the studies that assessed screening use after a negative test result for a known pathogenic variant in the family, only a few assessed indications for that screening, such as follow-up of a previously identified abnormality. Last, some studies have included patients in the uptake analysis who were actively undergoing treatment for another cancer, which could influence provider screening recommendations. Therefore, Table 18 is limited to studies with patients who had undergone Lynch syndrome genetic testing, larger sample sizes, longer follow-up, and analysis that included individuals of an appropriate screening age.
Study Citation | Study Population | Uptake of Gynecologic Screening Before Genetic Counseling and Testing | Uptake of Gynecologic Screening After Receipt of Genetic Test Results | Length of Follow-up | Comments |
---|---|---|---|---|---|
Claes et al. (2005)1,a | Carriers (n = 7) | Not reported | TVUS | 1 y | One noncarrier reported undergoing TVUS for a previous endometrial problem, while three noncarriers reported undergoing the procedure for preventive reasons. |
– Carriers 86% (6/7) | |||||
Noncarriers (n = 16) | |||||
– Noncarriers 27% (4/15) | |||||
Collins et al. (2007)1,a | Carriers (n = 13) | Not reported | TVUS | 3 y | Two of four carriers had an RRH/RRSO by the 3-year follow-up assessment. |
– Carriers 69% (9/13) | |||||
– Noncarriers 6% (2/32) | |||||
Noncarriers (n = 32) | ES | ||||
– Carriers 54% (7/13) | |||||
– Noncarriers 3% (1/32) | |||||
Yurgelun et al. (2012): Cohort 12,a | 77 at risk of Lynch syndrome–associated EC; 45 carriers; 19 no genetic testing but Lynch syndrome–associated family history | 75% (58/77) engaged in EC screening or EC risk-reduction intervention; 42 underwent annual TVUS and/or ES; 16 underwent RRH | Not reported | N/A | |
Yurgelun et al. (2012): Cohort 21,a | 40 women at clinical risk of Lynch syndrome | 65% (26/40) adhered to EC screening or risk reduction; 6 underwent RRH; 13 underwent annual ES and/or TVUS; 6 had not reached recommended screening age | Carriers: 100% (n = 16) adhered to EC screening or risk-reducing strategies; 4 underwent pretest RRH; 5 underwent RRH; 5 underwent EC screening (TVUS and/or ES); 2 had not reached recommended screening age | 1 y | |
Carriers (n = 16) | |||||
Noncarriers (n = 9); 14 indeterminate results; 1 variant of uncertain significance | Noncarriers: 11% (1/9) underwent EC screening; 11% (1/9) underwent RRH | ||||
EC = endometrial cancer; ES = endometrial sampling; RRH = risk-reducing total abdominal hysterectomy; RRSO = risk-reducing salpingo-oophorectomy; TVUS = transvaginal ultrasound. | |||||
Noncarrier(s) = negative for known pathogenic variant in family. | |||||
1Prospective study design. | |||||
2Retrospective study design. | |||||
aSelf-report as data source. |
Overall, these studies have included relatively small numbers of women and suggest that screening rates for Lynch syndrome–associated gynecologic cancers are low before genetic counseling and testing. However, after participation in genetic education and counseling and the receipt of Lynch syndrome pathogenic variant test results, uptake of gynecologic cancer screening in carriers generally increases, while noncarriers decrease use.
There is no consensus regarding the use of risk-reducing colectomy for Lynch syndrome, and little is known about decision-making and psychological sequelae surrounding risk-reducing colectomy for Lynch syndrome.
Among persons who received positive test results, a greater proportion indicated interest in having risk-reducing colectomy after disclosure of results than at baseline.
