由于流行病学经常将结肠癌和直肠癌放在一起(如结直肠癌),很难在流行病方面将直肠癌和结肠癌区分。我国肿瘤登记报告也是将结肠癌和直肠癌放在了一起,统称为结直肠癌。
在世界范围内,结直肠癌是第三大恶性肿瘤。2012年,结直肠癌新发病例估计为136万,死亡病例为69.4万。在中国,结直肠癌也是第三大恶性肿瘤
2019年美国结直肠癌的预计新发病例和死亡病例:
据估计,2015年中国结直肠癌中国人口标化发病率为18.02/10万,死亡率为8.21/10万 ;世界人口标化发病率17.81/10万,死亡率8.12/10万。
结直肠癌对男性和女性的影响几乎相同。在美国所有种族中,非裔美国人的散发性结直肠癌发病率和死亡率最高。男性结直肠癌发病率高于女性。直肠位于盆腔内,从肛门齿状线的移行粘膜延伸到腹膜折返处的乙状结肠;;通过乙状结肠镜检查,距离肛缘10cm-15cm为直肠
直肠肿瘤的位置通常由肛缘、齿状线或肛肠环与肿瘤下缘之间的距离来标识,测量值因使用的内窥镜软硬度不同或数字不同而有所差异。
肿瘤与肛门括约肌之间的距离代表着保肛手术的可能性。盆腔的骨性结构约束了直肠的手术入路,这就降低了获得广泛阴性切缘的可能性,并增加了局部复发的风险。
年龄的增长是大多数癌症的一个最重要的风险因素。结直肠癌的其他风险因素还包括:
建议将直肠癌筛查作为50岁及以上成人的常规体检项目,特别是直系亲属结直肠癌家族史的人群,主要有以下几点因素:
(更多信息,请参考PDQ摘要“结直肠癌筛查”相关内容。)
与结肠癌相似,直肠癌的症状可能包括:
除了梗阻症状外,其他症状不一定与疾病的分期有关,也不一定代表着某种特定的诊断。
起始临床评估可能包括:
体格检查可能发现直肠内有明显的肿块和鲜血。淋巴结肿大、肝肿大或肺部征象可提示转移。
实验室检查可发现缺铁性贫血、电解质和肝功能异常。
直肠癌患者的预后与以下因素相关,包括:
在多机构前瞻性研究中,只有疾病分期(由肿瘤[T]、区域淋巴结[N]和远处转移[M]定义)被证明是预后因素。
一项大型的汇总分析评估了T和N分期的作用以及辅助治疗对直肠癌患者生存和复发的影响,并且这一结果已被发表并得到证实。
已有大量研究对其他的临床、病理和分子指标进行了评估。
迄今为止,还未有一个得到多中心前瞻性试验的证实。例如,高度微卫星不稳定性与林奇综合征相关的直肠癌具有关联性。在一个以人群为基础的纳入607例样本的临床试验中,结果提示微卫星不稳定或能改善发病年龄在50岁或以下结直肠癌患者的生存期,但与临床分期无关。
此外,据报道,基因表达谱分析有助于预测直肠腺癌对术前放化疗的反应,并预测5-FU为基础的新辅助放化疗用于临床Ⅱ期和Ⅲ期直肠癌的预后。
直肠癌辅助治疗后患者的总生存率(OS)存在种族和种族差异。黑人的总体生存期较白人短。这种差异的影响因素可能包括肿瘤的位置、手术方式和是否共病。
直肠癌术后监测项目的主要目标是:
直肠癌治疗后,应定期复查,有助于复发疾病早期发现、早期治疗。
两项临床试验中已证实密切随诊对患者的生存期具有重要的临床意义。一项荟萃分析将这2项试验和另外4项报告结合分析,指出密切随诊可改善患者的生存率,具有统计学意义。
美国和欧洲主流的肿瘤学会对结直肠癌初次治疗后的监测指南各不相同,因此最佳监测方案未达成统一。
需要进行大规模、合理设计的、前瞻性、多中心、随机研究,以建立基于循证证据的共识,进行随访评估。
癌胚抗原(CEA)是一种血清糖蛋白,常用于直肠癌患者的治疗和随访。一篇文章就肿瘤标志物CEA的临床应用提出以下几点:
在一项荷兰回顾性研究中,对全直肠系膜切除治疗直肠癌进行了研究。研究者发现大多数直肠癌患者的术前血清CEA水平正常,而复发患者的血清CEA水平至少上升了50%。作者得出结论,术前血清CEA水平正常的直肠癌患者,不能忽视术后CEA监测。
包含直肠癌信息的其他PDQ摘要如下:
It is difficult to separate epidemiological considerations of rectal cancer from those of colon cancer because epidemiological studies often consider colon and rectal cancer (i.e., colorectal cancer) together.
Worldwide, colorectal cancer is the third most common form of cancer. In 2012, there were an estimated 1.36 million new cases of colorectal cancer and 694,000 deaths.
Estimated new cases and deaths from rectal and colon cancer in the United States in 2019:
Colorectal cancer affects men and women almost equally. Among all racial groups in the United States, African Americans have the highest sporadic colorectal cancer incidence and mortality rates.
The rectum is located within the pelvis, extending from the transitional mucosa of the anal dentate line to the sigmoid colon at the peritoneal reflection; by rigid sigmoidoscopy, the rectum measures between 10 cm and 15 cm from the anal verge.
The location of a rectal tumor is usually indicated by the distance between the anal verge, dentate line, or anorectal ring and the lower edge of the tumor, with measurements differing depending on the use of a rigid or flexible endoscope or digital examination.
The distance of the tumor from the anal sphincter musculature has implications for the ability to perform sphincter-sparing surgery. The bony constraints of the pelvis limit surgical access to the rectum, which results in a lesser likelihood of attaining widely negative margins and a higher risk of local recurrence.
Increasing age is the most important risk factor for most cancers. Other risk factors for colorectal cancer include the following:
Evidence supports screening for rectal cancer as a part of routine care for all adults aged 50 years and older, especially for those with first-degree relatives with colorectal cancer, for the following reasons:
(Refer to the PDQ summary on Colorectal Cancer Screening for more information.)
Similar to colon cancer, symptoms of rectal cancer may include the following:
With the exception of obstructive symptoms, these symptoms do not necessarily correlate with the stage of disease or signify a particular diagnosis.
The initial clinical evaluation may include the following:
Physical examination may reveal a palpable mass and bright blood in the rectum. Adenopathy, hepatomegaly, or pulmonary signs may be present with metastatic disease.
Laboratory examination may reveal iron-deficiency anemia and electrolyte and liver function abnormalities.
The prognosis of patients with rectal cancer is related to several factors, including the following:
Only disease stage (designated by tumor [T], nodal status [N], and distant metastasis [M]) has been validated as a prognostic factor in multi-institutional prospective studies.
A major pooled analysis evaluating the impact of T and N stage and treatment on survival and relapse in patients with rectal cancer who are treated with adjuvant therapy has been published and confirms these findings.
A large number of studies have evaluated other clinical, pathologic, and molecular parameters.
As yet, none has been validated in multi-institutional prospective trials. For example, microsatellite instability–high, also associated with Lynch syndrome–related rectal cancer, was shown to be associated with improved survival independent of tumor stage in a population-based series of 607 patients with colorectal cancer who were 50 years old or younger at the time of diagnosis.
In addition, gene expression profiling has been reported to be useful in predicting the response of rectal adenocarcinomas to preoperative chemoradiation therapy and in determining the prognosis of stages II and III rectal cancer after neoadjuvant 5-fluorouracil-based chemoradiation therapy.
Racial and ethnic differences in overall survival (OS) after adjuvant therapy for rectal cancer have been observed, with shorter OS for blacks than for whites. Factors contributing to this disparity may include tumor position, type of surgical procedure, and presence of comorbid conditions.
The primary goals of postoperative surveillance programs for rectal cancer are:
Routine, periodic studies following treatment for rectal cancer may lead to earlier identification and management of recurrent disease.
A statistically significant survival benefit has been demonstrated for more intensive follow-up protocols in two clinical trials. A meta-analysis that combined these two trials with four others reported a statistically significant improvement in survival for patients who were intensively followed.
Guidelines for surveillance after initial treatment with curative intent for colorectal cancer vary between leading U.S. and European oncology societies, and optimal surveillance strategies remain uncertain.
Large, well-designed, prospective, multi-institutional, randomized studies are required to establish an evidence-based consensus for follow-up evaluation.
Measurement of CEA, a serum glycoprotein, is frequently used in the management and follow-up of patients with rectal cancer. A review of the use of this tumor marker for rectal cancer suggests the following:
In one Dutch retrospective study of total mesorectal excision for the treatment of rectal cancer, investigators found that the preoperative serum CEA level was normal in the majority of patients with rectal cancer, and yet, serum CEA levels rose by at least 50% in patients with recurrence. The authors concluded that serial, postoperative CEA testing cannot be discarded based on a normal preoperative serum CEA level in patients with rectal cancer.
Other PDQ summaries containing information related to rectal cancer include the following:
在美国,绝大多数的直肠癌是腺癌。其他组织学类型占结直肠肿瘤的2%-5%。
世界卫生组织对结直肠癌的分类包括:
(更多信息,请参考PDQ摘要“非霍奇金淋巴瘤治疗”相关内容。)
Adenocarcinomas account for the vast majority of rectal tumors in the United States. Other histologic types account for an estimated 2% to 5% of colorectal tumors.
The World Health Organization classification of tumors of the colon and rectum includes the following:
(Refer to the PDQ summary on Adult Non-Hodgkin Lymphoma Treatment for more information.)
准确的临床分期可提供直肠原发肿瘤的位置、大小以及是否有转移病灶,转移灶大小、数目和位置等重要信息。准确的初始分期可以帮助确定手术类型和新辅助治疗与否,从而最大限度地提高手术切缘阴性率,并最终影响临床疗效。
临床评估和分期措施可能包括:
在直肠癌的肿瘤(T)分期中,数项研究表明直肠内超声的准确率达80%~95%,而CT和MRI的准确率分别为65%~75%和75%~85%。通过直肠内超声确定受累转移性淋巴结的准确率约为70%-75%,而CT和MRI分别为55%-65%和60%-70%。
在一项纳入84项研究的荟萃分析中,就评估淋巴结(N)方面,没有发现直肠内超声、CT和MRI三者中的任何一种明显优于其他两种方法。
硬管式与软管式内镜的直肠内超声相比,二者在T和N分期上方面具有同样的准确率;然而,有一些技术上的问题,直肠内超声对T分期和N分期可能会得出不确定或不准确的结论。因此,可以考虑通过MRI或软管式直肠内超声进行进一步评估。
CRM被定义为腹膜后或腹膜外膜软组织边界距离肿瘤浸润最深部位的最短距离
AJCC采用TNM分类方法对直肠癌进行临床分期。
同种分类方法适用于临床分期和病理分期。
制定临床决策时,应参考TNM分级(肿瘤、淋巴结和转移癌),而不是陈旧的Dukes或改良版Astler-Coller分类体系。
该分期体系常用于下列癌症,包括结肠和直肠的腺癌、高级神经内分泌癌和鳞癌。未采用该分期系统的癌症包括以下组织病理学类型的癌症:阑尾癌、肛门癌和高分化神经内分泌肿瘤(类癌)。
(更多信息,请参考PDQ摘要“肛门癌治疗和胃肠道类癌治疗”相关内容。)
AJCC和一个美国国家癌症研究所资助的工作组建议,针对未接受新辅助治疗的患者,在根治性结肠和直肠切除术中至少应检查10到14个淋巴结。在肿瘤姑息性切除或术前接受放疗的患者中,淋巴结数目要求可少一些。
术中检测淋巴结的数目可以反映淋巴血管、肠系膜的侵袭程度,还有助于肿瘤的病理诊断。
回顾性研究INT-0089(EST-2288)显示结直肠癌手术中检查的淋巴结数目与患者的临床转归具有相关性。
最近提出了一种新的淋巴结阳性直肠癌-转移分期方案
分期 | TNM | 说明 | 图示 |
---|---|---|---|
分期 | TNM | 说明 | 图示 |
分期 | TNM | 说明 | 图示 |
分期 | TNM | 说明 | 图示 |
分期 | TNM | 说明 | 图示 |
0 | Tis, N0, M0 | Tis=原位癌,粘膜内层癌(侵犯固有层,但未穿透黏膜肌层)。 | |
N0=无区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(不在病理诊断范围内。) | |||
分期 | TNM | 说明 | 图示 |
I | T1–T2, N0, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |
T2=肿瘤侵犯固有肌层。 | |||
N0=无区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
分期 | TNM | 说明 | 图示 |
IIA | T3, N0, M0 | T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |
N0=无区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
IIB | T4a, N0, M0 | T4a=肿瘤穿透脏层腹膜,包括穿透肿瘤的严重肠穿孔,接着肿瘤通过炎症区域侵袭到脏层腹膜表层 | |
N0=无区域淋巴结转移 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
IIC | T4b, N0, M0 | T4b=肿瘤直接侵犯或粘连于相邻器官或结构。 | |
N0=无区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
分期 | TNM | 说明 | 图示 |
IIIA | T1, N2a, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |
N2a= 4-6枚区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
T1–2, N1/N1c, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | ||
T2=肿瘤侵犯固有肌层。 | |||
N1=1-3枚区域淋巴结转移(淋巴结内癌结节≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1c=无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现肿瘤种植。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
IIIB | T1–T2, N2b, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |
T2=肿瘤侵犯固有肌层。 | |||
N2b=7枚或以上区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
T2–T3, N2a, M0 | T2=肿瘤侵犯固有肌层。 | ||
T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |||
N2a=4-6枚区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
T3–T4a, N1/N1c, M0 | T3=肿瘤穿透固有肌层进入结直肠旁组织。 | ||
T4=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
–T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透肠壁,并通过炎症部位连续侵犯脏层腹膜表层)。 | |||
N1=1-3枚区域淋巴结转移(淋巴结内癌结节≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1c= 无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现肿瘤种植。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
IIIC | T3–T4a, N2b, M0 | T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |
T4=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
–T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透肠壁,并通过炎症部位连续侵犯脏层腹膜表层)。 | |||
N2b=7枚或以上区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
T4a, N2a, M0 | T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透肠壁,并通过炎症部位连续侵犯脏层腹膜表层)。 | ||
N2a=4-6枚区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
T4b, N1–N2, M0 | T4b=肿瘤直接侵犯或粘连于相邻器官或结构。 | ||
N1=1-3枚区域淋巴结转移(淋巴结内癌结节≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1a=1枚区域淋巴结转移。 | |||
–N1b=2-3枚区域淋巴结转移。 | |||
–N1c= 无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现肿瘤种植。 | |||
N2=4枚或以上区域淋巴结转移。 | |||
–N2a= 4-6枚区域淋巴结转移。 | |||
–N2b=7枚或以上区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
分期 | TNM | 说明 | 图示 |
IVA | 任何T、任何N、M1a | TX=原发肿瘤无法评估。 | |
T0=无原发肿瘤证据。 | |||
Tis=原位癌,粘膜内癌(侵犯固有层,但未穿透黏膜肌层)。 | |||
T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |||
T2=肿瘤侵犯固有肌层。 | |||
T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |||
T4=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
–T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透引起的肠穿孔,以及肿瘤通过炎症区域侵袭到脏层腹膜表层) | |||
–T4b=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
NX=区域淋巴结无法评估。 | |||
N0=无区域淋巴结转移。 | |||
N1=1-3枚区域淋巴结转移(淋巴结内肿瘤≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1a=1枚区域淋巴结转移。 | |||
–N1b=2-3枚区域淋巴结转移。 | |||
–N1c= 无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现癌结节。 | |||
N2=4枚或以上区域淋巴结转移。 | |||
–N2a= 4-6枚区域淋巴结转移。 | |||
–N2b=7枚或以上区域淋巴结转移。 | |||
M1a=单个部位或器官转移,未见腹膜转移。 | |||
IVB | 任何 T、任何 N、 M1b | 任何T=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的T说明部分。 | |
任何N=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的N说明部分。 | |||
M1b=2个或2个以上部位或器官转移,未见腹膜转移。 | |||
IVC | 任何 T、任何 N、 M1c | 任何T=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的T说明部分。 | |
任何N=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的N说明部分。 | |||
M1c=仅腹膜转移或/伴随其他部位或器官转移。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
