你现在的位置: 首页 > 癌症医典(食管癌) > 食管癌的临床治疗(成人)
中文版 / English
食管癌的临床治疗(成人)(PDQ®)

有关食管癌的基本信息

食管恶性肿瘤主要有两种病理组织学类型:腺癌和鳞状细胞癌。典型的腺癌源于食管下段,鳞状细胞癌可起源于食管全段。两种病理类型的食管癌在流行病学方面具有显著差异。

发病率和死亡率

2020年美国食管癌的预计新发病例和死亡病例:

  • 新发病例:18440例。
  • 死亡病例:16170例。
  • 据估计,2015年中国食管癌新发病例和死亡病例分别为:

  • 新发病例:24.6万
  • 死亡病例:18.8万。
  • 数十年来,食管癌的发病率逐年升高。同时,组织学特征和原发肿瘤部位亦发生转变。在美国,鳞状细胞癌历来更为普遍,尽管在过去几十年里,腺癌在美国和西欧的发病率急剧上升。

    从全球范围来看,鳞状细胞癌仍占多数。然而,在美国和西欧,食管腺癌的发病率高于鳞状细胞癌。

    在白人男性人群中,腺癌的发病率明显升高。

    在美国,食管癌的中位发病年龄为67岁。

    多数腺癌位于远端食管。发病率升高的原因和人口学改变尚不明确。

    解剖

    食管和胃属于上消化道系统。

    食管,是与胃肠道相连的一条管道,主要用于输送食物。食管上连于咽,下连接于胃,位于胸腔的后纵隔,邻近纵隔胸膜、腹膜、心包、横膈膜。当进入腹腔时,食管呈急转的回转弯与胃相连。在消化道系统中,食管具有最显著的肌性管道结构,食管的肌层分为内环和外纵两层。上段和下段食管分别由环咽肌的括约肌和胃食管括约肌控制。食管的固有层和黏膜下层含丰富的淋巴管,有助于黏膜下的纵向淋巴引流。

    食管肿瘤,通常是以病灶上缘到门齿的距离来进行描述。通过内镜测量距门齿距离,食管长约30-40cm。食管主要分为四段:

  • 颈段食管(距门齿15-20cm)
  • 胸上段食管(距门齿20-25cm)
  • 胸中段食管(距门齿25-30cm)
  • 胸下段食管和食管胃结合部(距门齿30-40cm)
  • 危险因素

    食管鳞状细胞癌的危险因素包括

  • 吸烟
  • 酗酒
  • 食管腺癌的危险因素并不明确。

    Barrett(巴雷特)食管是一个例外,常提示患食管腺癌的风险升高。慢性返流病是诱发Barrett化生的一个重要危险因素。一项瑞典的基于人群的对照研究结果强烈提示,有症状的胃食管返流病是食管腺癌的一个危险因素。返流症状的发生率、严重程度、持续时间均与食管腺癌的危险因素呈正相关性。

    (更多详情,请参考食管癌预防部分PDQ章节)

    预后因素

    有利的预后因素包括:

  • 早期病变
  • 完全切除
  • 远端食管Barrett粘膜重度异形增生的患者通常会罹患增生部位的原位癌或浸润癌。手术切除后,此类患者的预后较佳。

    多数情况下,食管癌是一类可治性疾病,但很少能被治愈。采用正规治疗的食管癌患者的5年总体生存率为5%-30%。偶有非常早期的食管癌患者获得较好的生存期。

    相关章节

    其他PDQ章节也包括食管癌的相关信息,如

  • 食管癌预防
  • 食管癌筛查
  • 罕见儿童癌症的治疗
  • 胃肠道间质瘤,亦可发生于食管,通常为良性。相关信息,请参考以下章节:

  • 胃肠道间质瘤治疗(成人)
  • 有关食管癌患者的支持治疗,请参考以下章节:

  • 癌症护理的营养支持
  • 化疗和头颈部放疗后口腔并发症章节的吞咽困难部分
  • 参考文献

  • American Cancer Society: Cancer Facts and Figures 2020. Atlanta, Ga: American Cancer Society, 2020. Available online. Last accessed January 17, 2020.
  • Brown LM, Devesa SS, Chow WH: Incidence of adenocarcinoma of the esophagus among white Americans by sex, stage, and age. J Natl Cancer Inst 100 (16): 1184-7, 2008.
  • Blot WJ, McLaughlin JK: The changing epidemiology of esophageal cancer. Semin Oncol 26 (5 Suppl 15): 2-8, 1999.
  • Schmassmann A, Oldendorf MG, Gebbers JO: Changing incidence of gastric and oesophageal cancer subtypes in central Switzerland between 1982 and 2007. Eur J Epidemiol 24 (10): 603-9, 2009.
  • Kubo A, Corley DA: Marked multi-ethnic variation of esophageal and gastric cardia carcinomas within the United States. Am J Gastroenterol 99 (4): 582-8, 2004.
  • Ginsberg RJ: Cancer treatment in the elderly. J Am Coll Surg 187 (4): 427-8, 1998.
  • Lagergren J, Bergström R, Lindgren A, et al.: Symptomatic gastroesophageal reflux as a risk factor for esophageal adenocarcinoma. N Engl J Med 340 (11): 825-31, 1999.
  • Reed MF, Tolis G, Edil BH, et al.: Surgical treatment of esophageal high-grade dysplasia. Ann Thorac Surg 79 (4): 1110-5; discussion 1110-5, 2005.
  • Esophageal Cancer Treatment (Adult) (PDQ®)

    General Information About Esophageal Cancer

    Two histologic types account for the majority of malignant esophageal neoplasms: adenocarcinoma and squamous cell carcinoma. Adenocarcinomas typically start in the lower esophagus and squamous cell carcinoma can develop throughout the esophagus. The epidemiology of these types varies markedly.

    Incidence and Mortality

    Estimated new cases and deaths from esophageal cancer in the United States in 2020:

  • New cases: 18,440.
  • Deaths: 16,170.
  • The incidence of esophageal cancer has risen in recent decades, coinciding with a shift in histologic type and primary tumor location. In the United States, squamous cell carcinoma has historically been more prevalent although the incidence of adenocarcinoma has risen dramatically in the last few decades in the United States and western Europe.

    Worldwide, squamous cell carcinoma remains the predominant histology, however, adenocarcinoma of the esophagus is now more prevalent than squamous cell carcinoma in the United States and western Europe.

    The incidence of adenocarcinoma has increased most notably among white males.

    In the United States, the median age of patients who present with esophageal cancer is 67 years.

    Most adenocarcinomas are located in the distal esophagus. The cause for the rising incidence and demographic alterations is unknown.

    Anatomy

    The esophagus and stomach are part of the upper gastrointestinal (digestive) system.

    The esophagus serves as a conduit to the gastrointestinal tract for food. The esophagus extends from the larynx to the stomach and lies in the posterior mediastinum within the thorax near the lung pleura, peritoneum, pericardium, and diaphragm. As it travels into the abdominal cavity, the esophagus makes an abrupt turn and enters the stomach. The esophagus is the most muscular segment of the gastrointestinal system and is composed of inner circular and outer longitudinal muscle layers. The upper and lower esophagus are controlled by the sphincter function of the cricopharyngeus muscle and gastroesophageal sphincter, respectively. The esophagus has a rich network of lymphatic channels concentrated in the lamina propria and submucosa, which drains longitudinally along the submucosa.