This study also indicated that consideration of risk-reducing surgery for Lynch syndrome may motivate participation in genetic testing. Before receiving results, 46% indicated that they were considering risk-reducing colectomy, and 69% of women were considering risk-reducing total abdominal hysterectomy (RRH) and risk reducing bilateral salpingo-oophorectomy (RRSO); however, this study did not assess whether persons actually followed through with risk-reducing surgery after they received their test results. Before undergoing Lynch syndrome genetic counseling and testing, 5% of cancer-unaffected individuals at risk of a MMR variant in a longitudinal study reported that they would consider colectomy, and 5% of women indicated that they would have an RRH and an RRSO, if they were found to be pathogenic variant–positive. At 3 years after disclosure of results, no participants had undergone risk-reducing colectomy.
Two women who had undergone an RRH before genetic testing underwent RRSO within 1 year after testing,
however, no other female carriers of pathogenic variants in the study reported having either procedure at 3 years after test result disclosure.
In a cross-sectional quality-of-life and functional outcome survey of Lynch syndrome patients with more extensive (subtotal colectomy) or less extensive (segmental resection or hemicolectomy) resections, global quality-of-life outcomes were comparable, although patients with greater extent of resection described more frequent bowel movements and related dysfunction.
Family communication about genetic testing for hereditary CRC susceptibility, and specifically about the results of such testing, is complex. It is generally accepted that communication about genetic risk information within families is largely the responsibility of family members themselves. A few studies have examined communication patterns in families who had been offered Lynch syndrome genetic counseling and testing. Studies have focused on whether individuals disclosed information about Lynch syndrome genetic testing to their family members, to whom they disclosed this information, and family-based characteristics or issues that might facilitate or inhibit such communication. These studies examined communication and disclosure processes in families after notification by health care professionals about a Lynch syndrome predisposition and have comprised relatively small samples.
Research findings indicate that persons generally are willing to share information about the presence of a Lynch syndrome pathogenic variant within their families.
Motivations for sharing genetic risk information include a desire to increase family awareness about personal risk, health promotion options and predictive genetic testing, a desire for emotional support, and a perceived moral obligation and responsibility to help others in the family.
Findings across studies suggest that most study participants believed that Lynch syndrome genetic risk information is shared openly within families; however, such communication is more likely to occur with first-degree relatives (e.g., siblings, children) than with more distant relatives.
One Finnish study recruited parents aged 40 years or older and known to carry an MMR pathogenic variant to complete a questionnaire that investigated how parents shared knowledge of genetic risk with their adult and minor offspring. The study also identified challenges in the communication process.
Of 248 parents, 87% reported that they had disclosed results to their children. Reasons for nondisclosure were consistent with previous studies (young age of offspring, socially distant relationships, or feelings of difficulty in discussing the topic).
Nearly all parents had informed their adult offspring about their genetic risk and the possibility of genetic testing, but nearly one-third were unsure of how their offspring had used the information. Parents identified discussing their children’s cancer risk as the most difficult aspect of the communication process. Of the 191 firstborn children informed, 69% had undergone genetic testing. One-third of the parents suggested that health professionals should be involved in disclosure of the information and that a family appointment at the genetics clinic should be made at the time of disclosure.
In regard to informing second- and third-degree relatives, individuals may favor a cascade approach; for example, it is assumed that once a relative is given information about the family’s risk of Lynch syndrome, he or she would then be responsible for informing his or her first-degree relatives.
This cascade approach to communication is distinctly preferred in regard to informing relatives’ offspring, particularly those of minor age, and the consensus suggests that it would be inappropriate to disclose such information to a second-degree or third-degree relative without first proceeding through the family relational hierarchy.
In one study, persons who had undergone testing and were found to carry a Lynch syndrome–predisposing pathogenic variant were more likely than persons who had received true negative or uninformative results to inform at least one second-degree or third-degree relative about their genetic test results.
While communication about genetic risk is generally viewed as an open process, some communication barriers were reported across studies. Reasons for not informing a relative included lack of a close relationship and lack of contact with the individual; in fact, emotional, rather than relational, closeness seemed to be a more important determinant of the degree of risk communication. A desire to not worry relatives with information about test results and the perception that relatives would not understand the meaning of this information also have been cited as communication barriers.
Disclosure seemed less likely if at-risk individuals were considered too young to receive the information (i.e., children), if information about the hereditary cancer risk had previously created conflict in the family,
or if it was assumed that relatives would be uninterested in information about testing.