上标b和c的释义参见表5末尾。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
上标b和c的释义参见表5末尾。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
上标b和c的释义参见表5末尾。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
上标b和c的释义参见表5末尾。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
b T4中的直接侵犯包括侵犯其他器官或穿透浆膜直接侵犯结直肠其他部位,并经内镜确诊(如通过盲肠癌侵犯乙状结肠),或腹膜后腹膜下癌,或穿透固有肌层直接侵犯其他器官和结构(如降结肠后壁肿瘤侵犯左肾或侧腹壁;或直肠中远端肿瘤侵犯前列腺、精索、宫颈和阴道)。 | |||
c 肿瘤粘连于其他器官和结构,大体上归为cT4b。然而,如果镜下观察未见肿瘤粘连,按照肠壁侵犯深度归为pT1-4a。应将V和L分类标准用于鉴别是否伴血管和淋巴管侵犯,而PN预后影响因素应用于周围神经侵犯。 |
分期 | TNM | 说明 | 图示 |
---|---|---|---|
分期 | TNM | 说明 | 图示 |
分期 | TNM | 说明 | 图示 |
分期 | TNM | 说明 | 图示 |
I | T1–T2, N0, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |
T2=肿瘤侵犯固有肌层。 | |||
N0=无区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
分期 | TNM | 说明 | 图示 |
IIA | T3, N0, M0 | T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |
N0=无区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
IIB | T4a, N0, M0 | T4a=肿瘤穿透脏层腹膜,包括穿透肿瘤的严重肠穿孔,接着肿瘤通过炎症区域侵袭到脏层腹膜表层 | |
N0=无区域淋巴结转移 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
IIC | T4b, N0, M0 | T4b=肿瘤直接侵犯或粘连于相邻器官或结构。 | |
N0=无区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
分期 | TNM | 说明 | 图示 |
IIIA | T1, N2a, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |
N2a= 4-6枚区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
T1–2, N1/N1c, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | ||
T2=肿瘤侵犯固有肌层。 | |||
N1=1-3枚区域淋巴结转移(淋巴结内癌结节≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1c=无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现肿瘤种植。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
IIIB | T1–T2, N2b, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |
T2=肿瘤侵犯固有肌层。 | |||
N2b=7枚或以上区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
T2–T3, N2a, M0 | T2=肿瘤侵犯固有肌层。 | ||
T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |||
N2a=4-6枚区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
T3–T4a, N1/N1c, M0 | T3=肿瘤穿透固有肌层进入结直肠旁组织。 | ||
T4=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
–T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透肠壁,并通过炎症部位连续侵犯脏层腹膜表层)。 | |||
N1=1-3枚区域淋巴结转移(淋巴结内癌结节≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1c= 无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现肿瘤种植。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
IIIC | T3–T4a, N2b, M0 | T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |
T4=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
–T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透肠壁,并通过炎症部位连续侵犯脏层腹膜表层)。 | |||
N2b=7枚或以上区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
T4a, N2a, M0 | T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透肠壁,并通过炎症部位连续侵犯脏层腹膜表层)。 | ||
N2a=4-6枚区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
T4b, N1–N2, M0 | T4b=肿瘤直接侵犯或粘连于相邻器官或结构。 | ||
N1=1-3枚区域淋巴结转移(淋巴结内癌结节≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1a=1枚区域淋巴结转移。 | |||
–N1b=2-3枚区域淋巴结转移。 | |||
–N1c= 无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现肿瘤种植。 | |||
N2=4枚或以上区域淋巴结转移。 | |||
–N2a= 4-6枚区域淋巴结转移。 | |||
–N2b=7枚或以上区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
分期 | TNM | 说明 | 图示 |
IVA | 任何T、任何N、M1a | TX=原发肿瘤无法评估。 | |
T0=无原发肿瘤证据。 | |||
Tis=原位癌,粘膜内癌(侵犯固有层,但未穿透黏膜肌层)。 | |||
T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |||
T2=肿瘤侵犯固有肌层。 | |||
T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |||
T4=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
–T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透引起的肠穿孔,以及肿瘤通过炎症区域侵袭到脏层腹膜表层) | |||
–T4b=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
NX=区域淋巴结无法评估。 | |||
N0=无区域淋巴结转移。 | |||
N1=1-3枚区域淋巴结转移(淋巴结内肿瘤≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1a=1枚区域淋巴结转移。 | |||
–N1b=2-3枚区域淋巴结转移。 | |||
–N1c= 无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现癌结节。 | |||
N2=4枚或以上区域淋巴结转移。 | |||
–N2a= 4-6枚区域淋巴结转移。 | |||
–N2b=7枚或以上区域淋巴结转移。 | |||
M1a=单个部位或器官转移,未见腹膜转移。 | |||
IVB | 任何 T、任何 N、 M1b | 任何T=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的T说明部分。 | |
任何N=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的N说明部分。 | |||
M1b=2个或2个以上部位或器官转移,未见腹膜转移。 | |||
IVC | 任何 T、任何 N、 M1c | 任何T=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的T说明部分。 | |
任何N=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的N说明部分。 | |||
M1c=仅腹膜转移或/伴随其他部位或器官转移。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
上标b和c的释义参见表5末尾。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
上标b和c的释义参见表5末尾。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
上标b和c的释义参见表5末尾。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
b T4中的直接侵犯包括侵犯其他器官或穿透浆膜直接侵犯结直肠其他部位,并经内镜确诊(如通过盲肠癌侵犯乙状结肠),或腹膜后腹膜下癌,或穿透固有肌层直接侵犯其他器官和结构(如降结肠后壁肿瘤侵犯左肾或侧腹壁;或直肠中远端肿瘤侵犯前列腺、精索、宫颈和阴道)。 | |||
c 肿瘤粘连于其他器官和结构,大体上归为cT4b。然而,如果镜下观察未见肿瘤粘连,按照肠壁侵犯深度归为pT1-4a。应将V和L分类标准用于鉴别是否伴血管和淋巴管侵犯,而PN预后影响因素应用于周围神经侵犯。 |
分期 | TNM | 说明 | 图示 |
---|---|---|---|
分期 | TNM | 说明 | 图示 |
分期 | TNM | 说明 | 图示 |
IIA | T3, N0, M0 | T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |
N0=无区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
IIB | T4a, N0, M0 | T4a=肿瘤穿透脏层腹膜,包括穿透肿瘤的严重肠穿孔,接着肿瘤通过炎症区域侵袭到脏层腹膜表层 | |
N0=无区域淋巴结转移 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
IIC | T4b, N0, M0 | T4b=肿瘤直接侵犯或粘连于相邻器官或结构。 | |
N0=无区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
分期 | TNM | 说明 | 图示 |
IIIA | T1, N2a, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |
N2a= 4-6枚区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
T1–2, N1/N1c, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | ||
T2=肿瘤侵犯固有肌层。 | |||
N1=1-3枚区域淋巴结转移(淋巴结内癌结节≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1c=无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现肿瘤种植。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
IIIB | T1–T2, N2b, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |
T2=肿瘤侵犯固有肌层。 | |||
N2b=7枚或以上区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
T2–T3, N2a, M0 | T2=肿瘤侵犯固有肌层。 | ||
T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |||
N2a=4-6枚区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
T3–T4a, N1/N1c, M0 | T3=肿瘤穿透固有肌层进入结直肠旁组织。 | ||
T4=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
–T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透肠壁,并通过炎症部位连续侵犯脏层腹膜表层)。 | |||
N1=1-3枚区域淋巴结转移(淋巴结内癌结节≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1c= 无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现肿瘤种植。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
IIIC | T3–T4a, N2b, M0 | T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |
T4=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
–T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透肠壁,并通过炎症部位连续侵犯脏层腹膜表层)。 | |||
N2b=7枚或以上区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
T4a, N2a, M0 | T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透肠壁,并通过炎症部位连续侵犯脏层腹膜表层)。 | ||
N2a=4-6枚区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
T4b, N1–N2, M0 | T4b=肿瘤直接侵犯或粘连于相邻器官或结构。 | ||
N1=1-3枚区域淋巴结转移(淋巴结内癌结节≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1a=1枚区域淋巴结转移。 | |||
–N1b=2-3枚区域淋巴结转移。 | |||
–N1c= 无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现肿瘤种植。 | |||
N2=4枚或以上区域淋巴结转移。 | |||
–N2a= 4-6枚区域淋巴结转移。 | |||
–N2b=7枚或以上区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
分期 | TNM | 说明 | 图示 |
IVA | 任何T、任何N、M1a | TX=原发肿瘤无法评估。 | |
T0=无原发肿瘤证据。 | |||
Tis=原位癌,粘膜内癌(侵犯固有层,但未穿透黏膜肌层)。 | |||
T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |||
T2=肿瘤侵犯固有肌层。 | |||
T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |||
T4=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
–T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透引起的肠穿孔,以及肿瘤通过炎症区域侵袭到脏层腹膜表层) | |||
–T4b=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
NX=区域淋巴结无法评估。 | |||
N0=无区域淋巴结转移。 | |||
N1=1-3枚区域淋巴结转移(淋巴结内肿瘤≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1a=1枚区域淋巴结转移。 | |||
–N1b=2-3枚区域淋巴结转移。 | |||
–N1c= 无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现癌结节。 | |||
N2=4枚或以上区域淋巴结转移。 | |||
–N2a= 4-6枚区域淋巴结转移。 | |||
–N2b=7枚或以上区域淋巴结转移。 | |||
M1a=单个部位或器官转移,未见腹膜转移。 | |||
IVB | 任何 T、任何 N、 M1b | 任何T=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的T说明部分。 | |
任何N=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的N说明部分。 | |||
M1b=2个或2个以上部位或器官转移,未见腹膜转移。 | |||
IVC | 任何 T、任何 N、 M1c | 任何T=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的T说明部分。 | |
任何N=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的N说明部分。 | |||
M1c=仅腹膜转移或/伴随其他部位或器官转移。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
上标b和c的释义参见表5末尾。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
上标b和c的释义参见表5末尾。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
b T4中的直接侵犯包括侵犯其他器官或穿透浆膜直接侵犯结直肠其他部位,并经内镜确诊(如通过盲肠癌侵犯乙状结肠),或腹膜后腹膜下癌,或穿透固有肌层直接侵犯其他器官和结构(如降结肠后壁肿瘤侵犯左肾或侧腹壁;或直肠中远端肿瘤侵犯前列腺、精索、宫颈和阴道)。 | |||
c 肿瘤粘连于其他器官和结构,大体上归为cT4b。然而,如果镜下观察未见肿瘤粘连,按照肠壁侵犯深度归为pT1-4a。应将V和L分类标准用于鉴别是否伴血管和淋巴管侵犯,而PN预后影响因素应用于周围神经侵犯。 |
分期 | TNM | 说明 | 图示 |
---|---|---|---|
分期 | TNM | 说明 | 图示 |
IIIA | T1, N2a, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |
N2a= 4-6枚区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
T1–2, N1/N1c, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | ||
T2=肿瘤侵犯固有肌层。 | |||
N1=1-3枚区域淋巴结转移(淋巴结内癌结节≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1c=无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现肿瘤种植。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
IIIB | T1–T2, N2b, M0 | T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |
T2=肿瘤侵犯固有肌层。 | |||
N2b=7枚或以上区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
T2–T3, N2a, M0 | T2=肿瘤侵犯固有肌层。 | ||
T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |||
N2a=4-6枚区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴。) | |||
T3–T4a, N1/N1c, M0 | T3=肿瘤穿透固有肌层进入结直肠旁组织。 | ||
T4=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
–T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透肠壁,并通过炎症部位连续侵犯脏层腹膜表层)。 | |||
N1=1-3枚区域淋巴结转移(淋巴结内癌结节≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1c= 无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现肿瘤种植。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
IIIC | T3–T4a, N2b, M0 | T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |
T4=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
–T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透肠壁,并通过炎症部位连续侵犯脏层腹膜表层)。 | |||
N2b=7枚或以上区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
T4a, N2a, M0 | T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透肠壁,并通过炎症部位连续侵犯脏层腹膜表层)。 | ||
N2a=4-6枚区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
T4b, N1–N2, M0 | T4b=肿瘤直接侵犯或粘连于相邻器官或结构。 | ||
N1=1-3枚区域淋巴结转移(淋巴结内癌结节≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1a=1枚区域淋巴结转移。 | |||
–N1b=2-3枚区域淋巴结转移。 | |||
–N1c= 无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现肿瘤种植。 | |||
N2=4枚或以上区域淋巴结转移。 | |||
–N2a= 4-6枚区域淋巴结转移。 | |||
–N2b=7枚或以上区域淋巴结转移。 | |||
M0=影像检查等未见远处转移;未见远处部位或器官转移证据。(病理科医生不负责该范畴) | |||
分期 | TNM | 说明 | 图示 |
IVA | 任何T、任何N、M1a | TX=原发肿瘤无法评估。 | |
T0=无原发肿瘤证据。 | |||
Tis=原位癌,粘膜内癌(侵犯固有层,但未穿透黏膜肌层)。 | |||
T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |||
T2=肿瘤侵犯固有肌层。 | |||
T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |||
T4=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
–T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透引起的肠穿孔,以及肿瘤通过炎症区域侵袭到脏层腹膜表层) | |||
–T4b=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
NX=区域淋巴结无法评估。 | |||
N0=无区域淋巴结转移。 | |||
N1=1-3枚区域淋巴结转移(淋巴结内肿瘤≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1a=1枚区域淋巴结转移。 | |||
–N1b=2-3枚区域淋巴结转移。 | |||
–N1c= 无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现癌结节。 | |||
N2=4枚或以上区域淋巴结转移。 | |||
–N2a= 4-6枚区域淋巴结转移。 | |||
–N2b=7枚或以上区域淋巴结转移。 | |||
M1a=单个部位或器官转移,未见腹膜转移。 | |||
IVB | 任何 T、任何 N、 M1b | 任何T=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的T说明部分。 | |
任何N=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的N说明部分。 | |||
M1b=2个或2个以上部位或器官转移,未见腹膜转移。 | |||
IVC | 任何 T、任何 N、 M1c | 任何T=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的T说明部分。 | |
任何N=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的N说明部分。 | |||
M1c=仅腹膜转移或/伴随其他部位或器官转移。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
上标b和c的释义参见表5末尾。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
b T4中的直接侵犯包括侵犯其他器官或穿透浆膜直接侵犯结直肠其他部位,并经内镜确诊(如通过盲肠癌侵犯乙状结肠),或腹膜后腹膜下癌,或穿透固有肌层直接侵犯其他器官和结构(如降结肠后壁肿瘤侵犯左肾或侧腹壁;或直肠中远端肿瘤侵犯前列腺、精索、宫颈和阴道)。 | |||
c 肿瘤粘连于其他器官和结构,大体上归为cT4b。然而,如果镜下观察未见肿瘤粘连,按照肠壁侵犯深度归为pT1-4a。应将V和L分类标准用于鉴别是否伴血管和淋巴管侵犯,而PN预后影响因素应用于周围神经侵犯。 |
分期 | TNM | 说明 | 图示 |
---|---|---|---|
IVA | 任何T、任何N、M1a | TX=原发肿瘤无法评估。 | |
T0=无原发肿瘤证据。 | |||
Tis=原位癌,粘膜内癌(侵犯固有层,但未穿透黏膜肌层)。 | |||
T1=肿瘤侵犯黏膜下层(穿透黏膜肌层,但未穿透固有肌层)。 | |||
T2=肿瘤侵犯固有肌层。 | |||
T3=肿瘤穿透固有肌层进入结直肠旁组织。 | |||
T4=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
–T4a=肿瘤穿透脏层腹膜(包括肿瘤穿透引起的肠穿孔,以及肿瘤通过炎症区域侵袭到脏层腹膜表层) | |||
–T4b=肿瘤直接侵犯或粘连于相邻器官或结构。 | |||
NX=区域淋巴结无法评估。 | |||
N0=无区域淋巴结转移。 | |||