    Tumors of the esophagus are conventionally described in terms of distance of the upper border of the tumor to the incisors. When measured from the incisors via endoscopy, the esophagus extends approximately 30 to 40 cm. The esophagus is divided into four main segments:

  • Cervical esophagus (~15–20 cm from the incisors).
  • Upper thoracic esophagus (~20–25 cm from the incisors).
  • Middle thoracic esophagus (~25–30 cm from the incisors).
  • Lower thoracic esophagus and gastroesophageal junction (~30–40 cm from the incisors).
  • Risk Factors

    Risk factors for squamous cell carcinoma of the esophagus include:

  • Tobacco.
  • Alcohol.
  • Risk factors associated with esophageal adenocarcinoma are less clear.

    Barrett esophagus is an exception and its presence is associated with an increased risk of developing adenocarcinoma of the esophagus. Chronic reflux is considered the predominant cause of Barrett metaplasia. The results of a population-based, case-controlled study from Sweden strongly suggest that symptomatic gastroesophageal reflux is a risk factor for esophageal adenocarcinoma. The frequency, severity, and duration of reflux symptoms were positively correlated with increased risk of esophageal adenocarcinoma.

    (Refer to the PDQ summary on Esophageal Cancer Prevention for more information.)

    Prognostic Factors

    Favorable prognostic factors include the following:

  • Early-stage disease.
  • Complete resection.
  • Patients with severe dysplasia in distal esophageal Barrett mucosa often have in situ or invasive cancer within the dysplastic area. After resection, these patients usually have excellent prognoses.

    In most cases, esophageal cancer is a treatable disease, but it is rarely curable. The overall 5-year survival rate in patients amenable to definitive treatment ranges from 5% to 30%. The occasional patient with very early disease has a better chance of survival.

    Other PDQ summaries containing information related to esophageal cancer include the following:

  • Esophageal Cancer Prevention
  • Esophageal Cancer Screening
  • Unusual Cancers of Childhood Treatment
  • For information about gastrointestinal stromal tumors, which can occur in the esophagus and are usually benign, refer to the following summary:

  • Gastrointestinal Stromal Tumors Treatment (Adult)
  • For information about supportive care for patients with esophageal cancer, refer to the following summaries:

  • Nutrition in Cancer Care
  • Dysphagia section in the Oral Complications of Chemotherapy and Head/Neck Radiation summary
  • ReferenceSection

  • American Cancer Society: Cancer Facts and Figures 2020. Atlanta, Ga: American Cancer Society, 2020. Available online. Last accessed January 17, 2020.
  • Brown LM, Devesa SS, Chow WH: Incidence of adenocarcinoma of the esophagus among white Americans by sex, stage, and age. J Natl Cancer Inst 100 (16): 1184-7, 2008.
  • Blot WJ, McLaughlin JK: The changing epidemiology of esophageal cancer. Semin Oncol 26 (5 Suppl 15): 2-8, 1999.
  • Schmassmann A, Oldendorf MG, Gebbers JO: Changing incidence of gastric and oesophageal cancer subtypes in central Switzerland between 1982 and 2007. Eur J Epidemiol 24 (10): 603-9, 2009.
  • Kubo A, Corley DA: Marked multi-ethnic variation of esophageal and gastric cardia carcinomas within the United States. Am J Gastroenterol 99 (4): 582-8, 2004.
  • Ginsberg RJ: Cancer treatment in the elderly. J Am Coll Surg 187 (4): 427-8, 1998.
  • Lagergren J, Bergström R, Lindgren A, et al.: Symptomatic gastroesophageal reflux as a risk factor for esophageal adenocarcinoma. N Engl J Med 340 (11): 825-31, 1999.
  • Reed MF, Tolis G, Edil BH, et al.: Surgical treatment of esophageal high-grade dysplasia. Ann Thorac Surg 79 (4): 1110-5; discussion 1110-5, 2005.
  • 食管癌的临床治疗(成人)(PDQ®)

    食管癌的细胞学分类

    典型起源于Barrett食管的腺癌至少占食管癌的50%。该组织类型食管癌的发病率逐年上升。Barrett食管内含向食管胃结合部生长的胃腺上皮。

    主要有三种类型的腺上皮

  • 化生型柱状上皮
  • 食管壁内的化生型壁细胞腺上皮
  • 化生型肠上皮伴典型的杯状细胞。特别是增生,很有可能发展为肠型粘膜。
  • 鳞状细胞癌在食管癌中不足50%。

    胃肠道间质瘤可发生于食管,通常是良性肿瘤。(更多信息,请参考胃肠道间质瘤治疗(成人)PDQ章节)

    Esophageal Cancer Treatment (Adult) (PDQ®)

    Cellular Classification of Esophageal Cancer

    Adenocarcinomas, typically arising in Barrett esophagus, account for at least 50% of malignant lesions, and the incidence of this histology appears to be rising. Barrett esophagus contains glandular epithelium cephalad to the esophagogastric junction.

    Three different types of glandular epithelium can be seen:

  • Metaplastic columnar epithelium.
  • Metaplastic parietal cell glandular epithelium within the esophageal wall.
  • Metaplastic intestinal epithelium with typical goblet cells. Dysplasia is particularly likely to develop in the intestinal type mucosa.
  • Fewer than 50% of esophageal cancers are squamous cell carcinomas.

    Gastrointestinal stromal tumors can occur in the esophagus and are usually benign. (Refer to the PDQ summary on Gastrointestinal Stromal Tumors Treatment (Adult) for more information.)

    食管癌的临床治疗(成人)(PDQ®)

    食管癌的分期信息

    很难对食管癌的多种治疗方法进行比较,其中最为重要的一个难题是缺乏精准的术前分期。临床分期决定了治疗目标,是选择根治性的还是姑息性治疗。

    分期评估

    标准非侵入性分期方法,主要有以下几种:

  • 内镜超声
  • 胸腹部CT检查
  • PET-CT检查
  • 内镜超声对肿瘤深度分期评估的准确率高达85%-90%,而CT为50%-80%。针对区域性淋巴结分期评估,内镜超声的准确率为70-80%,CT为50%-70%。

    一项回顾性系列研究显示内镜超声引导下的细针穿刺(FNA)在区域性淋巴结的分期评估中的敏感度为93%,特异度为100%。内镜超声引导下的细针穿刺对淋巴结分期评估的研究正处于前瞻性评估阶段。

    在部分外科中心,胸腔镜和腹腔镜已被用于食管癌的临床分期。

    一项针对107例患者采用胸腔镜/腹腔镜进行评估的临床试验,结果显示阳性淋巴结检出率高达56%,而采用非侵入性检查(如CT、MRI、内镜超声)的检出率为41%。二组患者的并发症和死亡率无明显差

    针对远处转移灶的筛查,非侵入性PET扫描采用放射性同位素氟代脱氧葡萄糖(18F-FDG)用于食管癌的术前临床分期评估更具敏感性,优于CT检查和内镜检查。近期一项262例潜在可切除食管癌患者在行标准评估后,其中至少4.8%患者采用18F-FDG PET检查发现了明确的远处转移病变。

    AJCC分期系统

    AJCC分期系统采用TNM(肿瘤、淋巴结、转移灶)对食管癌及食管胃结合部癌进行分期。

    肿瘤位于贲门处,距离食管胃结合部5cm范围内,同时侵及食管或食管胃结合部归为食管癌。肿瘤中心位于贲门,距离食管胃结合部5cm范围外,或未侵及食管归为胃癌。

    (更多信息,请查阅胃癌治疗章节的胃癌分期部分)