Prior existence of conflict seemed to inhibit discussions about hereditary cancer risk, particularly if such discussions involved disclosure of bad news.
For most participants in these studies, the news that the pattern of cancers in their families was attributable to a Lynch syndrome–predisposing pathogenic variant did not come as a surprise,
as individuals had suspected a hereditary cause for the familial cancers or had prior family discussions about cancer. Identification of a Lynch syndrome–predisposing pathogenic variant in the family was considered a private matter but not necessarily a secret,
and many individuals had discussed the family’s pathogenic variant status with someone outside of the family. Knowledge about the detection of a Lynch syndrome–predisposing pathogenic variant in the family was not viewed as stigmatizing, though individuals expressed concern about the potential impact of this information on insurance discrimination.
Also, while there may be a willingness to disclose information about the presence of a pathogenic variant in the family, one study suggests a tendency to remain more private about the disclosure of individual results, distinguishing personal results from familial risk information.
In a few cases, individuals reported that their relatives expressed anger, shock, or other negative emotional reactions after receiving news about the family’s Lynch syndrome risk;
however, most indicated little to no difficulty in informing their relatives.
It was suggested that families who are more comfortable and open with cancer-related discussions may be more receptive and accepting of news about genetic risk.
In some cases, probands reported feeling particularly obliged to inform family members about a hereditary cancer risk
and were often the strongest advocates for encouraging their family members to undergo genetic counseling and testing for the family pathogenic variant.
Some gender and family role differences also emerged in regard to the dissemination of hereditary cancer risk information. One study reported that female probands were more comfortable discussing genetic information than were male probands and that male probands showed a greater need for professional support during the family communication process.
Another study suggested that mothers may be particularly influential members of the family network in regard to communicating health risk information.
Pathogenic variant–negative individuals, persons who chose not to be tested, and spouses of at-risk persons reported not feeling as personally involved with the risk communication process compared with probands and other at-risk persons who had undergone genetic testing.
Various modes of communication (e.g., in-person, telephone, or written contact) may typically be used to disclose genetic risk information within families.
In one study, communication aids such as a genetic counseling summary letter or Lynch syndrome booklet were viewed as helpful adjuncts to the communication process but were not considered central or necessary to its success.
Studies have suggested that recommendations by health care providers to inform relatives about hereditary cancer risk may encourage communication about Lynch syndrome
and that support by health care professionals may be helpful in overcoming barriers to communicating such information to family members.
Much of the literature to date on family communication has focused on disclosure of test results; however, other elements of family communication are currently being explored. One study evaluated the role of older family members in providing various types of support (e.g., instrumental, emotional, crisis help, and dependability when needed) among individuals with Lynch syndrome and their family members (206 respondents from 33 families).
Respondents completed interviews about their family social network (biological and non-biological relatives and others outside the family) and patterns of communication within their family. The median age of the respondents and the members of their family social network did not differ (age 43 y). The study found that 23% of the members of the family social network encouraged CRC screening (other types of support, such as social support, were reported much more frequently). Those who encouraged screening were older, female, and significant others or biological family members, rather than nonfamily members. Given that many of the members of the family social network did not live in the same household, the study points out the importance of extended family in the context of screening encouragement and support.
The uptake for genetic testing for FAP may be higher than testing for Lynch syndrome. A study of asymptomatic individuals in the United States at risk of FAP who were enrolled in a CRC registry and were offered genetic counseling found that 82% of adults and 95% of minors underwent genetic testing.
Uptake rates close to 100% have been reported in the United Kingdom.
A possible explanation for the greater uptake of APC genetic testing is that it may be more cost-effective than annual endoscopic screening
and can eliminate the burden of annual screening, which must often be initiated before puberty. The opportunity to eliminate worry about potential risk-reducing surgery is another possible benefit of genetic testing for FAP. The decision to have APC genetic testing may be viewed as a medical management decision;
the potential psychosocial factors that may influence the testing decision are not as well studied for FAP as for other hereditary cancer syndromes. The higher penetrance of APC pathogenic variants, earlier onset of disease, and the unambiguous phenotype also may influence the decision to undergo genetic testing for this condition, possibly because of a greater awareness of the disease and more experience with multiple family members being affected.