N1=1-3枚区域淋巴结转移(淋巴结内肿瘤≥0.2mm),或任意数目的淋巴结出现肿瘤沉积且可识别的淋巴结均为阴性。 | |||
–N1a=1枚区域淋巴结转移。 | |||
–N1b=2-3枚区域淋巴结转移。 | |||
–N1c= 无区域淋巴结转移,但浆膜下、肠系膜或无腹膜覆盖的结肠旁组织,或直肠旁/直肠系膜组织出现癌结节。 | |||
N2=4枚或以上区域淋巴结转移。 | |||
–N2a= 4-6枚区域淋巴结转移。 | |||
–N2b=7枚或以上区域淋巴结转移。 | |||
M1a=单个部位或器官转移,未见腹膜转移。 | |||
IVB | 任何 T、任何 N、 M1b | 任何T=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的T说明部分。 | |
任何N=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的N说明部分。 | |||
M1b=2个或2个以上部位或器官转移,未见腹膜转移。 | |||
IVC | 任何 T、任何 N、 M1c | 任何T=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的T说明部分。 | |
任何N=见上方关于任何肿瘤、任何区域淋巴结、M1a TNM分期中的N说明部分。 | |||
M1c=仅腹膜转移或/伴随其他部位或器官转移。 | |||
T=原发肿瘤;N=局部淋巴结;M=远处转移。 | |||
a 已取得AJCC授权复印:Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74。 | |||
b T4中的直接侵犯包括侵犯其他器官或穿透浆膜直接侵犯结直肠其他部位,并经内镜确诊(如通过盲肠癌侵犯乙状结肠),或腹膜后腹膜下癌,或穿透固有肌层直接侵犯其他器官和结构(如降结肠后壁肿瘤侵犯左肾或侧腹壁;或直肠中远端肿瘤侵犯前列腺、精索、宫颈和阴道)。 | |||
c 肿瘤粘连于其他器官和结构,大体上归为cT4b。然而,如果镜下观察未见肿瘤粘连,按照肠壁侵犯深度归为pT1-4a。应将V和L分类标准用于鉴别是否伴血管和淋巴管侵犯,而PN预后影响因素应用于周围神经侵犯。 |
Accurate staging provides crucial information about the location and size of the primary tumor in the rectum, and, if present, the size, number, and location of any metastases. Accurate initial staging can influence therapy by helping to determine the type of surgical intervention and the choice of neoadjuvant therapy to maximize the likelihood of resection with clear margins. In primary rectal cancer, pelvic imaging helps determine the following:
Clinical evaluation and staging procedures may include the following:
In the tumor (T) staging of rectal carcinoma, several studies indicate that the accuracy of endorectal ultrasound ranges from 80% to 95% compared with 65% to 75% for CT and 75% to 85% for MRI. The accuracy in determining metastatic nodal involvement by endorectal ultrasound is approximately 70% to 75% compared with 55% to 65% for CT and 60% to 70% for MRI.
In a meta-analysis of 84 studies, none of the three imaging modalities, including endorectal ultrasound, CT, and MRI, were found to be significantly superior to the others in staging nodal (N) status.
Endorectal ultrasound using a rigid probe may be similarly accurate in T and N staging when compared with endorectal ultrasound using a flexible scope; however, a technically difficult endorectal ultrasound may give an inconclusive or inaccurate result for both T stage and N stage. In this case, further assessment by MRI or flexible endorectal ultrasound may be considered.
In patients with rectal cancer, the circumferential resection margin is an important pathological staging parameter. Measured in millimeters, it is defined as the retroperitoneal or peritoneal adventitial soft-tissue margin closest to the deepest penetration of tumor.
The AJCC has designated staging by TNM (tumor, node, metastasis) classification to define rectal cancer.
The same classification is used for both clinical and pathologic staging.
Treatment decisions are made with reference to the TNM classification system, rather than the older Dukes or Modified Astler-Coller classification schema.
Cancers staged using this staging system include adenocarcinomas, high-grade neuroendocrine carcinomas, and squamous carcinomas of the colon and rectum. Cancers not staged using this staging system include these histopathologic types of cancer: appendiceal carcinomas, anal carcinomas, well-differentiated neuroendocrine tumors (carcinoids).
(Refer to the PDQ summaries on Anal Cancer Treatment and the Gastrointestinal Carcinoid Tumors Treatment for more information.)
The AJCC and a National Cancer Institute-sponsored panel suggested that at least 10 to 14 lymph nodes be examined in radical colon and rectum resections in patients who did not receive neoadjuvant therapy. In cases in which a tumor is resected for palliation or in patients who have received preoperative radiation therapy, fewer lymph nodes may be present.
This takes into consideration that the number of lymph nodes examined is a reflection of both the aggressiveness of lymphovascular mesenteric dissection at the time of surgical resection and the pathologic identification of nodes in the specimen.
Retrospective studies, such as Intergroup trial INT-0089 (EST-2288), have demonstrated that the number of lymph nodes examined during colon and rectal surgery may be associated with patient outcome.
A new tumor-metastasis staging strategy for node-positive rectal cancer has been proposed.
Stage | TNM | Description | Illustration |
---|---|---|---|
Stage | TNM | Description | Illustration |
Stage | TNM | Description | Illustration |
Stage | TNM | Description | Illustration |
Stage | TNM | Definition | Illustration |
0 | Tis, N0, M0 | Tis = Carcinoma in situ, intramucosal carcinoma (involvement of lamina propria with no extension through muscularis mucosae). | |
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
Stage | TNM | Description | Illustration |
I | T1–T2, N0, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |
T2 = Tumor invades the muscularis propria. | |||
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
Stage | TNM | Description | Illustration |
IIA | T3, N0, M0 | T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIB | T4a, N0, M0 | T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIC | T4b, N0, M0 | T4b = Tumor directly invades or adheres to adjacent organs or structures. | |
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
Stage | TNM | Description | Illustration |
IIIA | T1, N2a, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |
N2a = Four to six regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T1–2, N1/N1c, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | ||
T2 = Tumor invades the muscularis propria. | |||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIIB | T1–T2, N2b, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |
T2 = Tumor invades the muscularis propria. | |||
N2b = Seven or more regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T2–T3, N2a, M0 | T2 = Tumor invades the muscularis propria. | ||
T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |||
N2a = Four to six regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T3–T4a, N1/N1c, M0 | T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | ||
T4 = Tumor invades the visceral peritoneum or invades or adheres to adjacent organ or structure. | |||
–T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIIC | T3–T4a, N2b, M0 | T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |
T4 = Tumor invades the visceral peritoneum or invades or adheres to adjacent organ or structure. | |||
–T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |||
N2b = Seven or more regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T4a, N2a, M0 | T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | ||
N2a = Four to six regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T4b, N1–N2, M0 | T4b = Tumor directly invades or adheres to adjacent organs or structures. | ||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1a = One regional lymph node is positive. | |||
–N1b = Two or three regional lymph nodes are positive. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
N2 = Four or more regional nodes are positive. | |||
–N2a = Four to six regional lymph nodes are positive. | |||
–N2b = Seven or more regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
Stage | TNM | Definition | Illustration |
IVA | Any T, Any N, M1a | TX = Primary tumor cannot be assessed. | |
T0 = No evidence of primary tumor. | |||
Tis = Carcinoma in situ, intramucosal carcinoma (involvement of lamina propria with no extension through muscularis mucosae). | |||
T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |||
T2 = Tumor invades the muscularis propria. | |||
T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |||
T4 = Tumor invades the visceral peritoneum or invades or adheres to adjacent organ or structure. | |||
–T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |||
–T4b = Tumor directly invades or adheres to adjacent organs or structures. | |||
NX = Regional lymph nodes cannot be assessed. | |||
N0 = No regional lymph node metastasis. | |||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1a = One regional lymph node is positive. | |||
–N1b = Two or three regional lymph nodes are positive. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
N2 = Four or more regional nodes are positive. | |||
–N2a = Four to six regional lymph nodes are positive. | |||
–N2b = Seven or more regional lymph nodes are positive. | |||
M1a = Metastasis to one site or organ is identified without peritoneal metastasis. | |||
IVB | Any T, Any N, M1b | Any T = See T descriptions above in Any T, Any N, M1a TNM stage group. | |
Any N = See N descriptions above in Any T, Any N1, M1a TNM stage group. | |||
M1b = Metastasis to two or more sites or organs is identified without peritoneal metastasis. | |||
IVC | Any T, Any N, M1c | Any T = See T descriptions above in Any T, Any N, M1a TNM stage group. | |
Any N = See N descriptions above in Any T, Any N1, M1a TNM stage group. | |||
M1c = Metastasis to the peritoneal surface is identified alone or with other site or organ metastases. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
The explanations for superscripts b and c are at the end of Table 5. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
The explanations for superscripts b and c are at the end of Table 5. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
The explanations for superscripts b and c are at the end of Table 5. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
The explanations for superscripts b and c are at the end of Table 5. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
b Direct invasion in T4 includes invasion of other organs or other segments of the colorectum as a result of direct extension through the serosa, as confirmed on microscopic examination (e.g., invasion of the sigmoid colon by a carcinoma of the cecum) or, for cancers in a retroperitoneal or subperitoneal location, direct invasion of other organs or structures by virtue of extension beyond the muscularis propria (i.e., respectively, a tumor on the posterior wall of the descending colon invading the left kidney or lateral abdominal wall; or a mid or distal rectal cancer with invasion of prostate, seminal vesicles, cervix, or vagina). | |||
cTumor that is adherent to other organs or structures, grossly, is classified cT4b. However, if no tumor is present in the adhesion, microscopically, the classification should be pT1-4a depending on the anatomical depth of wall invasion. The V and L classification should be used to identify the presence or absence of vascular or lymphatic invasion whereas the PN prognostic factor should be used for perineural invasion. |
Stage | TNM | Description | Illustration |
---|---|---|---|
Stage | TNM | Description | Illustration |
Stage | TNM | Description | Illustration |
Stage | TNM | Definition | Illustration |
I | T1–T2, N0, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |
T2 = Tumor invades the muscularis propria. | |||
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
Stage | TNM | Description | Illustration |
IIA | T3, N0, M0 | T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIB | T4a, N0, M0 | T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIC | T4b, N0, M0 | T4b = Tumor directly invades or adheres to adjacent organs or structures. | |
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
Stage | TNM | Description | Illustration |
IIIA | T1, N2a, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |
N2a = Four to six regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T1–2, N1/N1c, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | ||
T2 = Tumor invades the muscularis propria. | |||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIIB | T1–T2, N2b, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |
T2 = Tumor invades the muscularis propria. | |||
N2b = Seven or more regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T2–T3, N2a, M0 | T2 = Tumor invades the muscularis propria. | ||
T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |||
N2a = Four to six regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T3–T4a, N1/N1c, M0 | T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | ||
T4 = Tumor invades the visceral peritoneum or invades or adheres to adjacent organ or structure. | |||
–T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIIC | T3–T4a, N2b, M0 | T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |
T4 = Tumor invades the visceral peritoneum or invades or adheres to adjacent organ or structure. | |||