    将腹腔淋巴结受累划分为M1,一直都存有争议。腹腔淋巴结阳性对预后的影响远不如远处器官的转移。

    如患者出现区域性和/或腹主动脉旁转移病灶,则没有必要认为患者已因为转移失去手术机会。如果可能,可尝试行原发肿瘤的完全切除和淋巴结清扫术。

    表1. 食管鳞状细胞癌和腺癌的原发肿瘤、区域淋巴结、远处转移、组织学分级定义,及食管鳞状细胞癌的分段食管鳞状细胞癌的临床分期表2. 食管鳞状细胞癌pTNM 0期的定义表3. 食管鳞状细胞癌pTNM IA 和 IB 期的定义表4. 食管鳞状细胞癌pTNM IIA 和 IIB 期的定义表5. 食管鳞状细胞癌pTNM IIIA 和 IIIB 期的定义表6. 食管鳞状细胞癌pTNM IVA 和 IVB 期的定义当前的临床试验
    T 类别/标准N 类别/标准M 类别/标准G定义L 类别/标准
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    TX=原发肿瘤不能确定NX=区域淋巴结不能确定M0=无远处转移GX=病理分级不能确定X=肿瘤原发部位不明
    T0=找不到原发灶N0=无淋巴结转移M1=远处转移G1=高分化上段=颈段食管至奇静脉下缘
    Tis=高级别不典型增生,指恶性肿瘤细胞被基底膜局限于黏膜上皮层。N1=1-2个区域淋巴结转移G2=中分化中段=奇静脉下缘至下肺静脉下缘
    G3=低分化,未分化下段:下肺静脉下缘至胃,包括食管胃结合部。
    T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。N2=3-6个区域淋巴结转移
    N3=7个及7个以上区域淋巴结转移
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    T2=肿瘤侵犯固有肌层
    T3=肿瘤侵犯外膜
    T4=肿瘤侵犯邻近组织结构
    T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜、
    T4b=肿瘤侵犯其他邻近组织结构,如主动脉、椎体、气管。
    分期TNM病理分级肿瘤位置描述图示
    0Tis, N0, M0不适用任一Tis=高级别不典型增生,指恶性肿瘤细胞被基底膜局限于黏膜上皮层。
    N0=无淋巴结转移
    M0=无远处转移
    G1不适用
    任意肿瘤位置=见表1
    分期TNM病理分级肿瘤位置描述图示
    IATia, N0, M0G1任一T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1 =高分化
    任意肿瘤位置=见表1
    T1a, N0, M0GX任一T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX =病理分级不能确定
    任意肿瘤位置=见表1
    IBT1a, N0, M0G2–G3任一T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G2 =中分化
    G3=低分化,未分化
    任意肿瘤位置=见表1
    T1b, N0, M0G1–G3任一T1b=肿瘤侵犯黏膜下层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1=高分化
    G2 =中分化
    G3 =低分化,未分化
    任意肿瘤位置=见表1
    T1b, N0, M0GX任一T1b=肿瘤侵犯+E186:E785下层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX=病理分级不能确定
    任意肿瘤位置=见表1
    T2, N0, M0G1任一T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1=高分化
    IIA
    分期TNM病理分级肿瘤位置描述图示
    IIAT2, N0, M0GX任一T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX=病理分级不能确定
    任意肿瘤位置=见表1
    T2, N0, M0G2–G3任一T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G2 =中分化
    G3 =低分化,未分化
    任意肿瘤位置=见表1
    T3, N0, M0任一下段T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    下段:下肺静脉下缘至胃,包括食管胃结合部。
    T3, N0, M0G1上段/中段T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1 =高分化
    上段=颈段食管至奇静脉下缘
    中段=奇静脉下缘至下肺静脉下缘
    IIBT3, N0, M0G2–G3上段/中段T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G2 =中分化
    G3 =低分化,未分化
    上段=颈段食管至奇静脉下缘
    中段=奇静脉下缘至下肺静脉下缘
    T3, N0, M0GX任一T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0=无远处转移
    GX=病理分级不能确定
    任意肿瘤位置=见表1
    T3, N0, M0任一肿瘤位置XT3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    肿瘤位置X=原发灶不明
    T1, N1, M0任一任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    N1=1-2个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    分期TNM病理分级肿瘤位置描述图示
    IIIAT1, N2, M0任一任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T2, N1, M0任一任一T2=肿瘤侵犯固有肌层
    N1=1-2个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    IIIBT2, N2, M0任一任一T2=肿瘤侵犯固有肌层
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T3, N1–N2, M0任一任一T3=肿瘤侵犯外膜
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T4a, N0–1, M0任一任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    分期TNM病理分级肿瘤位置描述图示
    IVAT4a, N2, M0任一任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T4b, N0–2, M0任一任一T4b=肿瘤侵犯其他邻近组织结构,如主动脉、椎体、气管。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    Any T, N3, M0任一任一任一T分期=见表1
    N3=7个及7个以上区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    IVB任一T分期、任一N分期, M1任一任一任一T分期=见表1
    任一N分期=见表1
    M1=远处转移
    任一病理分级=见表1
    任一T分期=见表1
    T=原发肿瘤定义;N=区域淋巴结定义;M=远处转移定义;G=组织学分级定义;L=肿瘤位置定义
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; N/A = 不适用;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准