Genetic testing for FAP is presently offered to children with affected parents, often at the age of 10 to 12 years, when endoscopic screening is recommended. Because it is optimal to diagnose FAP before age 18 years to prevent CRC and because screening and possibly surgery are warranted at the time an individual is identified as a carrier of an APC pathogenic variant, genetic testing of minors is justified in this instance. (Refer to the Testing in children section in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for a more detailed discussion regarding the ethical, psychosocial, and genetic counseling issues related to genetic testing in children.)
In a survey conducted in the Netherlands of members of families with FAP, one-third (34%) believed that it was most suitable to offer APC gene testing to children before age 12 years, whereas 38% preferred to offer testing to children between the ages of 12 and 16 years, when children would be better able to understand the DNA testing process. Only 4% felt that children should not undergo DNA testing at all.
Results of qualitative interview data from 28 U.S. parents diagnosed with FAP showed that 61% favored genetic testing of APC variants in their at-risk children (aged 10–17 y); 71% believed that their children should receive their test results. The primary reasons why parents chose to test their children included early detection and management, reduction in parental anxiety and uncertainty, and help with decision making regarding surveillance. Reasons provided for not testing focused on discrimination concerns and cost.
Clinical observations suggest that children who have family members affected with FAP are very aware of the possibility of risk-reducing surgery, and focus on the test result as the factor that determines the need for such surgery.
It is important to consider the timing of disclosure of genetic test results to children in regard to their age, developmental issues, and psychological concerns about FAP. Children who carry an APC pathogenic variant have expressed concern regarding how they will be perceived by peers and might benefit from assistance in formulating an explanation for others that preserves self-esteem.
Studies evaluating psychological outcomes after genetic testing for FAP suggest that some individuals, particularly carriers of pathogenic variants, may be at risk of experiencing increased distress. In a cross-sectional study of adults who had previously undergone APC genetic testing, those who were carriers of pathogenic variants exhibited higher levels of state anxiety than noncarriers and were more likely to exhibit clinically significant anxiety levels.
Lower optimism and lower self-esteem were associated with higher anxiety in this study,
and FAP-related distress, perceived seriousness of FAP, and belief in the accuracy of genetic testing were associated with more state anxiety among carriers.
However, in an earlier study that compared adults who had undergone genetic testing for FAP, Huntington disease, and hereditary breast/ovarian cancer syndrome, FAP-specific distress was somewhat elevated within 1 week after disclosure of either positive or negative test results and was lower overall than the other syndromes.
In a cross-sectional Australian study focusing on younger adults aged 18 to 35 years diagnosed with FAP (N = 88), participants most frequently reported the following FAP-related issues for which they perceived the need for moderate-to-high levels of support or assistance: anxiety regarding their children’s risk of developing FAP, fear about developing cancer, and uncertainty about the impact of FAP.
Seventy-five percent indicated that they would consider prenatal testing for FAP; 61% would consider PGT, and 61% would prefer that their children undergo genetic testing at birth or before age 10 years. A small proportion of respondents (16%) reported experiencing some FAP-related discrimination, primarily indicating that attending to their medical or self-care needs (e.g., time off work for screening, need for frequent toilet breaks, and physical limitations) may engender negative attitudes in colleagues and managers.
Another large cross-sectional study of FAP families conducted in the Netherlands included persons aged 16 to 84 years who either had an FAP diagnosis, were at 50% risk of having an APC pathogenic variant, or were proven APC noncarriers.