–T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |||
N2b = Seven or more regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T4a, N2a, M0 | T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | ||
N2a = Four to six regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T4b, N1–N2, M0 | T4b = Tumor directly invades or adheres to adjacent organs or structures. | ||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1a = One regional lymph node is positive. | |||
–N1b = Two or three regional lymph nodes are positive. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
N2 = Four or more regional nodes are positive. | |||
–N2a = Four to six regional lymph nodes are positive. | |||
–N2b = Seven or more regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
Stage | TNM | Definition | Illustration |
IVA | Any T, Any N, M1a | TX = Primary tumor cannot be assessed. | |
T0 = No evidence of primary tumor. | |||
Tis = Carcinoma in situ, intramucosal carcinoma (involvement of lamina propria with no extension through muscularis mucosae). | |||
T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |||
T2 = Tumor invades the muscularis propria. | |||
T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |||
T4 = Tumor invades the visceral peritoneum or invades or adheres to adjacent organ or structure. | |||
–T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |||
–T4b = Tumor directly invades or adheres to adjacent organs or structures. | |||
NX = Regional lymph nodes cannot be assessed. | |||
N0 = No regional lymph node metastasis. | |||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1a = One regional lymph node is positive. | |||
–N1b = Two or three regional lymph nodes are positive. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
N2 = Four or more regional nodes are positive. | |||
–N2a = Four to six regional lymph nodes are positive. | |||
–N2b = Seven or more regional lymph nodes are positive. | |||
M1a = Metastasis to one site or organ is identified without peritoneal metastasis. | |||
IVB | Any T, Any N, M1b | Any T = See T descriptions above in Any T, Any N, M1a TNM stage group. | |
Any N = See N descriptions above in Any T, Any N1, M1a TNM stage group. | |||
M1b = Metastasis to two or more sites or organs is identified without peritoneal metastasis. | |||
IVC | Any T, Any N, M1c | Any T = See T descriptions above in Any T, Any N, M1a TNM stage group. | |
Any N = See N descriptions above in Any T, Any N1, M1a TNM stage group. | |||
M1c = Metastasis to the peritoneal surface is identified alone or with other site or organ metastases. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
The explanations for superscripts b and c are at the end of Table 5. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
The explanations for superscripts b and c are at the end of Table 5. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
The explanations for superscripts b and c are at the end of Table 5. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
b Direct invasion in T4 includes invasion of other organs or other segments of the colorectum as a result of direct extension through the serosa, as confirmed on microscopic examination (e.g., invasion of the sigmoid colon by a carcinoma of the cecum) or, for cancers in a retroperitoneal or subperitoneal location, direct invasion of other organs or structures by virtue of extension beyond the muscularis propria (i.e., respectively, a tumor on the posterior wall of the descending colon invading the left kidney or lateral abdominal wall; or a mid or distal rectal cancer with invasion of prostate, seminal vesicles, cervix, or vagina). | |||
cTumor that is adherent to other organs or structures, grossly, is classified cT4b. However, if no tumor is present in the adhesion, microscopically, the classification should be pT1-4a depending on the anatomical depth of wall invasion. The V and L classification should be used to identify the presence or absence of vascular or lymphatic invasion whereas the PN prognostic factor should be used for perineural invasion. |
Stage | TNM | Description | Illustration |
---|---|---|---|
Stage | TNM | Description | Illustration |
Stage | TNM | Definition | Illustration |
IIA | T3, N0, M0 | T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIB | T4a, N0, M0 | T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIC | T4b, N0, M0 | T4b = Tumor directly invades or adheres to adjacent organs or structures. | |
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
Stage | TNM | Description | Illustration |
IIIA | T1, N2a, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |
N2a = Four to six regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T1–2, N1/N1c, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | ||
T2 = Tumor invades the muscularis propria. | |||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIIB | T1–T2, N2b, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |
T2 = Tumor invades the muscularis propria. | |||
N2b = Seven or more regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T2–T3, N2a, M0 | T2 = Tumor invades the muscularis propria. | ||
T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |||
N2a = Four to six regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T3–T4a, N1/N1c, M0 | T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | ||
T4 = Tumor invades the visceral peritoneum or invades or adheres to adjacent organ or structure. | |||
–T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIIC | T3–T4a, N2b, M0 | T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |
T4 = Tumor invades the visceral peritoneum or invades or adheres to adjacent organ or structure. | |||
–T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |||
N2b = Seven or more regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T4a, N2a, M0 | T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | ||
N2a = Four to six regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T4b, N1–N2, M0 | T4b = Tumor directly invades or adheres to adjacent organs or structures. | ||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1a = One regional lymph node is positive. | |||
–N1b = Two or three regional lymph nodes are positive. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
N2 = Four or more regional nodes are positive. | |||
–N2a = Four to six regional lymph nodes are positive. | |||
–N2b = Seven or more regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
Stage | TNM | Definition | Illustration |
IVA | Any T, Any N, M1a | TX = Primary tumor cannot be assessed. | |
T0 = No evidence of primary tumor. | |||
Tis = Carcinoma in situ, intramucosal carcinoma (involvement of lamina propria with no extension through muscularis mucosae). | |||
T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |||
T2 = Tumor invades the muscularis propria. | |||
T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |||
T4 = Tumor invades the visceral peritoneum or invades or adheres to adjacent organ or structure. | |||
–T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |||
–T4b = Tumor directly invades or adheres to adjacent organs or structures. | |||
NX = Regional lymph nodes cannot be assessed. | |||
N0 = No regional lymph node metastasis. | |||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1a = One regional lymph node is positive. | |||
–N1b = Two or three regional lymph nodes are positive. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
N2 = Four or more regional nodes are positive. | |||
–N2a = Four to six regional lymph nodes are positive. | |||
–N2b = Seven or more regional lymph nodes are positive. | |||
M1a = Metastasis to one site or organ is identified without peritoneal metastasis. | |||
IVB | Any T, Any N, M1b | Any T = See T descriptions above in Any T, Any N, M1a TNM stage group. | |
Any N = See N descriptions above in Any T, Any N1, M1a TNM stage group. | |||
M1b = Metastasis to two or more sites or organs is identified without peritoneal metastasis. | |||
IVC | Any T, Any N, M1c | Any T = See T descriptions above in Any T, Any N, M1a TNM stage group. | |
Any N = See N descriptions above in Any T, Any N1, M1a TNM stage group. | |||
M1c = Metastasis to the peritoneal surface is identified alone or with other site or organ metastases. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
The explanations for superscripts b and c are at the end of Table 5. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
The explanations for superscripts b and c are at the end of Table 5. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
b Direct invasion in T4 includes invasion of other organs or other segments of the colorectum as a result of direct extension through the serosa, as confirmed on microscopic examination (e.g., invasion of the sigmoid colon by a carcinoma of the cecum) or, for cancers in a retroperitoneal or subperitoneal location, direct invasion of other organs or structures by virtue of extension beyond the muscularis propria (i.e., respectively, a tumor on the posterior wall of the descending colon invading the left kidney or lateral abdominal wall; or a mid or distal rectal cancer with invasion of prostate, seminal vesicles, cervix, or vagina). | |||
cTumor that is adherent to other organs or structures, grossly, is classified cT4b. However, if no tumor is present in the adhesion, microscopically, the classification should be pT1-4a depending on the anatomical depth of wall invasion. The V and L classification should be used to identify the presence or absence of vascular or lymphatic invasion whereas the PN prognostic factor should be used for perineural invasion. |
Stage | TNM | Description | Illustration |
---|---|---|---|
Stage | TNM | Definition | Illustration |
IIIA | T1, N2a, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |
N2a = Four to six regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T1–2, N1/N1c, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | ||
T2 = Tumor invades the muscularis propria. | |||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIIB | T1–T2, N2b, M0 | T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |
T2 = Tumor invades the muscularis propria. | |||
N2b = Seven or more regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T2–T3, N2a, M0 | T2 = Tumor invades the muscularis propria. | ||
T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |||
N2a = Four to six regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T3–T4a, N1/N1c, M0 | T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | ||
T4 = Tumor invades the visceral peritoneum or invades or adheres to adjacent organ or structure. | |||
–T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
IIIC | T3–T4a, N2b, M0 | T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |
T4 = Tumor invades the visceral peritoneum or invades or adheres to adjacent organ or structure. | |||
–T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |||
N2b = Seven or more regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T4a, N2a, M0 | T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | ||
N2a = Four to six regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
T4b, N1–N2, M0 | T4b = Tumor directly invades or adheres to adjacent organs or structures. | ||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1a = One regional lymph node is positive. | |||
–N1b = Two or three regional lymph nodes are positive. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
N2 = Four or more regional nodes are positive. | |||
–N2a = Four to six regional lymph nodes are positive. | |||
–N2b = Seven or more regional lymph nodes are positive. | |||
M0 = No distant metastasis by imaging, etc.; no evidence of tumor in distant sites or organs. (This category is not assigned by pathologists.) | |||
Stage | TNM | Definition | Illustration |
IVA | Any T, Any N, M1a | TX = Primary tumor cannot be assessed. | |
T0 = No evidence of primary tumor. | |||
Tis = Carcinoma in situ, intramucosal carcinoma (involvement of lamina propria with no extension through muscularis mucosae). | |||
T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |||
T2 = Tumor invades the muscularis propria. | |||
T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |||
T4 = Tumor invades the visceral peritoneum or invades or adheres to adjacent organ or structure. | |||
–T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |||
–T4b = Tumor directly invades or adheres to adjacent organs or structures. | |||
NX = Regional lymph nodes cannot be assessed. | |||