    食管鳞状细胞癌的临床分期

    表2. 食管鳞状细胞癌pTNM 0期的定义表3. 食管鳞状细胞癌pTNM IA 和 IB 期的定义表4. 食管鳞状细胞癌pTNM IIA 和 IIB 期的定义表5. 食管鳞状细胞癌pTNM IIIA 和 IIIB 期的定义表6. 食管鳞状细胞癌pTNM IVA 和 IVB 期的定义当前的临床试验
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    0Tis, N0, M0不适用任一Tis=高级别不典型增生,指恶性肿瘤细胞被基底膜局限于黏膜上皮层。
    N0=无淋巴结转移
    M0=无远处转移
    G1不适用
    任意肿瘤位置=见表1
    分期TNM病理分级肿瘤位置描述图示
    IATia, N0, M0G1任一T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1 =高分化
    任意肿瘤位置=见表1
    T1a, N0, M0GX任一T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX =病理分级不能确定
    任意肿瘤位置=见表1
    IBT1a, N0, M0G2–G3任一T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G2 =中分化
    G3=低分化,未分化
    任意肿瘤位置=见表1
    T1b, N0, M0G1–G3任一T1b=肿瘤侵犯黏膜下层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1=高分化
    G2 =中分化
    G3 =低分化,未分化
    任意肿瘤位置=见表1
    T1b, N0, M0GX任一T1b=肿瘤侵犯+E186:E785下层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX=病理分级不能确定
    任意肿瘤位置=见表1
    T2, N0, M0G1任一T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1=高分化
    IIA
    分期TNM病理分级肿瘤位置描述图示
    IIAT2, N0, M0GX任一T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX=病理分级不能确定
    任意肿瘤位置=见表1
    T2, N0, M0G2–G3任一T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G2 =中分化
    G3 =低分化,未分化
    任意肿瘤位置=见表1
    T3, N0, M0任一下段T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    下段:下肺静脉下缘至胃,包括食管胃结合部。
    T3, N0, M0G1上段/中段T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1 =高分化
    上段=颈段食管至奇静脉下缘
    中段=奇静脉下缘至下肺静脉下缘
    IIBT3, N0, M0G2–G3上段/中段T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G2 =中分化
    G3 =低分化,未分化
    上段=颈段食管至奇静脉下缘
    中段=奇静脉下缘至下肺静脉下缘
    T3, N0, M0GX任一T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0=无远处转移
    GX=病理分级不能确定
    任意肿瘤位置=见表1
    T3, N0, M0任一肿瘤位置XT3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    肿瘤位置X=原发灶不明
    T1, N1, M0任一任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    N1=1-2个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    分期TNM病理分级肿瘤位置描述图示
    IIIAT1, N2, M0任一任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T2, N1, M0任一任一T2=肿瘤侵犯固有肌层
    N1=1-2个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    IIIBT2, N2, M0任一任一T2=肿瘤侵犯固有肌层
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T3, N1–N2, M0任一任一T3=肿瘤侵犯外膜
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T4a, N0–1, M0任一任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    分期TNM病理分级肿瘤位置描述图示
    IVAT4a, N2, M0任一任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T4b, N0–2, M0任一任一T4b=肿瘤侵犯其他邻近组织结构,如主动脉、椎体、气管。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    Any T, N3, M0任一任一任一T分期=见表1
    N3=7个及7个以上区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    IVB任一T分期、任一N分期, M1任一任一任一T分期=见表1
    任一N分期=见表1
    M1=远处转移
    任一病理分级=见表1
    任一T分期=见表1
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; N/A = 不适用;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    表3. 食管鳞状细胞癌pTNM IA 和 IB 期的定义表4. 食管鳞状细胞癌pTNM IIA 和 IIB 期的定义表5. 食管鳞状细胞癌pTNM IIIA 和 IIIB 期的定义表6. 食管鳞状细胞癌pTNM IVA 和 IVB 期的定义当前的临床试验
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    IATia, N0, M0G1任一T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1 =高分化
    任意肿瘤位置=见表1
    T1a, N0, M0GX任一T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX =病理分级不能确定
    任意肿瘤位置=见表1
    IBT1a, N0, M0G2–G3任一T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G2 =中分化
    G3=低分化,未分化
    任意肿瘤位置=见表1
    T1b, N0, M0G1–G3任一T1b=肿瘤侵犯黏膜下层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1=高分化
    G2 =中分化
    G3 =低分化,未分化
    任意肿瘤位置=见表1
    T1b, N0, M0GX任一T1b=肿瘤侵犯+E186:E785下层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX=病理分级不能确定
    任意肿瘤位置=见表1
    T2, N0, M0G1任一T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1=高分化
    IIA
    分期TNM病理分级肿瘤位置描述图示
    IIAT2, N0, M0GX任一T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX=病理分级不能确定
    任意肿瘤位置=见表1
    T2, N0, M0G2–G3任一T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G2 =中分化
    G3 =低分化,未分化
    任意肿瘤位置=见表1
    T3, N0, M0任一下段T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    下段:下肺静脉下缘至胃,包括食管胃结合部。
    T3, N0, M0G1上段/中段T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1 =高分化
    上段=颈段食管至奇静脉下缘
    中段=奇静脉下缘至下肺静脉下缘
    IIBT3, N0, M0G2–G3上段/中段T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G2 =中分化
    G3 =低分化,未分化
    上段=颈段食管至奇静脉下缘
    中段=奇静脉下缘至下肺静脉下缘
    T3, N0, M0GX任一T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0=无远处转移
    GX=病理分级不能确定
    任意肿瘤位置=见表1
    T3, N0, M0任一肿瘤位置XT3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    肿瘤位置X=原发灶不明
    T1, N1, M0任一任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    N1=1-2个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    分期TNM病理分级肿瘤位置描述图示
    IIIAT1, N2, M0任一任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T2, N1, M0任一任一T2=肿瘤侵犯固有肌层
    N1=1-2个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    IIIBT2, N2, M0任一任一T2=肿瘤侵犯固有肌层
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T3, N1–N2, M0任一任一T3=肿瘤侵犯外膜
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T4a, N0–1, M0任一任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    分期TNM病理分级肿瘤位置描述图示
    IVAT4a, N2, M0任一任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T4b, N0–2, M0任一任一T4b=肿瘤侵犯其他邻近组织结构,如主动脉、椎体、气管。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    Any T, N3, M0任一任一任一T分期=见表1
    N3=7个及7个以上区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    IVB任一T分期、任一N分期, M1任一任一任一T分期=见表1
    任一N分期=见表1
    M1=远处转移
    任一病理分级=见表1
    任一T分期=见表1
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    表4. 食管鳞状细胞癌pTNM IIA 和 IIB 期的定义表5. 食管鳞状细胞癌pTNM IIIA 和 IIIB 期的定义表6. 食管鳞状细胞癌pTNM IVA 和 IVB 期的定义当前的临床试验
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    IIAT2, N0, M0GX任一T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX=病理分级不能确定
    任意肿瘤位置=见表1
    T2, N0, M0G2–G3任一T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G2 =中分化
    G3 =低分化,未分化
    任意肿瘤位置=见表1
    T3, N0, M0任一下段T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    下段:下肺静脉下缘至胃,包括食管胃结合部。
    T3, N0, M0G1上段/中段T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1 =高分化
    上段=颈段食管至奇静脉下缘
    中段=奇静脉下缘至下肺静脉下缘
    IIBT3, N0, M0G2–G3上段/中段T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G2 =中分化
    G3 =低分化,未分化
    上段=颈段食管至奇静脉下缘
    中段=奇静脉下缘至下肺静脉下缘
    T3, N0, M0GX任一T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0=无远处转移
    GX=病理分级不能确定
    任意肿瘤位置=见表1
    T3, N0, M0任一肿瘤位置XT3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    肿瘤位置X=原发灶不明
    T1, N1, M0任一任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    N1=1-2个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    分期TNM病理分级肿瘤位置描述图示
    IIIAT1, N2, M0任一任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T2, N1, M0任一任一T2=肿瘤侵犯固有肌层
    N1=1-2个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    IIIBT2, N2, M0任一任一T2=肿瘤侵犯固有肌层
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T3, N1–N2, M0任一任一T3=肿瘤侵犯外膜
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T4a, N0–1, M0任一任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    分期TNM病理分级肿瘤位置描述图示
    IVAT4a, N2, M0任一任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T4b, N0–2, M0任一任一T4b=肿瘤侵犯其他邻近组织结构,如主动脉、椎体、气管。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    Any T, N3, M0任一任一任一T分期=见表1
    N3=7个及7个以上区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    IVB任一T分期、任一N分期, M1任一任一任一T分期=见表1
    任一N分期=见表1
    M1=远处转移
    任一病理分级=见表1
    任一T分期=见表1
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    表5. 食管鳞状细胞癌pTNM IIIA 和 IIIB 期的定义表6. 食管鳞状细胞癌pTNM IVA 和 IVB 期的定义当前的临床试验
    分期TNM病理分级肿瘤位置描述图示
    分期TNM病理分级肿瘤位置描述图示
    IIIAT1, N2, M0任一任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T2, N1, M0任一任一T2=肿瘤侵犯固有肌层
    N1=1-2个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    IIIBT2, N2, M0任一任一T2=肿瘤侵犯固有肌层
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T3, N1–N2, M0任一任一T3=肿瘤侵犯外膜
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0=无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T4a, N0–1, M0任一任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    分期TNM病理分级肿瘤位置描述图示
    IVAT4a, N2, M0任一任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T4b, N0–2, M0任一任一T4b=肿瘤侵犯其他邻近组织结构,如主动脉、椎体、气管。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    Any T, N3, M0任一任一任一T分期=见表1
    N3=7个及7个以上区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    IVB任一T分期、任一N分期, M1任一任一任一T分期=见表1
    任一N分期=见表1
    M1=远处转移
    任一病理分级=见表1
    任一T分期=见表1
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准
    表6. 食管鳞状细胞癌pTNM IVA 和 IVB 期的定义当前的临床试验
    分期TNM病理分级肿瘤位置描述图示
    IVAT4a, N2, M0任一任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    T4b, N0–2, M0任一任一T4b=肿瘤侵犯其他邻近组织结构,如主动脉、椎体、气管。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    Any T, N3, M0任一任一任一T分期=见表1
    N3=7个及7个以上区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一肿瘤位置=见表1
    IVB任一T分期、任一N分期, M1任一任一任一T分期=见表1
    任一N分期=见表1
    M1=远处转移
    任一病理分级=见表1
    任一T分期=见表1
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;L=肿瘤部位; P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    b 肿瘤分段以食管肿瘤的中心位置为界定标准