Of those who had APC testing, 48% had done so at least 5 years or longer before this study. Of persons with an FAP diagnosis, 76% had undergone preventive colectomy, and 78% of those were at least 5 years postsurgery. The study evaluated the prevalence of generalized psychological distress, distress related specifically to FAP, and cancer-related worries. Mean scores on the Mental Health Index-5, a subscale of the SF-36 that assessed generalized distress, were comparable to the general Dutch population. Twenty percent of respondents were classified as having moderate to high levels of FAP-specific distress as measured by the Impact of Event scale (IES), with 23% of those with an FAP diagnosis, 11% of those at risk of FAP, and 17% of noncarriers reporting scores in this range. Five percent reported scores on the IES that indicated severe and clinically relevant distress; of those, the majority (78%) had an FAP diagnosis. Overall, mean scores on the Cancer Worry Scale were comparable to those found in another study of families with Lynch syndrome. Persons with an FAP diagnosis were more likely to report more frequent cancer worries, and the most commonly reported worries were the potential need for additional surgery (26%) and the likelihood that they (17%) or a family member (14%) will develop cancer. In multivariate analysis, factors associated with higher levels of FAP-specific distress included greater perceived risk of developing cancer, more frequent discussion about FAP with family or friends, and having no children. Factors associated with higher levels of cancer-specific worries included being female, poorer family functioning, greater actual and desired discussion about FAP with family or friends, greater perceived cancer risk, poorer general health perceptions, and having been a caregiver for a family member with cancer. The authors noted that most factors that were associated with higher levels of cancer- and FAP-specific distress or worry were psychosocial factors, rather than clinical or demographic factors.
Another cross-sectional study conducted in the Netherlands found that among FAP patients, 37% indicated that the disease had influenced their desire to have children (i.e., wanting fewer or no children). Thirty-three percent indicated that they would consider PND for FAP; 30% would consider PGT. Higher levels of guilt and more positive attitudes towards terminating pregnancy were associated with greater interest for both PND and PGT.
In a separate U.S. study, predictors of willingness to consider prenatal testing included having an affected child and experiencing a first-degree relative’s death secondary to FAP.
The psychological vulnerability of children undergoing testing is of particular concern in genetic testing for FAP. Research findings suggest that most children do not experience clinically significant psychological distress after APC testing. As in studies involving adults, however, subgroups may be vulnerable to increased distress and would benefit from continued psychological support. A study of children who had undergone genetic testing for FAP found that their mood and behavior remained in the normal range after genetic counseling and disclosure of test results. Aspects of the family situation, including illness in the mother or a sibling were associated with subclinical increases in depressive symptoms.
In a long-term follow-up study of 48 children undergoing testing for FAP, most children did not suffer psychological distress; however, a small proportion of children tested demonstrated clinically significant posttest distress.
Another study found that although APC pathogenic variant–positive children’s perceived risk of developing the disease increased after disclosure of results, anxiety and depression levels remain unchanged in the year after disclosure.
Pathogenic variant–negative children in this study experienced less anxiety and improved self-esteem over this same time period.
Less is known about psychological aspects of screening for FAP. One study of a small number of persons (aged 17–53 y) with a family history of FAP who were offered participation in a genetic counseling and testing protocol found that among those who were asymptomatic, all reported undergoing at least one endoscopic surveillance before participation in the study.
Only 33% (two of six patients) reported continuing screening at the recommended interval. Of the affected persons who had undergone colectomy, 92% (11 of 12 patients) were adherent to recommended colorectal surveillance. In a cross-sectional study of 150 persons with a clinical or genetic diagnosis of classic FAP or attenuated FAP (AFAP) and at-risk relatives, 52% of those with FAP and 46% of relatives at risk of FAP, had undergone recommended endoscopic screening.
Among persons who had or were at risk of AFAP, 58% and 33%, respectively, had undergone screening. Compared with persons who had undergone screening within the recommended time interval, those who had not screened were less likely to recall provider recommendations for screening, more likely to lack health insurance or insurance reimbursement for screening, and more likely to believe that they are not at increased risk of CRC. Only 42% of the study population had ever undergone genetic counseling. A small percentage of participants (14%–19%) described screening as a “necessary evil,” indicating a dislike for the bowel preparation, or experienced pain and discomfort. Nineteen percent reported that these issues might pose barriers to undergoing future endoscopies. Nineteen percent reported that improved techniques and the use of anesthesia have improved tolerance for screening procedures.