N0 = No regional lymph node metastasis. | |||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1a = One regional lymph node is positive. | |||
–N1b = Two or three regional lymph nodes are positive. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
N2 = Four or more regional nodes are positive. | |||
–N2a = Four to six regional lymph nodes are positive. | |||
–N2b = Seven or more regional lymph nodes are positive. | |||
M1a = Metastasis to one site or organ is identified without peritoneal metastasis. | |||
IVB | Any T, Any N, M1b | Any T = See T descriptions above in Any T, Any N, M1a TNM stage group. | |
Any N = See N descriptions above in Any T, Any N1, M1a TNM stage group. | |||
M1b = Metastasis to two or more sites or organs is identified without peritoneal metastasis. | |||
IVC | Any T, Any N, M1c | Any T = See T descriptions above in Any T, Any N, M1a TNM stage group. | |
Any N = See N descriptions above in Any T, Any N1, M1a TNM stage group. | |||
M1c = Metastasis to the peritoneal surface is identified alone or with other site or organ metastases. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
The explanations for superscripts b and c are at the end of Table 5. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
b Direct invasion in T4 includes invasion of other organs or other segments of the colorectum as a result of direct extension through the serosa, as confirmed on microscopic examination (e.g., invasion of the sigmoid colon by a carcinoma of the cecum) or, for cancers in a retroperitoneal or subperitoneal location, direct invasion of other organs or structures by virtue of extension beyond the muscularis propria (i.e., respectively, a tumor on the posterior wall of the descending colon invading the left kidney or lateral abdominal wall; or a mid or distal rectal cancer with invasion of prostate, seminal vesicles, cervix, or vagina). | |||
cTumor that is adherent to other organs or structures, grossly, is classified cT4b. However, if no tumor is present in the adhesion, microscopically, the classification should be pT1-4a depending on the anatomical depth of wall invasion. The V and L classification should be used to identify the presence or absence of vascular or lymphatic invasion whereas the PN prognostic factor should be used for perineural invasion. |
Stage | TNM | Definition | Illustration |
---|---|---|---|
IVA | Any T, Any N, M1a | TX = Primary tumor cannot be assessed. | |
T0 = No evidence of primary tumor. | |||
Tis = Carcinoma in situ, intramucosal carcinoma (involvement of lamina propria with no extension through muscularis mucosae). | |||
T1 = Tumor invades the submucosa (through the muscularis mucosa but not into the muscularis propria). | |||
T2 = Tumor invades the muscularis propria. | |||
T3 = Tumor invades through the muscularis propria into pericolorectal tissues. | |||
T4 = Tumor invades the visceral peritoneum or invades or adheres to adjacent organ or structure. | |||
–T4a = Tumor invades through the visceral peritoneum (including gross perforation of the bowel through tumor and continuous invasion of tumor through areas of inflammation to the surface of the visceral peritoneum). | |||
–T4b = Tumor directly invades or adheres to adjacent organs or structures. | |||
NX = Regional lymph nodes cannot be assessed. | |||
N0 = No regional lymph node metastasis. | |||
N1 = One to three regional lymph nodes are positive (tumor in lymph nodes measuring ≥0.2 mm), or any number of tumor deposits are present and all identifiable lymph nodes are negative. | |||
–N1a = One regional lymph node is positive. | |||
–N1b = Two or three regional lymph nodes are positive. | |||
–N1c = No regional lymph nodes are positive, but there are tumor deposits in the subserosa, mesentery, or nonperitonealized pericolic, or perirectal/mesorectal tissues. | |||
N2 = Four or more regional nodes are positive. | |||
–N2a = Four to six regional lymph nodes are positive. | |||
–N2b = Seven or more regional lymph nodes are positive. | |||
M1a = Metastasis to one site or organ is identified without peritoneal metastasis. | |||
IVB | Any T, Any N, M1b | Any T = See T descriptions above in Any T, Any N, M1a TNM stage group. | |
Any N = See N descriptions above in Any T, Any N1, M1a TNM stage group. | |||
M1b = Metastasis to two or more sites or organs is identified without peritoneal metastasis. | |||
IVC | Any T, Any N, M1c | Any T = See T descriptions above in Any T, Any N, M1a TNM stage group. | |
Any N = See N descriptions above in Any T, Any N1, M1a TNM stage group. | |||
M1c = Metastasis to the peritoneal surface is identified alone or with other site or organ metastases. | |||
T = primary tumor; N = regional lymph nodes; M = distant metastasis. | |||
aReprinted with permission from AJCC: Colon and rectum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 251–74. | |||
b Direct invasion in T4 includes invasion of other organs or other segments of the colorectum as a result of direct extension through the serosa, as confirmed on microscopic examination (e.g., invasion of the sigmoid colon by a carcinoma of the cecum) or, for cancers in a retroperitoneal or subperitoneal location, direct invasion of other organs or structures by virtue of extension beyond the muscularis propria (i.e., respectively, a tumor on the posterior wall of the descending colon invading the left kidney or lateral abdominal wall; or a mid or distal rectal cancer with invasion of prostate, seminal vesicles, cervix, or vagina). | |||
cTumor that is adherent to other organs or structures, grossly, is classified cT4b. However, if no tumor is present in the adhesion, microscopically, the classification should be pT1-4a depending on the anatomical depth of wall invasion. The V and L classification should be used to identify the presence or absence of vascular or lymphatic invasion whereas the PN prognostic factor should be used for perineural invasion. |
因为直肠癌局部复发的风险较高,因此,直肠癌的治疗方法与结肠癌有所不同。直肠癌的总体预后较差。两者治疗上,在手术、放疗、化疗给药方式方面具有差异。除了明确直肠癌手术的目的(即治疗性或姑息性)外,还应考虑恢复或维持肛门括约肌、泌尿生殖系统的生理功能及性功能等相关的治疗问题。
直肠癌的临床治疗需要多学科治疗模式,涉及多学科专家团队,包括胃肠病学、临床肿瘤学、肿瘤外科学、放射肿瘤学和影像学等科室。
标准治疗方法 | ||
---|---|---|
治疗方法 | ||
临床0期直肠癌 | ||
临床I 期直肠癌 | ||
临床II和III期直肠癌 | ||
治疗方法 | ||
临床IV期和复发性直肠癌 | ||
二线化疗 | ||
肝转移 | ||
肝切除后肝内动脉灌注化疗 |
治疗方法 | ||
---|---|---|
临床IV期和复发性直肠癌 | ||
二线化疗 | ||
肝转移 | ||
肝切除后肝内动脉灌注化疗 |
直肠癌的主要治疗方法是切除原发肿瘤病灶。手术可根据以下情况而有所调整:
手术类型包括:
单纯息肉切除术可用于部分(T1)病例,如息肉伴侵袭性癌,可完整切除,手术切缘阴性,组织学特征良好。
针对T1期肿瘤的局部切除术已被临床用于一部分符合适应症的患者。对于其他所有肿瘤,直肠系膜切除术是首选的治疗方法。只有非常少的T2期肿瘤患者才能行局部切除术。直肠切除联合全直肠系膜切除术(低位/中位肿瘤的全直肠系膜切除术和高位肿瘤的直肠系膜切除术,切缘距离肿瘤下方>5cm)的术后复发率为4%-8%。
针对中上段直肠晚期癌症患者,应选择低位前切术并行结直肠吻合。针对局部晚期直肠癌,应行根治性切除术,而经低位前切除的全直肠系膜切除术联合自主神经保留术优于经腹会阴联合切除术。
因为直肠系膜切除术后的局部肿瘤复发率低,一些研究者对术后的常规辅助放疗提出了质疑。由于直肠癌首次复发只是在局部位置,围手术期放疗对直肠癌的作用大于结肠癌。
直肠癌的新辅助治疗指术前放化疗,是临床II期和III期患者的首选治疗方案。然而,针对II期或III期直肠癌患者,术后放化疗也是一种治疗手段。
[循证依据等级:1iiA]
基于以下多项研究结果,术前放化疗已成为T3-T4或淋巴结阳性(临床II和III期)患者的标准治疗方法:
多项II和III期临床试验对术前放化疗的临床意义进行研究,包括:
术前放化疗的病理完全缓解率为10%-25%。
然而,与单纯手术相比,术前放疗可升高并发症的发生率;一些局部复发风险较低的癌症患者,采用手术和辅助化疗可能就已经足够。
(有关这些研究的更多信息,请参考本摘要“临床II和III期直肠癌”部分中的术前放化疗部分相关内容。)
术前放化疗是目前临床II和III期直肠癌的治疗标准。然而,在1990年以前,已有研究观察到术后多学科治疗可改善无疾病生存期(DFS)和总体生存期(OS),研究包括:
随后的研究拟通过改善放射增敏作用、寻找最佳化疗药物和改进用药途径来延长患者的生存期。
5-FU:以下研究对5-FU作为辅助治疗的最佳用药方式进行评估:
(有关这些研究的更多信息,请参考本摘要“临床II和III期直肠癌”部分相关内容。)
本研究排除已被临床接受的Ⅱ和Ⅲ期直肠癌术后放化疗方案,包括盆腔放疗(45 Gy-55 Gy) 期间,持续静脉输注5-FU以及4个周期的5-FU 静脉推注联合/不联合亚叶酸钙(LV)辅助维持化疗。
NSABP-R-01试验将单纯手术与手术联合术后放化疗进行对比研究。
随后,NSABP-R-02的研究探讨了在术后化疗中加入放疗是否能提高R-01研究报道的生存期优势。
[循证依据等级:1iiA]
在NSABP-R-02研究中,联用放疗显著降低了5年局部复发率(化疗联合放疗和仅化疗分别为8%和13%,P=0.02),但对生存期无明显影响。放疗或许能延长60岁以下已行经腹会阴联合切除术患者的生存期。
本项试验在肿瘤界引发了关于术后放疗临床意义的讨论,但由于局部复发导致的严重并发症,摒弃放疗似乎还为时尚早。
表8描述了用于直肠癌治疗的化疗方案。
方案名称 | 联合应用的药物 | 剂量 |
---|---|---|
AIO 或德国AIO | 叶酸,亦称为亚叶酸钙,5-FU、伊立替康 | 伊立替康(100 mg/m2)和亚叶酸钙(500 mg/m2),静脉滴注2小时,第1天;随后5-FU(2000 mg/m2)静脉推注,输液泵持续静脉输注24小时,每年4次(52周)。 |
CAPOX | 卡培他滨联合奥沙利铂 | 卡培他滨(1000 mg/m2),每日2次,第1-14天;奥沙利铂(70 mg/m2),第1天和第8天,每3周重复。 |
Douillard | 亚叶酸钙、5-FU、伊立替康 | 伊立替康(180 mg/m2),静脉滴注2小时,第1天;亚叶酸钙(200 mg/m2),静脉滴注2小时,第1天和第2天。随后负荷剂量5-FU(400 mg/m2)静脉推注,然后 5-FU(600 mg/m2)输液泵持续静脉输注22小时,第1天和第2天,每2周重复。 |
FOLFIRI | 亚叶酸钙 | 伊立替康(180 mg/m2)和亚叶酸钙(400 mg/m2),静脉滴注2小时,第1天;随后负荷剂量5-FU(400 mg/m2)静脉推注,第1天;然后 5-FU(2400-3000 mg/m2)输液泵持续静脉输注46小时,每2周重复。 |
FOLFOX-4 | 亚叶酸钙 | 奥沙利铂(85 mg/m2),静脉滴注2小时,第1天;亚叶酸钙(200 mg/m2),静脉滴注2小时,第1天和第2天。随后负荷剂量5-FU(400 mg/m2)静脉推注,然后 5-FU(600 mg/m2)输液泵持续静脉输注22小时,第1天和第2天,每2周重复。 |
FOLFOX-6 | 奥沙利铂、亚叶酸钙、5-FU | 奥沙利铂(85-100 mg/m2)、亚叶酸钙(400 mg/m2),静脉滴注2小时,第1天。随后负荷剂量5-FU(400 mg/m2)静脉推注,第1天;然后 5-FU(2400-3000 mg/m2)输液泵持续静脉输注46小时,每2周重复。 |
FOLFOXIRI | 伊立替康、奥沙利铂、亚叶酸钙、5-FU | 伊立替康(165 mg/m2)静脉滴注60分钟;同步滴注奥沙利铂(85mg/m2)和亚叶酸钙(200 mg/m2),静脉滴注超过120分钟;随后 5-FU(3200 mg/m2)静脉输注48小时。 |
FUFOX | 5-FU、亚叶酸钙、奥沙利铂 | 奥沙利铂(50 mg/m2)和亚叶酸钙(500 mg/m2)联用5-FU(2000 mg/m2)静脉输注22小时,第1、8、22、29天。每36天重复。 |
FUOX | 5-FU联合奥沙利铂 | 5-FU(2250 mg/m2)静脉输注48小时,第1、8、15、22、29、36天。联用奥沙利铂(85mg/m2),第1、15、29天,每6周重复。 |
IFL (or Saltz) | 伊立替康、5-FU、亚叶酸钙 | 伊立替康(125 mg/m2)联用5-FU(500 mg/m2)静脉推注,亚叶酸钙(20 mg/m2)静脉推注,周疗,连用4周,休息2周,每6周重复。 |
XELOX | 卡培他滨联合奥沙利铂 | 口服卡培他滨(1000 mg/m2),每日2次,连用14天;奥沙利铂(130 mg/m2),第1天,每3周重复。 |
5-FU = 5-氟尿嘧啶;AIO =肿瘤内科工作组; bid = 每日2次; IV = 静脉给药;LV = 亚叶酸钙 |
直肠癌盆腔放疗的急性不良反应主要是胃肠道毒反应,具有自限性,一般在治疗结束后4-6周内消失。
更令人担忧的是直肠癌治疗后有迟发性并发症的可能。行积极手术的直肠癌患者可能会出现慢性综合征,尤其在肛门括约肌受损的情况下。
与单纯手术相比,放疗增加了慢性肠功能障碍、肛门括约肌功能障碍(如果实施了保肛术)和性功能障碍的发生率。
对术后放化疗患者进行分析,结果显示放化疗组患者的慢性肠功能障碍发生率高于单纯手术组。
Cochrane综述强调了放疗会增加再次手术风险,并增高迟发性直肠和性功能障碍风险。
改进的放疗计划和技术可以最大限度地减少与治疗相关的急性和迟发性并发症。这些技术包括:
在欧洲,常采用术前放疗(5 Gy×5天)并于1周后行手术的治疗模式,而不是美国使用的长程放化疗。其中一个原因可能是,在美国担心高剂量照射可能会加重迟发不良反应。
波兰一项研究将316名患者随机分为术前长程放化疗组(50.4Gy,28次;5-FU/LV组 )和术前短程放疗组(25 Gy,5次)。
尽管主要终点是保肛,但长程组和短程组的迟发不良反应无显著差异(长程组7%,短疗程10%)。值得注意的是,未见肛门括约肌和性功能方面的相关数据。不良反应由医生决定,而不是患者本人。
比较术前和术后辅助放化疗的临床试验,除应进一步阐明两种方法对患者常规主要终点DFS和OS的影响外,还应探索不同治疗方法对肠道功能和其他重要关于生活质量方面的作用(如保肛)。
The management of rectal cancer varies somewhat from that of colon cancer because of the increased risk of local recurrence and a poorer overall prognosis. Differences include surgical technique, the use of radiation therapy, and the method of chemotherapy administration. In addition to determining the intent of rectal cancer surgery (i.e., curative or palliative), it is important to consider therapeutic issues related to the maintenance or restoration of normal anal sphincter, genitourinary function, and sexual function.
The approach to the management of rectal cancer is multimodal and involves a multidisciplinary team of cancer specialists with expertise in gastroenterology, medical oncology, surgical oncology, radiation oncology, and radiology.
Standard Treatment Options | ||
---|---|---|
Treatment Options | ||
Stage 0 Rectal Cancer | ||
Stage I Rectal Cancer | ||
Stages II and III Rectal Cancer | ||
Treatment Options | ||
Stage IV and Recurrent Rectal Cancer | ||
Second-line chemotherapy | ||
Liver Metastases | ||
Intra-arterial chemotherapy after liver resection |
Treatment Options | ||
---|---|---|
Stage IV and Recurrent Rectal Cancer | ||
Second-line chemotherapy | ||
Liver Metastases | ||
Intra-arterial chemotherapy after liver resection |
The primary treatment for patients with rectal cancer is surgical resection of the primary tumor. The surgical approach to treatment varies according to the following:
Types of surgical resection include the following:
Polypectomy alone may be used in certain instances (T1) in which polyps with invasive cancer can be completely resected with clear margins and have favorable histologic features.
Local excision of clinical T1 tumors is an acceptable surgical technique for appropriately selected patients. For all other tumors, a mesorectal excision is the treatment of choice. Very select patients with T2 tumors may be candidates for local excision. Local failure rates in the range of 4% to 8% after rectal resection with appropriate mesorectal excision (total mesorectal excision for low/middle rectal tumors and mesorectal excision at least 5 cm below the tumor for high rectal tumors) have been reported.
For patients with advanced cancers of the mid- to upper rectum, low-anterior resection followed by the creation of a colorectal anastomosis may be the treatment of choice. For locally advanced rectal cancers for which radical resection is indicated, however, total mesorectal excision with autonomic nerve preservation techniques via low-anterior resection is preferable to abdominoperineal resection.
The low incidence of local relapse after meticulous mesorectal excision has led some investigators to question the routine use of adjuvant radiation therapy. Because of an increased tendency for first failure in locoregional sites only, the impact of perioperative radiation therapy is greater in rectal cancer than in colon cancer.