    当前的临床试验

    采用我们先进的临床试验搜索引擎查找正在进行患者招募的NCI支持的癌症临床试验。搜索引擎可按试验所在地、治疗类型、药物名称和其他标准进行设置。还能查询有关临床试验的基本信息。

    食管腺癌的分期

    表7. 食管腺癌pTNM 0 期的定义表8. 食管腺癌pTNM IA, IB, IC 期的定义表9. 食管腺癌pTNM IIA 和 IIB 期的定义表10. 食管腺癌pTNM IIIA 和 IIIB 期的定义表11. 食管腺癌pTNM IVA 和 IVB 期的定义
    分期TNM病理分级描述图示
    分期TNM病理分级描述图示
    分期TNM病理分级描述图示
    分期TNM病理分级描述图示
    分期TNM病理分级描述图示
    0Tis, N0, M0不适用Tis=高级别不典型增生,指恶性肿瘤细胞被基底膜局限于黏膜上皮层。
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    分期TNM病理分级描述图示
    IAT1a, N0, M0G1T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1 =高分化
    T1a, N0, M0GXT1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX =病理分级不能确定
    IBT1a, N0, M0G2T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G2 =中分化
    T1b, N0, M0G1–2T1b=肿瘤侵犯黏膜下层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1 =高分化
    G2 =中分化
    T1b, N0, M0GXT1b=肿瘤侵犯黏膜下层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX =病理分级不能确定
    ICT1, N0, M0G3T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G3 =低分化,未分化
    T2, N0, M0G1–2T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1 =高分化
    G2 =中分化
    分期TNM病理分级描述图示
    IIAT2, N0, M0G3T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G3 =低分化,未分化
    T2, N0, M0GXT2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX =病理分级不能确定
    IIBT1, N1, M0任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T3, N0, M0任一T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    分期TNM病理分级描述图示
    IIIAT1, N2, M0任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T2, N1, M0任一T2=肿瘤侵犯固有肌层
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    IIIBT2, N2, M0任一T2=肿瘤侵犯固有肌层
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T3, N1–2, M0任一T3=肿瘤侵犯外膜
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T4a, N0–1, M0任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    分期TNM病理分级描述图示
    IVAT4a, N2, M0任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T4b, N0–2, M0任一T4b=肿瘤侵犯其他邻近组织结构,如主动脉、椎体、气管。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一T分期、N3, M1任一任一T分期=见表1
    N3=7个及7个以上区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    IVB任一T分期、任一N分期, M1任一任一T分期=见表1
    任一N分期=见表1
    M1=远处转移
    任一病理分级=见表1
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;N/A:不适用;L=肿瘤位置;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    表8. 食管腺癌pTNM IA, IB, IC 期的定义表9. 食管腺癌pTNM IIA 和 IIB 期的定义表10. 食管腺癌pTNM IIIA 和 IIIB 期的定义表11. 食管腺癌pTNM IVA 和 IVB 期的定义
    分期TNM病理分级描述图示
    分期TNM病理分级描述图示
    分期TNM病理分级描述图示
    分期TNM病理分级描述图示
    IAT1a, N0, M0G1T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1 =高分化
    T1a, N0, M0GXT1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX =病理分级不能确定
    IBT1a, N0, M0G2T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G2 =中分化
    T1b, N0, M0G1–2T1b=肿瘤侵犯黏膜下层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1 =高分化
    G2 =中分化
    T1b, N0, M0GXT1b=肿瘤侵犯黏膜下层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX =病理分级不能确定
    ICT1, N0, M0G3T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G3 =低分化,未分化
    T2, N0, M0G1–2T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G1 =高分化
    G2 =中分化
    分期TNM病理分级描述图示
    IIAT2, N0, M0G3T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G3 =低分化,未分化
    T2, N0, M0GXT2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX =病理分级不能确定
    IIBT1, N1, M0任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T3, N0, M0任一T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    分期TNM病理分级描述图示
    IIIAT1, N2, M0任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T2, N1, M0任一T2=肿瘤侵犯固有肌层
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    IIIBT2, N2, M0任一T2=肿瘤侵犯固有肌层
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T3, N1–2, M0任一T3=肿瘤侵犯外膜
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T4a, N0–1, M0任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    分期TNM病理分级描述图示
    IVAT4a, N2, M0任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T4b, N0–2, M0任一T4b=肿瘤侵犯其他邻近组织结构,如主动脉、椎体、气管。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一T分期、N3, M1任一任一T分期=见表1
    N3=7个及7个以上区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    IVB任一T分期、任一N分期, M1任一任一T分期=见表1
    任一N分期=见表1
    M1=远处转移
    任一病理分级=见表1
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    表9. 食管腺癌pTNM IIA 和 IIB 期的定义表10. 食管腺癌pTNM IIIA 和 IIIB 期的定义表11. 食管腺癌pTNM IVA 和 IVB 期的定义
    分期TNM病理分级描述图示
    分期TNM病理分级描述图示
    分期TNM病理分级描述图示
    IIAT2, N0, M0G3T2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    G3 =低分化,未分化
    T2, N0, M0GXT2=肿瘤侵犯固有肌层
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    GX =病理分级不能确定
    IIBT1, N1, M0任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T3, N0, M0任一T3=肿瘤侵犯外膜
    N0 = 无区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    分期TNM病理分级描述图示
    IIIAT1, N2, M0任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T2, N1, M0任一T2=肿瘤侵犯固有肌层
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    IIIBT2, N2, M0任一T2=肿瘤侵犯固有肌层
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T3, N1–2, M0任一T3=肿瘤侵犯外膜
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T4a, N0–1, M0任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    分期TNM病理分级描述图示
    IVAT4a, N2, M0任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T4b, N0–2, M0任一T4b=肿瘤侵犯其他邻近组织结构,如主动脉、椎体、气管。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一T分期、N3, M1任一任一T分期=见表1
    N3=7个及7个以上区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    IVB任一T分期、任一N分期, M1任一任一T分期=见表1
    任一N分期=见表1
    M1=远处转移
    任一病理分级=见表1
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    表10. 食管腺癌pTNM IIIA 和 IIIB 期的定义表11. 食管腺癌pTNM IVA 和 IVB 期的定义
    分期TNM病理分级描述图示
    分期TNM病理分级描述图示
    IIIAT1, N2, M0任一T1=肿瘤侵犯食管黏膜固有层、黏膜肌层、黏膜下层。
    T1a=肿瘤侵犯黏膜固有层或黏膜肌层
    T1b=肿瘤侵犯黏膜下层
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T2, N1, M0任一T2=肿瘤侵犯固有肌层
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    IIIBT2, N2, M0任一T2=肿瘤侵犯固有肌层
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T3, N1–2, M0任一T3=肿瘤侵犯外膜
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T4a, N0–1, M0任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    分期TNM病理分级描述图示
    IVAT4a, N2, M0任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T4b, N0–2, M0任一T4b=肿瘤侵犯其他邻近组织结构,如主动脉、椎体、气管。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一T分期、N3, M1任一任一T分期=见表1
    N3=7个及7个以上区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    IVB任一T分期、任一N分期, M1任一任一T分期=见表1
    任一N分期=见表1
    M1=远处转移
    任一病理分级=见表1
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页
    表11. 食管腺癌pTNM IVA 和 IVB 期的定义
    分期TNM病理分级描述图示
    IVAT4a, N2, M0任一T4a=肿瘤侵犯胸膜、心包、奇静脉、横膈膜和腹膜。
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    T4b, N0–2, M0任一T4b=肿瘤侵犯其他邻近组织结构,如主动脉、椎体、气管。
    N0 = 无区域淋巴结转移
    N1=1-2个区域淋巴结转移
    N2=3-6个区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    任一T分期、N3, M1任一任一T分期=见表1
    N3=7个及7个以上区域淋巴结转移
    M0 = 无远处转移
    任一病理分级=见表1
    IVB任一T分期、任一N分期, M1任一任一T分期=见表1
    任一N分期=见表1
    M1=远处转移
    任一病理分级=见表1
    T=原发肿瘤;N=区域淋巴结;M=远处转移;G=病理分级;P=病理的
    a 已取得AJCC授权版权:食管、食管胃结合部,参考Amin MB, Edge SB, Greene FL, et al. AJCC癌症分期手册 第8版 纽约 Springer, 2017,第185-202页