When persons at risk of FAP develop multiple polyps, risk-reducing surgery in the form of subtotal colectomy or proctocolectomy is the only effective way to reduce the risk of CRC. Most persons with FAP can avoid a permanent ostomy and preserve the anus and/or rectum, allowing some degree of bowel continence. (Refer to the Interventions for FAP section of this summary for more information about surgical management procedures in FAP.) Evidence on the quality-of-life outcomes from these interventions continues to accumulate and is summarized in Table 19.
Population | Length of Follow-up | Type of Procedure | Stool Frequency | Stool Continence | Body Image | Sexual Functioning | Comments |
---|---|---|---|---|---|---|---|
279 FAP-affected individuals (135 females and 144 males) after colectomy; controls included 1,771 individuals from the general Dutch population | IRA mean: 12 y (SD, 7.5 y) | IRA: n = 161 | Not assessed | Not assessed | EORTC QLQ-CR38 a | EORTC QLQ-CR38 a | SF-36b scores (Dutch version) on all subscales were significantly lower than the scores in the general population (IRA: P < .001; IPAA: P < .001). |
IRA: 87.5 (SD, 21.9) | IRA: 38.9 (SD, 26.6) | ||||||
IPAA mean: 6.8 y (SD, 4.9 y) | IPAA: n = 118 | IPAA: 84.4 (SD, 22.7) | IPAA: 42.2 (SD, 26.3) | ||||
88 Australian individuals (63 females and 25 males) aged 18–35 y, including 57 after colectomy and 14 with FAP but no surgery | Not reported | IRA: n = 33 | Not assessed | Not assessed | SF-36 b | SF-36 b | |
IPAA: n = 21 | IRA: 89.9 (SD, 16.1) | IRA: 86.2 (SD, 21.6) | |||||
Ileostomy: n = 1 | IPAA: 72.1 (SD, 23) | IPAA: 77.5 (SD, 26.2) | |||||
Unknown surgery type: n = 2 | No surgery: 94.1 (SD, 9.4) | No surgery: 91 (SD, 19) | |||||
525 individuals (283 females and 242 males) including 296 after colectomy, 45 with FAP but no surgery, 50 at risk for FAP and no surgery, and 134 noncarriers | Range: 0–1 y to >10 y | IRA: n = 136 | Not assessed | Not assessed | EORTC QLQ-CR38 a | EORTC QLQ-CR38 a | 41% of FAP patients reported employment disruptions: |
After colectomy: 85.4 (SD, 20.5) | After colectomy: 42.2 (SD, 23.2) | Part or complete disability: n = 73 (59%) | |||||
IPAA: n = 112 | FAP no surgery: 91.9 (SD, 16.1) | After colectomy: 42.2 (SD, 23.2) | Worked less: n = 30 (24%) | ||||
Ileostomy: n = 42 | At risk: 94.0 (SD, 13.1) | At risk: 47.6 (SD, 23.7) | Worked more n = 5 (4%) | ||||
Other: n = 6 | Noncarrier: 92.3 (SD, 13.1) | Noncarrier: 45.7 (SD, 21.2) | Worked more or less at different periods: n = 16 (13%) | ||||
209 Swedish FAP-affected individuals (116 females and 93 males) after colectomy aged 18–75 y | Mean time since last surgery: 14 y (SD, 10; range, 1–50 y) | IRA: n = 71 | Not assessed | Day: 71% (n = 149) | Not assessed | Not assessed | The mean number of 21 abdominal symptoms assessed was 7 (SD, 4.61; range, 1–18). Women reported more symptoms than men, but there were no differences between genders regarding the degree the symptoms were troublesome. Higher symptom number was an independent predictor of poorer physical and mental health. |
IPAA: n = 82 | |||||||
Ileostomy: n = 39 | Night: 61% (n = 128) | ||||||
Continent ileostomy: n = 14 | |||||||
Other: n = 3 | |||||||