Neoadjuvant therapy for rectal cancer, using preoperative chemoradiation therapy, is the preferred treatment option for patients with stages II and III disease. However, postoperative chemoradiation therapy for patients with stage II or III rectal cancer remains an acceptable option.
[Level of evidence: 1iA]
Preoperative chemoradiation therapy has become the standard of care for patients with clinically staged T3–T4 or node-positive disease (stages II/III), based on the results of several studies:
Multiple phase II and III studies examined the benefits of preoperative chemoradiation therapy, which include the following:
Complete pathologic response rates of 10% to 25% may be achieved with preoperative chemoradiation therapy.
However, preoperative radiation therapy is associated with increased complications compared with surgery alone; some patients with cancers at a lower risk of local recurrence might be adequately treated with surgery and adjuvant chemotherapy.
(Refer to the Preoperative chemoradiation therapy section in the Stages II and III Rectal Cancer section of this summary for more information about these studies.)
Preoperative chemoradiation therapy is the current standard of care for stages II and III rectal cancer. However, before 1990, the following studies noted an increase in both disease-free survival (DFS) and overall survival (OS) with the use of postoperative combined-modality therapy:
Subsequent studies have attempted to increase the survival benefit by improving radiation sensitization and by identifying the optimal chemotherapeutic agents and delivery systems.
Fluorouracil (5-FU): The following studies examined optimal delivery methods for adjuvant 5-FU:
(Refer to the Stages II and III Rectal Cancer section of this summary for detailed information about these study results.)
Acceptable postoperative chemoradiation therapy for patients with stage II or III rectal cancer not enrolled in clinical trials includes continuous-infusion 5-FU during 45 Gy to 55 Gy pelvic radiation and four cycles of adjuvant maintenance chemotherapy with bolus 5-FU with or without modulation with leucovorin (LV).
Findings from the NSABP-R-01 trial compared surgery alone with surgery followed by chemotherapy or radiation therapy.
Subsequently, the NSABP-R-02 study, addressed whether adding postoperative radiation therapy to chemotherapy would enhance the survival advantage reported in R-01.
[Level of evidence: 1iiA]
In the NSABP-R-02 study, the addition of radiation therapy significantly reduced local recurrence at 5 years (8% for chemotherapy and radiation vs. 13% for chemotherapy alone, P = .02) but failed to demonstrate a significant survival benefit. Radiation therapy appeared to improve survival among patients younger than 60 years and among patients who underwent abdominoperineal resection.
While this trial has initiated discussion in the oncologic community about the proper role of postoperative radiation therapy, omission of radiation therapy seems premature because of the serious complications of locoregional recurrence.
Table 8 describes the chemotherapy regimens used to treat rectal cancer.
Regimen Name | Drug Combination | Dose |
---|---|---|
AIO or German AIO | Folic acid, also known as LV, 5-FU, and irinotecan | Irinotecan (100 mg/m2) and LV (500 mg/m2) administered as 2-h infusions on d 1, followed by 5-FU (2,000 mg/m2) IV bolus administered via ambulatory pump weekly over 24 h, 4 times a y (52 wk). |
CAPOX | Capecitabine and oxaliplatin | Capecitabine (1,000 mg/m2) bid on d 1–14, plus oxaliplatin (70 mg/m2) on d 1 and 8 every 3 wk. |
Douillard | Folic acid, 5-FU, and irinotecan | Irinotecan (180 mg/m2) administered as a 2-h infusion on d 1, LV (200 mg/m2) administered as a 2-h infusion on d 1 and 2, followed by a loading dose of 5-FU (400 mg/m2) IV bolus, then 5-FU (600 mg/m2) administered via ambulatory pump over 22 h every 2 wk on d 1 and 2. |
FOLFIRI | LV, 5-FU, and irinotecan | Irinotecan (180 mg/m2) and LV (400 mg/m2) administered as 2-h infusions on d 1, followed by a loading dose of 5-FU (400 mg/m2) IV bolus administered on d 1, then 5-FU (2,400–3,000 mg/m2) administered via ambulatory pump over 46 h every 2 wk. |
FOLFOX4 | Oxaliplatin, LV, and 5-FU | Oxaliplatin (85 mg/m2) administered as a 2-h infusion on day 1, LV (200 mg/m2) administered as a 2-h infusion on d 1 and 2, followed by a loading dose of 5-FU (400 mg/m2) IV bolus, then 5-FU (600 mg/m2) administered via ambulatory pump over 22 h every 2 wk on d 1 and 2. |
FOLFOX6 | Oxaliplatin, LV, and 5-FU | Oxaliplatin (85–100 mg/m2) and LV (400 mg/m2) administered as 2-h infusions on d 1, followed by a loading dose of 5-FU (400 mg/m2) IV bolus on d 1, then 5-FU (2,400–3,000 mg/m2) administered via ambulatory pump over 46 h every 2 wk. |
FOLFOXIRI | Irinotecan, oxaliplatin, LV, 5-FU | Irinotecan (165 mg/m2) administered as a 60-min infusion, then concomitant infusion of oxaliplatin (85 mg/m2) and LV (200 mg/m2) over 120 min, followed by 5-FU (3,200 mg/m2) administered as a 48-h continuous infusion. |
FUFOX | 5-FU, LV, and oxaliplatin | Oxaliplatin (50 mg/m2) plus LV (500 mg/m2) plus 5-FU (2,000 mg/m2) administered as a 22-h continuous infusion on d 1, 8, 22, and 29 every 36 d. |
FUOX | 5-FU plus oxaliplatin | 5-FU (2,250 mg/m2) administered as a continuous infusion over 48 h on d 1, 8, 15, 22, 29, and 36 plus oxaliplatin (85 mg/m2) on d 1, 15, and 29 every 6 wk. |
IFL (or Saltz) | Irinotecan, 5-FU, and LV | Irinotecan (125 mg/m2) plus 5-FU (500 mg/m2) IV bolus and LV (20 mg/m2) IV bolus administered weekly for 4 out of 6 wk. |
XELOX | Capecitabine plus oxaliplatin | Oral capecitabine (1,000 mg/m2) administered bid for 14 d plus oxaliplatin (130 mg/m2) on d 1 every 3 wk. |
5-FU = fluorouracil; AIO = Arbeitsgemeinschaft Internistische Onkologie; bid = twice a day; IV = intravenous; LV = leucovorin. |
The acute side effects of pelvic radiation therapy for rectal cancer are mainly the result of gastrointestinal toxicity, are self-limiting, and usually resolve within 4 to 6 weeks of completing treatment.
Of greater concern is the potential for late morbidity after rectal cancer treatment. Patients who undergo aggressive surgical procedures for rectal cancer can have chronic symptoms, particularly if there is impairment of the anal sphincter.
Patients treated with radiation therapy appear to have increased chronic bowel dysfunction, anorectal sphincter dysfunction (if the sphincter was surgically preserved), and sexual dysfunction than do patients who undergo surgical resection alone.
An analysis of patients treated with postoperative chemotherapy and radiation therapy suggests that these patients may have more chronic bowel dysfunction than do patients who undergo surgical resection alone.
A Cochrane review highlights the risks of increased surgical morbidity as well as late rectal and sexual function in association with radiation therapy.
Improved radiation therapy planning and techniques may minimize these acute and late treatment-related complications. These techniques include the following:
In Europe, it is common to deliver preoperative radiation therapy alone in one week (5 Gy × five daily treatments) followed by surgery one week later, rather than the long-course chemoradiation approach used in the United States. One reason for this difference is the concern in the United States for heightened late effects when high radiation doses per fraction are given.
A Polish study randomly assigned 316 patients to preoperative long-course chemoradiation therapy (50.4 Gy in 28 daily fractions with 5-FU/LV) or short-course preoperative radiation therapy (25 Gy in 5 fractions).
Although the primary endpoint was sphincter preservation, late toxicity was not statistically significantly different between the two treatment approaches (7% long course vs. 10% short course). Of note, data on anal sphincter and sexual function were not reported, and toxicity was physician determined, not patient reported.
Ongoing clinical trials comparing preoperative and postoperative adjuvant chemoradiation therapy should further clarify the impact of either approach on bowel function and other important quality-of-life issues (e.g., sphincter preservation) in addition to the more conventional endpoints of DFS and OS.
0期直肠癌或原位癌是肿瘤最为表浅的一类直肠病变,并且局限于黏膜层,未侵犯固有层。
临床0期直肠癌的治疗的标准治疗方法包括:
临床0期直肠癌可采用局部切除术或单纯息肉切除术。
由于其局限性特点,临床0期直肠癌治愈率较高。对于不能行局部切除的大病灶,可行经肛门或经阴道全层直肠切除术。
采用我们的临床试验搜索引擎,可查询正招募患者的NCI支持的癌症临床试验。搜索可按试验地点、治疗类型、药物名称及其他标准进行设置。还有一些临床试验相关的基本信息。
Stage 0 rectal cancer or carcinoma in situ is the most superficial of all rectal lesions and is limited to the mucosa without invasion of the lamina propria.
Standard treatment options for stage 0 rectal cancer include the following:
Local excision or simple polypectomy may be indicated for stage 0 rectal cancer tumors.
Because of its localized nature at presentation, stage 0 rectal cancer has a high cure rate. For large lesions not amenable to local excision, full-thickness rectal resection by the transanal or transcoccygeal route may be performed.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
临床I期肿瘤在黏膜下侵犯至黏膜下层(T1)或侵犯但未穿透肠壁肌层(T2)。由于病灶部位较为局限,临床I期直肠癌治愈率较高。
临床I期直肠癌的标准治疗包括:
临床I期直肠癌的手术治疗,有三种可能的方式:
病理分期T1的患者可能不需要术后治疗。T2及以上肿瘤或伴20%转移淋巴结的患者,应考虑联合其他治疗,如放疗和化疗或广泛直肠切除术。
组织学分型差或局部切除后切缘阳性,可考虑低位前切除术或经腹会阴联合切除术,应按完整外科分期进行术后治疗。
对于T1和T2肿瘤患者,尚未有随机临床试验对局部切除术联合/不联合术后放化疗与广泛手术切除(低前位切除术和腹会阴联合切除术)进行对比研究。
循证依据(手术):
采用我们的临床试验搜索引擎,可查询正招募患者的NCI支持的癌症临床试验。搜索可按试验地点、治疗类型、药物名称及其他标准进行设置。还有一些临床试验相关的基本信息。
Stage I tumors extend beneath the mucosa into the submucosa (T1) or into, but not through, the bowel muscle wall (T2). Because of its localized nature at presentation, stage I rectal cancer has a high cure rate.
Standard treatment options for stage I rectal cancer include the following:
There are three potential options for surgical resection in stage I rectal cancer:
Patients with tumors that are pathologically T1 may not need postoperative therapy. Patients with tumors that are T2 or greater have lymph node involvement about 20% of the time. Patients may want to consider additional therapy, such as radiation therapy and chemotherapy, or wide surgical resection of the rectum.
Patients with poor histologic features or positive margins after local excision may consider low-anterior resection or abdominoperineal resection and postoperative treatment as dictated by full surgical staging.
For patients with T1 and T2 tumors, no randomized trials are available to compare local excision with or without postoperative chemoradiation therapy to wide surgical resection (low-anterior resection and abdominoperineal resection).
Evidence (surgery):
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
临床II和III期直肠癌的标准治疗包括:
临床II和III期直肠癌患者可在放化疗前后行低位前切除或经腹会阴联合切除联合全直肠系膜切除术。
回顾性研究表明,部分病理T3、N0患者虽然单纯采用手术治疗而未予任何其他治疗,但其局部和全身复发的风险仍非常低。
基于数项研究的结果,术前放化疗已成为临床T3或T4或淋巴结阳性的直肠癌患者的标准治疗。
循证依据(术前放化疗):
在欧洲和澳大利亚部分地区,术前短程放疗已成为临床的标准治疗。
循证依据(术前短程放疗)
随后,波兰直肠临床试验和Trans-Tasman放射肿瘤学组(TROG)比较了术前的短程放疗和标准长程联合5-FU化疗的临床疗效。
综合考虑这些研究结果表明,术前的短程放疗和长程放化疗均可用于临床II期或III期直肠癌。
由于全身化疗与放疗的结合与两种治疗手段不断改良,促进了术后辅助治疗的发展。现有一系列前瞻性随机临床试验对术后放疗联合5-FU为基础的化疗在II期和III期直肠癌的应用进行研究,包括:
[循证依据等级:1iiA]
这些研究结果显示,术后采用放化疗联合治疗,可延长患者的无疾病进展间隔期和总体生存期。这一结果发表于1990年,在国家癌症研究所主办的共识发展会议上,专家建议将术后联合治疗用于临床II和III期直肠癌。
此后,术前放化疗成为标准治疗。但术后放化疗仍是一个替代治疗方法。(有关更多信息,请参阅本摘要“术前放化疗部分”相关内容。)
其他证据(术后放化疗)
许多肿瘤学家将亚叶酸钙/5-FU/奥沙利铂(FOLFOX)作为直肠癌的标准辅助化疗。然而,目前还没有关于直肠癌的数据支持这一观点。在最新的直肠癌辅助治疗的临床试验中,FOLFOX已成为标准治疗。美国东部肿瘤协作组的临床试验(ECOG-E5202[NCT00217737])将术前或术后放化疗经治的临床II期或III期直肠癌患者随机分配,给予6个月FOLFOX单方案或FOLFOX联合贝伐单抗治疗。但由于进度过慢,该研究提早关闭,无相关数据。
在临床前模型,奥沙利铂已显示出放射增敏性。
有关联合奥沙利铂和氟嘧啶为基础的化疗II期临床试验曾报道病理完全缓解率为14%-30%。
多个研究的数据表明,病理完全缓解率与其他主要终点之间具有相关性,如无远处转移生存率、DFS和OS。
奥沙利铂联合放疗同步用于直肠癌患者中的非试验性临床价值,目前尚不清楚。
循证依据(术前奥沙利铂联合放化疗):
本研究的主要目的是控制局部区域肿瘤控制。
[循证依据等级:1ID] 本研究的初步结果以摘要形式发表在2011年美国临床肿瘤学会年会上,包括:
根据多项研究的结果,作为一种放射增敏剂,奥沙利铂在原发性肿瘤治疗中无任何临床意义,或与治疗导致的急性不良反应增加有关。是否应将奥沙利铂添加到5-FU/LV辅助治疗中用于临床II期和III期直肠癌患者的术后治疗,尚未有定论。目前,还没有随机的III期研究支持奥沙利铂辅助治疗直肠癌。然而,在5-FU/LV方案中加入奥沙利铂作为辅助治疗已成为结肠癌的标准治疗。
循证依据(术前奥沙利铂)
这些结肠癌的研究结果能否适用于直肠癌还不明确。目前还没有随机的III期研究支持FOLFOX作为常规辅助治疗用于直肠癌。
自从直肠癌术前放化疗出现以来,标准治疗方法一直是推荐经腹会阴联合切除术或腹腔镜辅助直肠癌切除术进行手术切除。在大多数系列研究中,经过长程放化疗,10%-20%患者获得完全临床缓解,即通过影像学检查、直肠指检或乙状结肠镜检查未发现肿瘤征象。长期以来,人们一直认为,大多数因个人或医疗原因未接受手术的患者会出现局部和/或全身复发。然而,术前采用放化疗随后手术,病理结果为完全缓解的患者,其无疾病生存期优于未见病理完全缓解的患者。
少数单一机构的研究对这一标准治疗表达了抗议,提出大多数具有临床完全缓解的患者即使不经手术也能治愈直肠癌。那么,许多患者在局部肿瘤复发时,便可通过手术(经腹会阴联合切除或腹腔镜辅助切除)予以切除。
这些系列研究因其规模较小和固有的的选择偏见而受到阻碍。
证据(初始放化疗获得完全临床缓解后,需密切随诊监测):
观察组患者采取了更密切的随诊方案,包括门诊直肠指诊、核磁共振成像(前2年,每4-6个月复查);麻醉下检查或内镜检查;胸部、腹部和盆腔CT;在最初的2年里,至少进行2次癌胚抗原(CEA)检测。目前尚无最佳随诊方案。
对于治疗后出现临床完全缓解的患者,可以考虑采用密切监测方法,而不是立即手术切除。
采用我们的临床试验搜索引擎,可查询正招募患者的NCI支持的癌症临床试验。搜索可按试验地点、治疗类型、药物名称及其他标准进行设置。还有一些临床试验相关的基本信息。
Standard treatment options for stages II and III rectal cancer include the following:
Total mesorectal excision with either low anterior resection or abdominoperineal resection is usually performed for stages II and III rectal cancer before or after chemoradiation therapy.