    参考文献

  • Ziegler K, Sanft C, Zeitz M, et al.: Evaluation of endosonography in TN staging of oesophageal cancer. Gut 32 (1): 16-20, 1991.
  • Tio TL, Coene PP, den Hartog Jager FC, et al.: Preoperative TNM classification of esophageal carcinoma by endosonography. Hepatogastroenterology 37 (4): 376-81, 1990.
  • Vazquez-Sequeiros E, Norton ID, Clain JE, et al.: Impact of EUS-guided fine-needle aspiration on lymph node staging in patients with esophageal carcinoma. Gastrointest Endosc 53 (7): 751-7, 2001.
  • Bonavina L, Incarbone R, Lattuada E, et al.: Preoperative laparoscopy in management of patients with carcinoma of the esophagus and of the esophagogastric junction. J Surg Oncol 65 (3): 171-4, 1997.
  • Sugarbaker DJ, Jaklitsch MT, Liptay MJ: Thoracoscopic staging and surgical therapy for esophageal cancer. Chest 107 (6 Suppl): 218S-223S, 1995.
  • Luketich JD, Schauer P, Landreneau R, et al.: Minimally invasive surgical staging is superior to endoscopic ultrasound in detecting lymph node metastases in esophageal cancer. J Thorac Cardiovasc Surg 114 (5): 817-21; discussion 821-3, 1997.
  • Krasna MJ, Reed CE, Nedzwiecki D, et al.: CALGB 9380: a prospective trial of the feasibility of thoracoscopy/laparoscopy in staging esophageal cancer. Ann Thorac Surg 71 (4): 1073-9, 2001.
  • Flamen P, Lerut A, Van Cutsem E, et al.: Utility of positron emission tomography for the staging of patients with potentially operable esophageal carcinoma. J Clin Oncol 18 (18): 3202-10, 2000.
  • Flamen P, Van Cutsem E, Lerut A, et al.: Positron emission tomography for assessment of the response to induction radiochemotherapy in locally advanced oesophageal cancer. Ann Oncol 13 (3): 361-8, 2002.
  • Weber WA, Ott K, Becker K, et al.: Prediction of response to preoperative chemotherapy in adenocarcinomas of the esophagogastric junction by metabolic imaging. J Clin Oncol 19 (12): 3058-65, 2001.
  • van Westreenen HL, Westerterp M, Bossuyt PM, et al.: Systematic review of the staging performance of 18F-fluorodeoxyglucose positron emission tomography in esophageal cancer. J Clin Oncol 22 (18): 3805-12, 2004.
  • Meyers BF, Downey RJ, Decker PA, et al.: The utility of positron emission tomography in staging of potentially operable carcinoma of the thoracic esophagus: results of the American College of Surgeons Oncology Group Z0060 trial. J Thorac Cardiovasc Surg 133 (3): 738-45, 2007.
  • Rice TW, Kelsen D, Blackstone EH, et al.: Esophagus and Esophagogastric Junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 185-202.
  • Korst RJ, Rusch VW, Venkatraman E, et al.: Proposed revision of the staging classification for esophageal cancer. J Thorac Cardiovasc Surg 115 (3): 660-69; discussion 669-70, 1998.
  • Esophageal Cancer Treatment (Adult) (PDQ®)

    Stage Information for Esophageal Cancer

    One of the major difficulties in allocating and comparing treatment modalities for patients with esophageal cancer is the lack of precise preoperative staging. The stage determines whether the intent of the therapeutic approach will be curative or palliative.

    Staging Evaluation

    Standard noninvasive staging modalities include the following:

  • Endoscopic ultrasound.
  • Computed tomography (CT) scan of the chest and abdomen.
  • Positron emission tomography (PET)-CT scan.
  • The overall tumor depth staging accuracy of endoscopic ultrasound is 85% to 90%, compared with 50% to 80% for CT; the accuracy of regional nodal staging is 70% to 80% for endoscopic ultrasound and 50% to 70% for CT.

    One retrospective series reported 93% sensitivity and 100% specificity of regional nodal staging with endoscopic ultrasound-guided fine-needle aspiration (FNA). Endoscopic ultrasound-guided FNA for lymph node staging is under prospective evaluation.

    Thoracoscopy and laparoscopy have been used in esophageal cancer staging at some surgical centers.

    An intergroup trial reported an increase in positive lymph node detection to 56% of 107 evaluable patients with the use of thoracoscopy/laparoscopy, from 41% (with the use of noninvasive staging tests, e.g., CT, magnetic resonance imaging, and endoscopic ultrasound), with no major complications or deaths.

    Noninvasive PET scan using the radiolabeled glucose analog fluorine F 18-fludeoxyglucose (18F-FDG) for preoperative staging of esophageal cancer is more sensitive than a CT scan or endoscopic ultrasound in detection of distant metastases. A recent study of 262 patients with potentially resectable esophageal cancer demonstrated the utility of 18F-FDG PET in identifying confirmed distant metastatic disease in at least 4.8% of patients after standard evaluation.

    AJCC Staging System

    The AJCC has designated staging by TNM (tumor, node, metastasis) classification to define cancer of the esophagus and esophagogastric junction.

    Tumors located in the gastric cardia within 5 cm of the gastroesophageal junction with extension into the esophagus or the gastroesophageal junction are classified as esophageal cancer. Tumors with the epicenter of the tumor located in the gastric cardia beyond 5 cm of the gastroesophageal junction or without extension into the esophagus are classified as gastric cancer.

    (Refer to the Stage Information for Gastric Cancer section in the PDQ summary on Gastric Cancer Treatment for more information.)