28 individuals (10 females and 18 males) who underwent colectomy at age 14 y or younger | 12 y (SD, 8.4; range, 1–37 y) | IRA: n = 7 | Day: | Day: | Rosenberg self-esteem score : 25.53/30 c | Not assessed | 10/28 reported cancer-related worry post colectomy, with a trend that young age (<18 y) was associated with more cancer-related worry. |
IRA: 3.8 (SD, 1.5) | IRA: 71.4% (n = 7) | ||||||
IPAA: 5.3 (SD, 2.4) | IPAA: 85.7% (n = 21) | ||||||
IPAA: n = 21 | Night: | Night: | |||||
IRA: 1.3 (SD, 0.6) | IRA: 50.0% (n = 7) | ||||||
IPAA: 1.3 (SD, 0.5) | IPAA: 61.9% (n = 21) | ||||||
EORTC QLQ = European Organization for Research and Treatment of Cancer Colorectal Quality of Life Questionnaire; IPAA = ileal pouch-anal anastomosis; IRA = ileorectal anastomosis; SD = standard deviation; SF-36 = Short Form (36) Health Survey. | |||||||
aEORTC QLQ-C38 scores range from 0–100. Functional scales: 0 = lowest level of function and 100 = highest/healthy level of function. Symptom scales: 0 = lowest level of symptomatology and 100 = highest level of symptomatology. | |||||||
bSF-36 scores range from 0–100, with 0 = lowest possible health status and 100 = best possible health status. | |||||||
cWithin normal ranges for same age group. |
Studies of risk-reducing surgery for FAP have found that general measures of quality of life have been within normal range, and the majority reported no negative impact on their body image. However, these studies suggest that risk-reducing surgery for FAP may have negative quality-of-life effects for at least some proportion of those affected.
Chemoprevention trials are currently under way to evaluate the effectiveness of various therapies for persons at risk of Lynch syndrome and FAP.
In a sample of persons diagnosed with FAP who were invited to take part in a 5-year trial to evaluate the effects of vitamins and fiber on the development of adenomatous polyps, 55% agreed to participate.
Participants were more likely to be younger, to have been more recently diagnosed with FAP, and to live farther from the trial center, but did not differ from nonparticipants on any other psychosocial variables.
The possibility of transmitting a pathogenic variant to a child may pose a concern to families affected by hereditary CRC syndromes to the extent that some carriers may avoid childbearing. These concerns also may prompt individuals to consider using prenatal diagnosis (PND) methods to help reduce the risk of transmission. PND 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.
Both procedures carry a small risk of miscarriage.
Moreover, discovering the fetus is a carrier of a cancer susceptibility variant may impose a difficult decision for couples regarding pregnancy continuation or termination and may require additional professional consultation and support.
An alternative to these tests is preimplantation genetic testing (PGT), a procedure used to test fertilized embryos for genetic disorders before uterine implantation.
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 APC.
From the limited studies published to date, there appears to be interest in the use of ART for FAP, Lynch syndrome, and PJS.
However, actual uptake rates have not been reported.