Retrospective studies have demonstrated that some patients with pathological T3, N0 disease treated with surgery and no additional therapy have a very low risk of local and systemic recurrence.
Preoperative chemoradiation therapy has become the standard of care for patients with clinically staged T3 or T4 or node-positive disease, based on the results of several studies.
Evidence (preoperative chemoradiation therapy):
The use of short-course radiation therapy before surgery has been a standard approach in parts of Europe and Australia.
Evidence (short-course preoperative radiation therapy):
Subsequently, the Polish Rectal Trial and the Trans-Tasman Radiation Oncology Group (TROG) compared short-course preoperative radiation therapy with the standard long-course preoperative chemoradiation therapy administered with 5-FU.
Taken together, these studies demonstrate that short-course preoperative radiation therapy and long-course preoperative chemoradiation therapy are both reasonable treatment strategies for patients with stage II or III rectal adenocarcinoma.
Progress in the development of postoperative treatment regimens relates to the integration of systemic chemotherapy and radiation therapy, as well as redefining the techniques for both modalities. The efficacy of postoperative radiation therapy and 5-FU-based chemotherapy for stages II and III rectal cancer was established by a series of prospective, randomized clinical trials, including the following:
[Level of evidence: 1iiA]
These studies demonstrated an increase in DFS interval and OS when radiation therapy was combined with chemotherapy after surgical resection. After the publication in 1990 of the results of these trials, experts at a National Cancer Institute-sponsored Consensus Development Conference recommended postoperative combined-modality treatment for patients with stages II and III rectal carcinoma.
Since that time, preoperative chemoradiation therapy has become the standard of care, although postoperative chemoradiation therapy is still an acceptable alternative. (Refer to the Preoperative chemoradiation therapy section of this summary for more information.)
Additional evidence (postoperative chemoradiation therapy):
Many academic oncologists suggest that LV/5-FU/oxaliplatin (FOLFOX) be considered the standard for adjuvant chemotherapy in rectal cancer. However, there are no data about rectal cancer to support this consideration. FOLFOX has become the standard arm in the latest Intergroup study evaluating adjuvant chemotherapy in rectal cancer. An Eastern Cooperative Oncology Group trial (ECOG-E5202 [NCT00217737]) randomly assigned patients with stage II or III rectal cancer who received preoperative or postoperative chemoradiation therapy to receive 6 months of FOLFOX with or without bevacizumab, but this trial closed because of poor accrual; no efficacy data are available.
Oxaliplatin has also been shown to have radiosensitizing properties in preclinical models.
Phase II studies that combined oxaliplatin with fluoropyrimidine-based chemoradiation therapy have reported pathologic complete response rates ranging from 14% to 30%.
Data from multiple studies have demonstrated a correlation between rates of pathologic complete response and endpoints including distant metastasis-free survival, DFS, and OS.
There is no current role for off-trial use of concurrent oxaliplatin and radiation therapy in the treatment of patients with rectal cancer.
Evidence (preoperative oxaliplatin with chemoradiation therapy):
The primary objective of this study is locoregional disease control.
[Level of evidence: 1iiD] Preliminary results, reported in abstract form at the 2011 American Society of Clinical Oncology annual meeting, demonstrated the following:
On the basis of results of several studies, oxaliplatin as a radiation sensitizer does not appear to add any benefit in terms of primary tumor response, and it has been associated with increased acute treatment-related toxicity. The question of whether oxaliplatin should be added to adjuvant 5-FU/LV for postoperative management of stages II and III rectal cancer is an ongoing debate. There are no randomized phase III studies to support the use of oxaliplatin for the adjuvant treatment of rectal cancer. However, the addition of oxaliplatin to 5-FU/LV for the adjuvant treatment of colon cancer is now considered standard care.
Evidence (postoperative oxaliplatin):
It is unclear whether the results of these colon cancer trials can be applied to the management of patients with rectal cancer. There are no randomized phase III studies to support the routine practice of administering FOLFOX as adjuvant therapy to patients with rectal cancer.
Since the advent of preoperative chemoradiation therapy in rectal cancer, the standard approach has been to recommend definitive surgical resection by either abdominoperineal resection or laparoscopic-assisted resection. In most series, after long-course chemoradiation therapy, 10% to 20% of patients will have a complete clinical response in which there is no sign of persistent cancer by imaging, rectal exam, or direct visualization during sigmoidoscopy. It was a long-held belief that most patients who did not undergo surgery for personal or medical reasons would experience a local and/or systemic recurrence. However, it became clear that patients with a pathologic complete response to preoperative chemoradiation therapy followed by definitive surgery had a better DFS than did patients who did not have a pathologic clinical response.
Several single-institution studies have challenged this standard of care by demonstrating that most patients with complete clinical response will be cured of rectal cancer without surgery and that many patients who experience a local recurrence can be treated with surgical resection (abdominoperineal resection or laparoscopic-assisted resection) at the time of their recurrence.
These institutional series were hampered by their small size and inherent selection bias.
Evidence (primary chemoradiation therapy followed by intensive surveillance for complete clinical responders):
Patients managed by watch and wait underwent a more intensive follow-up protocol consisting of outpatient digital rectal examination; MRI (every 4–6 months in the first 2 years); examination under anesthesia or endoscopy; computed tomography scan of the chest, abdomen, and pelvis; and at least two carcinoembryonic antigen measurements in the first 2 years. The optimal follow-up has not been determined.
For patients who have a complete clinical response to therapy, it is reasonable to consider a watch-and-wait approach with intensive surveillance instead of immediate surgical resection.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
制定进展期或复发性直肠癌治疗决策的基础是肿瘤部位
临床IV期和复发性直肠癌的标准治疗包括:
对于局部复发、仅肝或仅肺转移性的患者,如果可行的话,手术切除是唯一的潜在治愈方法
单纯肺转移或者仅有肝肺转移的患者也可以考虑手术切除,严格选择手术适应症可使得病人有望获得5年生存。
与肿瘤复发相关的肾积水往往意味着无法获得手术根治
局部复发的直肠癌可采用手术切除,特别既往手术史较少的患者。已行治愈性切除术的单纯局部肿瘤复发的患者,应采取积极的局部治疗,再次行低位前切除、结肠肛管吻合术、经腹会阴联合切除术、后盆腔或全盆腔清扫术,可获得长期无疾病生存收益。
诱导放化疗用于局部晚期盆腔复发(盆腔侧壁、骶骨和/或邻近器官受累)且既往未行放疗的患者,或能提高手术可切除率和保肛率。
术中放射治疗在接受过外照射治疗的患者中可改善局部复发患者的局部控制率,并发症率也在可接受范围。
下面将介绍一些美国FDA批准的用于治疗转移性结直肠癌的临床药物,可单药使用,也可联合应用,包括:
在其他有效的化疗药上市前,5-FU是唯一一个有效的化疗药物,对局部晚期、不可手术切除、或转移性患者具有一定作用,并能延长患者的疾病进展时间(TTP)
与支持治疗相比,还能改善患者的生存期和生活质量。
多项临床试验对各种5-FU/LV方案(不同剂量和用药方式)的疗效和不良反应进行分析,结果显示在12个月内,中位生存期无明显差异。
有3项针对转移性结直肠癌患者的随机研究结果显示,伊立替康或奥沙利铂联合5-FU/LV应用可提高临床缓解率,改善无进展生存期和总体生存期
循证依据(伊立替康vs.奥沙利铂)
自这些研究发表以来,FOLFOX或FOLFIRI被认为可用于转移性结直肠癌患者的一线治疗。然而,当使用伊立替康作为转移性结直肠癌的一线治疗方案时,FOLFIRI是首选方案。
[循证依据等级:1iiDiii]
在多药联合化疗前,有两项随机试验提示卡培他滨的临床疗效等同于Mayo方案中的5-FU/LV方案。
[循证依据等级:1iiA]
随机III期临床试验结果已提出可采用卡培他滨替代静脉输注5-FU。两项III期临床试验对 5-FU/奥沙利铂 (FUOX) 与卡培他滨/奥沙利铂 (CAPOX)进行对照研究。
循证依据(伊立替康vs.奥沙利铂)
当采用奥沙利铂为基础的方案作为转移性结直肠癌的一线治疗时,CAPOX方案并不劣于FUOX方案。
可在FOLFIRI或FOLFOX方案中联用贝伐珠单抗,用于转移性结直肠癌的一线治疗。
循证依据(贝伐珠单抗):
循证依据(FOLFOXIRI)
西妥昔单抗是一种抗EGFR的部分人源性单克隆抗体。当西妥昔单抗加入含贝伐单抗的多药化疗方案时,KRAS突变型患者的临床预后可能更差。
循证依据(西妥昔单抗)
分别对A和B组以及和A和C组进行对比分析,并将研究成果单独发表。
阿柏西普是一种新型抗VEGF分子制剂,常被用于转移性结直肠癌的二线治疗。
循证依据(阿柏西普)
雷莫芦单抗是一种与血管内皮生长因子受体2结合的完全人源化单克隆抗体。
循证依据(雷莫芦单抗)
帕尼单抗是一种抗EGFR的完全人源化抗体。FDA批准帕尼单抗用于化疗无效的转移性结直肠癌。
在临床试验中,帕尼单抗作为单药或联合治疗均具有一定的临床疗效。该药对患者的PFS和OS影响与西妥昔单抗相似。二者或有一致的药物类效应。
循证依据(帕尼单抗):
在临床IV期结直肠癌患者的治疗中,KRAS野生型患者是否应采用抗EGFR抗体联合化疗或抗VEGF抗体联合化疗,目前尚不清楚。有两项临床试验尝试对此进行研究。
循证依据(抗EGFR抗体与抗VEGF抗体联合一线化疗的对比研究):
根据这两项研究,化疗联合贝伐单抗或西妥昔单抗用于KRAS野生型的转移性结直肠癌患者无明显差异。然而,在KRAS野生型患者的临床治疗中,联用抗EGFR抗体可有效改善患者的总体生存期。
瑞戈非尼是多种酪氨酸激酶途径的抑制剂,包括血管内皮生长因子。2012年9月,FDA批准瑞戈非尼用于既往治疗失败的癌症患者。
循证依据(瑞戈非尼):
TAS-102(Lonsurf)是一种口服的胸腺嘧啶核苷类似物、曲氟尿苷、胸腺嘧啶磷酸化酶抑制剂、盐酸替吡拉西的复合制剂。曲氟尿苷,以三磷酸形式抑制胸苷酸合成酶,具有抗肿瘤作用。盐酸替吡拉西是胸苷磷酸化酶的有效抑制剂,能有效降解曲氟尿苷。曲氟尿苷和替吡拉西结合可升高血浆中曲氟尿苷水平。
循证依据(曲氟尿苷替匹嘧啶):
基于RECOURSE临床试验结果,FDA批准TAS-102用于治疗转移性结直肠癌。
约4%临床IV期结直肠癌患者的肿瘤有微卫星不稳定,这也称为高度微卫星不稳定(MSI-H)。MSI-H表型与MLH1、MSH2、MSH6和PMS2基因的胚系缺陷有关,是遗传性非息肉样结直肠癌(HNPCC)或林奇综合征患者中常见的表型。由于上述某个基因在DNA甲基化后沉默,患者也出现MSI-H表型。可采用分子遗传学检测微卫星不稳定性,在肿瘤组织观察微卫星不稳定性,或者通过免疫组化检测错配修复蛋白的缺失。
2017年5月,FDA批准派姆单抗,即程序性细胞死亡蛋白1(PD-1)抗体,用于治疗微卫星不稳定的肿瘤。
伊立替康作为二线化疗用于5-FU/LV一线治疗的患者。结果显示,与其他静脉输注5-FU或支持治疗相比,伊立替康的二线化疗能改善患者的总体生存期。
同样,还有一项III期临床试验将伊立替康和5-FU/LV经治后出现进展的患者随机给予静推和静脉输注5-FU/LV (LV5FU2)、奥沙利铂单药治疗、FOLFOX-4方案。FOLFOX-4组和LV5FU2组的中位疾病无进展期分别为4.6个月和2.7个月(log-rank检测,二项式 P<0.001)。
[循证依据等级:1iiDiii]
姑息放疗
化疗
放化疗
姑息性、内镜下置入支架有助于缓解梗阻。
约15%-25%结直肠癌患者在确诊时,就已出现肝转移,另有25%-50%患者在原发肿瘤切除术后,会发展为异时性肝转移。
仅有少数肝转移患者具有手术机会。不断改良的肿瘤消融术和局部化疗和全身化疗可作为临床治疗手段,包括:
肝转移可行手术切除,主要基于以下几点:
包括美国中北部癌症治疗组(NCCTG-934653 [NCT00002575])在内的多项非随机的临床试验发现,具有手术可能的肝转移患者,如肿瘤完全切除,其5年生存率可达25%-40%。
[循证依据等级:3iiiDiv] 不断改进的手术技术和术前影像检查可有效筛选手术患者。此外,多项研究显示多药联合化疗用于曾被认为不可手术的孤立的肝转移患者,部分患者可在化疗后行手术治疗。
曾被认定为不可手术切除的肝转移患者,如果临床化疗有效,则仍有手术机会。这些患者的5年生存率与可手术切除的患者无差异。
射频消融作为一种安全的治疗手段(术后并发症发生率2%,死亡率<1%),病人也会获得长期的生存获益
射频消融和和冷冻消融可用于不可手术切除及不宜行肝切除的患者。
辅助化疗在肝转移潜在治愈性手术切除后的作用尚不清楚。
循证依据(辅助化疗)
需要进一步的研究来评估这种治疗方法,并确定是否有更有效的联合化疗方案,仅行全身化疗便可获得与肝动脉灌注化疗联合全身化疗相似的结果。
肝转移患者采用氟脲苷的肝内动脉灌注化疗具有较好的临床疗效。与全身化疗相比,未观察到持续的生存期延长。
针对局部化疗的临床疗效的,一直存有诸多争议。但这也是大规模多中心III期临床试验(Leuk-9481)(NCT00002716)肝动脉灌注与全身化疗的基础。采用肝内动脉灌注化疗联合肝区放疗,特别是利用转移灶的局灶型放疗,正处于临床评估阶段。
多项研究显示肝内动脉灌注化疗可增加局部不良反应,如肝功能异常和致命的胆道硬化症。
采用我们的临床试验搜索引擎,可查询正招募患者的NCI支持的癌症临床试验。搜索可按试验地点、治疗类型、药物名称及其他标准进行设置。还有一些临床试验相关的基本信息。
Treatment of patients with advanced or recurrent rectal cancer depends on the location of the disease.