    The classification of involved abdominal lymph nodes as M1 disease is controversial. The presence of positive abdominal lymph nodes does not appear to have a prognosis as grave as that for metastases to distant organs.

    Patients with regional and/or celiac axis lymphadenopathy should not necessarily be considered to have unresectable disease caused by metastases. Complete resection of the primary tumor and appropriate lymphadenectomy is attempted when possible.

    Table 1. Definitions of Primary Tumor, Regional Lymph Node, Distant Metastasis, Histologic Grade for Squamous Cell Carcinoma and Adenocarcinoma, and Location for Squamous Cell Carcinoma of the EsophagusStaging for squamous cell carcinoma of the esophagusTable 2. Definitions of pTNM Stage 0 for Squamous Cell Carcinoma of the EsophagusTable 3. Definitions of pTNM Stages IA and IB for Squamous Cell Carcinoma of the EsophagusTable 4. Definitions of pTNM Stages IIA and IIB for Squamous Cell Carcinoma of the EsophagusTable 5. Definitions of pTNM Stages IIIA and IIIB for Squamous Cell Carcinoma of the EsophagusTable 6. Definitions of pTNM Stages IVA and IVB for Squamous Cell Carcinoma of the EsophagusCurrent Clinical Trials
    T Category/CriteriaN Category/CriteriaM Category/CriteriaG DefinitionL Category/Criteria
    StageTNMGradeTumor LocationDescriptionIllustration
    StageTNMGradeTumor LocationDescriptionIllustration
    StageTNMGradeTumor LocationDescriptionIllustration
    Stage TNM GradeTumor LocationDescriptionIllustration
    Stage TNM GradeTumor LocationDescriptionIllustration
    TX = Tumor cannot be assessed.NX = Regional lymph nodes cannot be assessed.M0 = No distant metastasis.GX = Grade cannot be assessed.X = Location unknown.
    T0 = No evidence of primary tumor.N0 = No regional lymph node metastasis.M1 = Distant metastasis.G1 = Well differentiated.Upper = Cervical esophagus to lower border of azygos vein.
    Tis = High-grade dysplasia, defined as malignant cells confined to the epithelium by the basement membrane.N1 = Metastasis in one or two regional lymph nodes.G2 = Moderately differentiated.Middle = Lower border of azygos vein to lower border of inferior pulmonary vein.
    G3 = Poorly differentiated, undifferentiated.Lower = Lower border of inferior pulmonary vein to stomach, including gastroesophageal junction.
    T1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.N2 = Metastasis in three to six regional lymph nodes.
    N3 = Metastasis in seven or more regional lymph nodes.
    T1a = Tumor invades the lamina propria or muscularis mucosae.
    T1b = Tumor invades the submucosa.
    T2 = Tumor invades the muscularis propria.
    T3 = Tumor invades adventitia.
    T4 = Tumor invades adjacent structures.
    T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    T4b = Tumor invades other adjacent structures, such as the aorta, vertebral body, or airway.
    StageTNMGradeTumor LocationDescriptionIllustration
    0Tis, N0, M0N/AAnyTis = High grade dysplasia, defined as malignant cells confined to the epithelium by the basement membrane.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = N/A.
    Any L = See Table 1.
    StageTNMGradeTumor LocationDescriptionIllustration
    IATia, N0, M0G1Any–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    Any L = See Table 1.
    T1a, N0, M0GXAny–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    IBT1a, N0, M0G2–G3Any–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Any L = See Table 1.
    T1b, N0, M0G1–G3Any–T1b = Tumor invades the submucosa.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Any L = See Table 1.
    T1b, N0, M0GXAny–T1b = Tumor invades the submucosa.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    T2, N0, M0G1AnyT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    Any L = See Table 1.
    StageTNMGradeTumor LocationDescriptionIllustration
    IIAT2, N0, M0GXAnyT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    T2, N0, M0G2–G3AnyT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Any L = See Table 1.
    T3, N0, M0AnyLowerT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Lower = Lower border of inferior pulmonary vein to stomach, including gastroesophageal junction.
    T3, N0, M0G1Upper/middleT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    Upper = Cervical esophagus to lower border of azygos vein.
    Middle = Lower border of azygos vein to lower border of inferior pulmonary vein.
    IIBT3, N0, M0G2–G3Upper/middleT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Upper = Cervical esophagus to lower border of azygos vein.
    Middle = Lower border of azygos vein to lower border of inferior pulmonary vein.
    T3, N0, M0GXAnyT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    T3, N0, M0AnyLocation XT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Location X = Location unknown.
    T1, N1, M0AnyAnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Stage TNM GradeTumor LocationDescriptionIllustration
    IIIAT1, N2, M0AnyAnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T2, N1, M0AnyAnyT2 = Tumor invades the muscularis propria.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    IIIBT2, N2, M0AnyAnyT2 = Tumor invades the muscularis propria.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T3, N1–N2, M0AnyAnyT3 = Tumor invades adventitia.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T4a, N0–1, M0AnyAny–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Stage TNM GradeTumor LocationDescriptionIllustration
    IVAT4a, N2, M0AnyAny–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T4b, N0–2, M0AnyAny–T4b = Tumor invades other adjacent structures, such as the aorta, vertebral body, or airway.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Any T, N3, M0AnyAnyAny T = See Table 1.
    N3 = Metastasis in seven or more regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    IVBAny T, Any N, M1AnyAnyAny T = See Table 1.
    Any N = See Table 1.
    M1 = Distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T = definition of primary tumor; N = definition of regional lymph nodes; M = definition of distant metastasis; G = definition of histologic grade; L = definition of location.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; N/A = not applicable; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.