Study Population | N c | Interest or Intention in ART | Comments |
---|---|---|---|
FAP-affected individuals | 20 | 95% (19/20) would consider prenatal GT for FAP; 90% (18/20) would consider PGT; 75% (15/20) would consider amniocentesis or chorionic villous sampling | |
FAP-affected individuals | 341 | 33% (16/64) would consider PND for FAP; 30% (76/256) would consider PGT; 15% (52/341) felt terminating pregnancy for FAP was acceptable | 24% and 25% of patients did not respond to questions about attitudes toward PND and PGT, respectively. |
Individuals with an APC pathogenic variant associated with FAP | 65 d | 25% (16/64) were aware of PGT; 78% (50/64) thought PGT should be offered; 55% (31/56) would consider PGT | |
Individuals undergoing genetic testing for Lynch syndrome | 48 e | 21% 10/48) would consider PND and/or PGT; 19% (9/48) would consider only PND; 2% (1/48) would consider only PGT | At 1 year after disclosure of GT results, two of nine carriers reported that they were considering PGT for future pregnancy. |
Individuals with an identified Lynch syndrome pathogenic variant | 43 f | 19% (8/42) were aware of PGT; 69% (29/42) thought PGT should be offered; 41% (16/39) would consider PGT | |
PJS-affected individualsa | 52 | 15% (8/52) indicated that pregnancy termination was acceptable if PND identified a fetus with PJS; 52% (27/52) indicated PGT was acceptable for persons with PJS | Ten (19%) individuals, nine of whom were female, reported that they had decided not to conceive a child because of PJS. |
GT = genetic testing; PGT = preimplantation genetic testing; PND = prenatal diagnosis. | |||
aStudies used a cross-sectional design and were conducted in the United States, and in the Netherlands. | |||
bParticipants were invited to complete questionnaires before clinical genetic testing for Lynch syndrome and at 3 months and 1 year after disclosure of genetic test results. | |||
cIndicates number of participants older than 18 y, unless otherwise specified. | |||
dTotal number of individuals with an APC pathogenic variant. Not all individuals answered or were eligible to answer each question. | |||
eRepresents the number who indicated that they were considering having children in the future, out of a total of 130 individuals who answered a questionnaire before genetic testing. | |||
fTotal number of individuals with a Lynch syndrome pathogenic variant. Not all individuals answered or were eligible to answer each question. |
对PDQ癌症信息总结定期进行审查,并在获得新信息后进行更新。本节描述了截至上述日期对此总结所做的最新变更。
更新美国国家综合癌症网络 (NCCN) 作为参考 55。
这一节作了广泛的修订。
更新NCCN 作为参考 121。
增加文本以说明,2019年一项使用大型、基于社区、整合的美国医疗保健系统数据的回顾性研究,通过所有结直肠癌 (CRC) 的错配修复蛋白 (MMR) 免疫组化法比较了年龄限制筛查策略与无年龄上限的普遍筛查策略对林奇综合征的确诊效果。(引用Li等人作为参考331)。林奇综合征的确诊在70岁到75岁之间显著下降,在80岁之后增长幅度最小。
增加文本以说明,对151例CRC患者进行的小型单机构分析,通过多基因(组合)检测在9.9%患者中发现了致病性生殖系变体(引自You等人作为参考350)。
增加文本以说明,一项对14例早发性CRC和MMR缺陷患者的回顾性研究表明,43%的患者患有林奇综合征,57%患有林奇样综合征(引用Antelo等人作为参考530)。
本总结由独立于NCI的PDQ癌症遗传学编辑委员会编写和维护。总结反映了对文献的独立审查,并不代表NCI或NIH的政策声明。关于总结政策和PDQ编辑委员会在维护PDQ总结中的作用的更多信息,请参见关于本PDQ总结和PDQ®-NCI的综合性癌症数据库页面。
The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.
Updated National Comprehensive Cancer Network (NCCN) as reference 55.
This section was extensively revised.
Updated NCCN as reference 121.
Added text to state that a 2019 retrospective study using data from a large, community-based, integrated U.S. health care system compared the diagnostic performance of age-restricted screening strategies for Lynch syndrome by reflex mismatch repair (MMR) immunohistochemistry of all colorectal cancers (CRCs) versus a universal screening strategy without an upper age limit (cited Li et al. as reference 331). Lynch syndrome identification decreased substantially after age 70 years to age 75 years, with minimal incremental gain after age 80 years.
Added text to state that a small single-institution analysis of 151 individuals with CRC identified pathogenic germline variants in 9.9% of individuals through multigene (panel) testing (cited You et al. as reference 350).
Added text to state that a retrospective study of 14 patients with early-onset CRC and MMR deficiency reported that 43% of patients had Lynch syndrome and 57% had Lynch-like syndrome (cited Antelo et al. as reference 530).
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 colorectal cancer. 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 Colorectal Cancer 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 Colorectal Cancer. 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 colorectal cancer. 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 Colorectal Cancer 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 Colorectal Cancer. 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.