Standard treatment options for stage IV and recurrent rectal cancer include the following:
For patients with locally recurrent, liver-only, or lung-only metastatic disease, surgical resection, if feasible, is the only potentially curative treatment.
Patients with limited pulmonary metastasis, and patients with both pulmonary and hepatic metastasis, may also be considered for surgical resection, with 5-year survival possible in highly selected patients.
The presence of hydronephrosis associated with recurrence appears to be a contraindication to surgery with curative intent.
Locally recurrent rectal cancer may be resectable, particularly if an inadequate prior operation was performed. For patients with local recurrence alone after an initial, attempted curative resection, aggressive local therapy with repeat low anterior resection and coloanal anastomosis, abdominoperineal resection, or posterior or total pelvic exenteration can lead to long-term disease-free survival.
The use of induction chemoradiation therapy for previously nonirradiated patients with locally advanced pelvic recurrence (pelvic side-wall, sacral, and/or adjacent organ involvement) may increase resectability and allow for sphincter preservation.
Intraoperative radiation therapy in patients who underwent previous external-beam radiation therapy may improve local control in patients with locally recurrent disease, with acceptable morbidity.
The following are active U.S. Food and Drug Administration (FDA)-approved drugs that are used alone and in combination with other drugs for patients with metastatic colorectal cancer:
When 5-FU was the only active chemotherapy drug, trials in patients with locally advanced, unresectable, or metastatic disease demonstrated partial responses and prolongation of the time-to-progression (TTP) of disease,
and improved survival and quality of life in patients who received chemotherapy versus best supportive care.
Several trials have analyzed the activity and toxic effects of various 5-FU/LV regimens using different doses and administration schedules and showed essentially equivalent results with a median survival time in the approximately 12-month range.
Three randomized studies in patients with metastatic colorectal cancer demonstrated improved response rates, progression-free survival (PFS), and overall survival (OS) when irinotecan or oxaliplatin was combined with 5-FU/LV.
Evidence (irinotecan vs. oxaliplatin):
Since the publication of these studies, the use of either FOLFOX or FOLFIRI is considered acceptable for first-line treatment of patients with metastatic colorectal cancer. However, when using an irinotecan-based regimen as first-line treatment of metastatic colorectal cancer, FOLFIRI is preferred.
[Level of evidence: 1iiDiii]
Before the advent of multiagent chemotherapy, two randomized studies demonstrated that capecitabine was associated with equivalent efficacy when compared with the Mayo Clinic regimen of 5-FU/LV.
[Level of evidence: 1iiA]
Randomized phase III trials have addressed the equivalence of substituting capecitabine for infusional 5-FU. Two phase III studies have evaluated capcitabine/oxaliplatin (CAPOX) versus 5-FU/oxaliplatin regimens (FUOX or FUFOX).
Evidence (oxaliplatin vs. capecitabine):
When using an oxaliplatin-based regimen as first-line treatment of metastatic colorectal cancer, a CAPOX regimen is not inferior to a 5-FU/oxaliplatin regimen.
Bevacizumab can reasonably be added to either FOLFIRI or FOLFOX for patients undergoing first-line treatment of metastatic colorectal cancer. There are currently no completed randomized controlled studies evaluating whether continued use of bevacizumab in second-line or third-line treatment after progressing on a first-line bevacizumab regimen extends survival.
Evidence (bevacizumab):
Evidence (FOLFOXIRI):
Cetuximab is a partially humanized monoclonal antibody against EGFR. Importantly, patients with mutant KRAS tumors may experience worse outcome when cetuximab is added to multiagent chemotherapy regimens containing bevacizumab.
Evidence (cetuximab):
The comparisons between arms A and B and arms A and C were analyzed and published separately.
Aflibercept is a novel anti-VEGF molecule and has been evaluated as a component of second-line therapy in patients with metastatic colorectal cancer.
Evidence (aflibercept):
Ramucirumab is a fully humanized monoclonal antibody that binds to vascular endothelial growth factor receptor-2 (VEGFR-2).
Evidence (ramucirumab):
Panitumumab is a fully humanized antibody against the EGFR. The FDA approved panitumumab for use in patients with metastatic colorectal cancer refractory to chemotherapy.
In clinical trials, panitumumab demonstrated efficacy as a single agent or in combination therapy, which was consistent with the effects on PFS and OS with cetuximab. There appears to be a consistent class effect.
Evidence (panitumumab):
In the management of patients with stage IV colorectal cancer, it is unknown whether patients with KRAS wild-type cancer should receive an anti-EGFR antibody with chemotherapy or an anti-VEGF antibody with chemotherapy. Two studies attempted to answer this question.
Evidence (anti-EGFR antibody vs. anti-VEGF antibody with first-line chemotherapy):
On the basis of these two studies, no apparent significant difference is evident about starting treatment with chemotherapy/bevacizumab or chemotherapy/cetuximab in patients with KRAS wild-type metastatic colorectal cancer. However, in patients with KRAS wild-type cancer, administration of an anti-EGFR antibody at some point in the course of management improves OS.
Regorafenib is an inhibitor of multiple tyrosine kinase pathways including VEGF. In September 2012, the FDA granted approval for the use of regorafenib in patients who had progressed on previous therapy.
Evidence (regorafenib):
TAS-102 (Lonsurf) is an orally administered combination of a thymidine-based nucleic acid analog, trifluridine, and a thymidine phosphorylase inhibitor, tipiracil hydrochloride. Trifluridine, in its triphosphate form, inhibits thymidylate synthase; therefore, trifluridine, in this form, has an anti-tumor effect. Tipiracil hydrochloride is a potent inhibitor of thymidine phosphorylase, which actively degrades trifluridine. The combination of trifluridine and tipiracil allows for adequate plasma levels of trifluridine.
Evidence (TAS-102):
TAS-102 was approved by the FDA for the treatment of metastatic colorectal cancer patientsbased on the results of the RECOURSE trial.
Approximately 4% of patients with stage IV colorectal cancer will have tumors that are microsatellite unstable; this designation is also known as microsatellite-high (MSI-H). The MSI-H phenotype is associated with germline defects in the MLH1, MSH2, MSH6, or PMS2 genes, and is the primary phenotype observed in tumors from patients with hereditary nonpolyposis colorectal cancer (HNPCC) or Lynch syndrome. Patients can also have the MSI-H phenotype because one of these genes was silenced via a process called DNA methylation. Testing for microsatellite instability can be done with molecular genetic tests, which look for microsatellite instability in the tumor tissue or with immunohistochemistry, which looks for the loss of mismatch repair proteins.
In May 2017, the FDA granted approval for using pembrolizumab, a programmed cell death protein 1 (PD-1) antibody, in patients with microsatellite unstable tumors.
Second-line chemotherapy with irinotecan in patients treated with 5-FU/LV as first-line therapy demonstrated improved OS when compared with either infusional 5-FU or supportive care.
Similarly, a phase III trial randomly assigned patients who progressed on irinotecan and 5-FU/LV to bolus and infusional 5-FU/LV, single-agent oxaliplatin, or FOLFOX4. The median TTP for FOLFOX4 versus 5-FU/LV was 4.6 months versus 2.7 months (stratified log-rank test, 2-sided P < .001).
[Level of evidence: 1iiDiii]
Palliative radiation therapy,
chemotherapy,
and chemoradiation therapy
may be indicated. Palliative, endoscopically-placed stents may be used to relieve obstruction.
Approximately 15% to 25% of colorectal cancer patients will present with liver metastases at diagnosis, and another 25% to 50% will develop metachronous hepatic metastasis after resection of the primary tumor.
Although only a small proportion of patients with liver metastasis are candidates for surgical resection, advances in tumor ablation techniques and in both regional and systemic chemotherapy administration provide a number of treatment options. These include the following:
Hepatic metastasis may be considered to be resectable based on the following factors:
For patients with hepatic metastasis that is considered to be resectable, a negative margin resection has been associated with 5-year survival rates of 25% to 40% in mostly nonrandomized studies, such as the North Central Cancer Treatment Group trial NCCTG-934653 (NCT00002575).
[Level of evidence: 3iiiDiv] Improved surgical techniques and advances in preoperative imaging have improved patient selection for resection. In addition, multiple studies with multiagent chemotherapy have demonstrated that patients with metastatic disease isolated to the liver, which historically would be considered unresectable, can occasionally be made resectable after the administration of neoadjuvant chemotherapy.
Patients with hepatic metastases that are deemed unresectable will occasionally become candidates for resection if they have a good response to chemotherapy. These patients have 5-year survival rates similar to patients who initially had resectable disease.
Radiofrequency ablation has emerged as a safe technique (2% major morbidity and <1% mortality rate) that may provide long-term tumor control.
Radiofrequency ablation and cryosurgical ablation remain options for patients with tumors that cannot be resected and for patients who are not candidates for liver resection.
The role of adjuvant chemotherapy after potentially curative resection of liver metastases is uncertain.
Evidence (adjuvant chemotherapy):
Additional studies are required to evaluate this treatment approach and to determine whether more effective systemic combination chemotherapy alone would provide results similar to hepatic intra-arterial therapy plus systemic treatment.
Hepatic intra-arterial chemotherapy with floxuridine for liver metastases has produced higher overall response rates but no consistent improvement in survival when compared with systemic chemotherapy.
Controversy regarding the efficacy of regional chemotherapy was the basis of a large multicenter phase III trial (Leuk-9481) (NCT00002716) of hepatic arterial infusion versus systemic chemotherapy. The use of combination intra-arterial chemotherapy with hepatic radiation therapy, especially employing focal radiation of metastatic lesions, is under evaluation.
Increased local toxic effects after hepatic infusional therapy are seen, including liver function abnormalities and fatal biliary sclerosis.
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
PDQ癌症信息定期评估和及时更新最新内容。这一部分会收录相关内容的最新信息(截至更新日期)。
2019年新发病例和死亡病例均采用最新资料(摘自美国癌症学会,见参考文献1)。
本章节的内容更新
本章节的内容更新
编辑对本部分内容进行了修改。
本篇内容由PDQ成人治疗编委会进行撰写和维护,编委会独立于NCI。本篇内容的选取立场公正,不代表NCI和NIH任何政治观点。有关本篇内容的政策及编委会在PDQ维护中的作用等更多信息,请参考PDQ摘要以及PDQ®-NCI综合癌症数据库页面内容。
The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.
Updated statistics with estimated new cases and deaths for 2019 (cited American Cancer Society as reference 2).
Editorial changes were made to this section.
Editorial changes were made to this section.
An editorial change was made to this section.
This summary is written and maintained by the PDQ Adult Treatment Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® - NCI's Comprehensive Cancer Database pages.
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the treatment of rectal 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 Adult Treatment 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 Rectal Cancer Treatment 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 Adult Treatment 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® Adult Treatment Editorial Board. PDQ Rectal Cancer Treatment. 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.
Based on the strength of the available evidence, treatment options may be described as either “standard” or “under clinical evaluation.” These classifications 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 treatment of rectal 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 Adult Treatment 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 Rectal Cancer Treatment 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 Adult Treatment 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® Adult Treatment Editorial Board. PDQ Rectal Cancer Treatment. 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.
Based on the strength of the available evidence, treatment options may be described as either “standard” or “under clinical evaluation.” These classifications 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.