    Staging for squamous cell carcinoma of the esophagus

    Table 2. Definitions of pTNM Stage 0 for Squamous Cell Carcinoma of the EsophagusTable 3. Definitions of pTNM Stages IA and IB for Squamous Cell Carcinoma of the EsophagusTable 4. Definitions of pTNM Stages IIA and IIB for Squamous Cell Carcinoma of the EsophagusTable 5. Definitions of pTNM Stages IIIA and IIIB for Squamous Cell Carcinoma of the EsophagusTable 6. Definitions of pTNM Stages IVA and IVB for Squamous Cell Carcinoma of the EsophagusCurrent Clinical Trials
    StageTNMGradeTumor LocationDescriptionIllustration
    StageTNMGradeTumor LocationDescriptionIllustration
    StageTNMGradeTumor LocationDescriptionIllustration
    Stage TNM GradeTumor LocationDescriptionIllustration
    Stage TNM GradeTumor LocationDescriptionIllustration
    0Tis, N0, M0N/AAnyTis = High grade dysplasia, defined as malignant cells confined to the epithelium by the basement membrane.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = N/A.
    Any L = See Table 1.
    StageTNMGradeTumor LocationDescriptionIllustration
    IATia, N0, M0G1Any–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    Any L = See Table 1.
    T1a, N0, M0GXAny–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    IBT1a, N0, M0G2–G3Any–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Any L = See Table 1.
    T1b, N0, M0G1–G3Any–T1b = Tumor invades the submucosa.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Any L = See Table 1.
    T1b, N0, M0GXAny–T1b = Tumor invades the submucosa.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    T2, N0, M0G1AnyT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    Any L = See Table 1.
    StageTNMGradeTumor LocationDescriptionIllustration
    IIAT2, N0, M0GXAnyT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    T2, N0, M0G2–G3AnyT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Any L = See Table 1.
    T3, N0, M0AnyLowerT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Lower = Lower border of inferior pulmonary vein to stomach, including gastroesophageal junction.
    T3, N0, M0G1Upper/middleT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    Upper = Cervical esophagus to lower border of azygos vein.
    Middle = Lower border of azygos vein to lower border of inferior pulmonary vein.
    IIBT3, N0, M0G2–G3Upper/middleT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Upper = Cervical esophagus to lower border of azygos vein.
    Middle = Lower border of azygos vein to lower border of inferior pulmonary vein.
    T3, N0, M0GXAnyT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    T3, N0, M0AnyLocation XT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Location X = Location unknown.
    T1, N1, M0AnyAnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Stage TNM GradeTumor LocationDescriptionIllustration
    IIIAT1, N2, M0AnyAnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T2, N1, M0AnyAnyT2 = Tumor invades the muscularis propria.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    IIIBT2, N2, M0AnyAnyT2 = Tumor invades the muscularis propria.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T3, N1–N2, M0AnyAnyT3 = Tumor invades adventitia.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T4a, N0–1, M0AnyAny–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Stage TNM GradeTumor LocationDescriptionIllustration
    IVAT4a, N2, M0AnyAny–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T4b, N0–2, M0AnyAny–T4b = Tumor invades other adjacent structures, such as the aorta, vertebral body, or airway.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Any T, N3, M0AnyAnyAny T = See Table 1.
    N3 = Metastasis in seven or more regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    IVBAny T, Any N, M1AnyAnyAny T = See Table 1.
    Any N = See Table 1.
    M1 = Distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; N/A = not applicable; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    Table 3. Definitions of pTNM Stages IA and IB for Squamous Cell Carcinoma of the EsophagusTable 4. Definitions of pTNM Stages IIA and IIB for Squamous Cell Carcinoma of the EsophagusTable 5. Definitions of pTNM Stages IIIA and IIIB for Squamous Cell Carcinoma of the EsophagusTable 6. Definitions of pTNM Stages IVA and IVB for Squamous Cell Carcinoma of the EsophagusCurrent Clinical Trials
    StageTNMGradeTumor LocationDescriptionIllustration
    StageTNMGradeTumor LocationDescriptionIllustration
    Stage TNM GradeTumor LocationDescriptionIllustration
    Stage TNM GradeTumor LocationDescriptionIllustration
    IATia, N0, M0G1Any–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    Any L = See Table 1.
    T1a, N0, M0GXAny–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    IBT1a, N0, M0G2–G3Any–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Any L = See Table 1.
    T1b, N0, M0G1–G3Any–T1b = Tumor invades the submucosa.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Any L = See Table 1.
    T1b, N0, M0GXAny–T1b = Tumor invades the submucosa.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    T2, N0, M0G1AnyT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    Any L = See Table 1.
    StageTNMGradeTumor LocationDescriptionIllustration
    IIAT2, N0, M0GXAnyT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    T2, N0, M0G2–G3AnyT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Any L = See Table 1.
    T3, N0, M0AnyLowerT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Lower = Lower border of inferior pulmonary vein to stomach, including gastroesophageal junction.
    T3, N0, M0G1Upper/middleT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    Upper = Cervical esophagus to lower border of azygos vein.
    Middle = Lower border of azygos vein to lower border of inferior pulmonary vein.
    IIBT3, N0, M0G2–G3Upper/middleT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Upper = Cervical esophagus to lower border of azygos vein.
    Middle = Lower border of azygos vein to lower border of inferior pulmonary vein.
    T3, N0, M0GXAnyT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    T3, N0, M0AnyLocation XT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Location X = Location unknown.
    T1, N1, M0AnyAnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Stage TNM GradeTumor LocationDescriptionIllustration
    IIIAT1, N2, M0AnyAnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T2, N1, M0AnyAnyT2 = Tumor invades the muscularis propria.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    IIIBT2, N2, M0AnyAnyT2 = Tumor invades the muscularis propria.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T3, N1–N2, M0AnyAnyT3 = Tumor invades adventitia.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T4a, N0–1, M0AnyAny–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Stage TNM GradeTumor LocationDescriptionIllustration
    IVAT4a, N2, M0AnyAny–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T4b, N0–2, M0AnyAny–T4b = Tumor invades other adjacent structures, such as the aorta, vertebral body, or airway.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Any T, N3, M0AnyAnyAny T = See Table 1.
    N3 = Metastasis in seven or more regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    IVBAny T, Any N, M1AnyAnyAny T = See Table 1.
    Any N = See Table 1.
    M1 = Distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    Table 4. Definitions of pTNM Stages IIA and IIB for Squamous Cell Carcinoma of the EsophagusTable 5. Definitions of pTNM Stages IIIA and IIIB for Squamous Cell Carcinoma of the EsophagusTable 6. Definitions of pTNM Stages IVA and IVB for Squamous Cell Carcinoma of the EsophagusCurrent Clinical Trials
    StageTNMGradeTumor LocationDescriptionIllustration
    Stage TNM GradeTumor LocationDescriptionIllustration
    Stage TNM GradeTumor LocationDescriptionIllustration
    IIAT2, N0, M0GXAnyT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    T2, N0, M0G2–G3AnyT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Any L = See Table 1.
    T3, N0, M0AnyLowerT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Lower = Lower border of inferior pulmonary vein to stomach, including gastroesophageal junction.
    T3, N0, M0G1Upper/middleT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    Upper = Cervical esophagus to lower border of azygos vein.
    Middle = Lower border of azygos vein to lower border of inferior pulmonary vein.
    IIBT3, N0, M0G2–G3Upper/middleT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G2 = Moderately differentiated.
    G3 = Poorly differentiated, undifferentiated.
    Upper = Cervical esophagus to lower border of azygos vein.
    Middle = Lower border of azygos vein to lower border of inferior pulmonary vein.
    T3, N0, M0GXAnyT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    Any L = See Table 1.
    T3, N0, M0AnyLocation XT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Location X = Location unknown.
    T1, N1, M0AnyAnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Stage TNM GradeTumor LocationDescriptionIllustration
    IIIAT1, N2, M0AnyAnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T2, N1, M0AnyAnyT2 = Tumor invades the muscularis propria.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    IIIBT2, N2, M0AnyAnyT2 = Tumor invades the muscularis propria.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T3, N1–N2, M0AnyAnyT3 = Tumor invades adventitia.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T4a, N0–1, M0AnyAny–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Stage TNM GradeTumor LocationDescriptionIllustration
    IVAT4a, N2, M0AnyAny–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T4b, N0–2, M0AnyAny–T4b = Tumor invades other adjacent structures, such as the aorta, vertebral body, or airway.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Any T, N3, M0AnyAnyAny T = See Table 1.
    N3 = Metastasis in seven or more regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    IVBAny T, Any N, M1AnyAnyAny T = See Table 1.
    Any N = See Table 1.
    M1 = Distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    Table 5. Definitions of pTNM Stages IIIA and IIIB for Squamous Cell Carcinoma of the EsophagusTable 6. Definitions of pTNM Stages IVA and IVB for Squamous Cell Carcinoma of the EsophagusCurrent Clinical Trials
    Stage TNM GradeTumor LocationDescriptionIllustration
    Stage TNM GradeTumor LocationDescriptionIllustration
    IIIAT1, N2, M0AnyAnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T2, N1, M0AnyAnyT2 = Tumor invades the muscularis propria.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    IIIBT2, N2, M0AnyAnyT2 = Tumor invades the muscularis propria.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T3, N1–N2, M0AnyAnyT3 = Tumor invades adventitia.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T4a, N0–1, M0AnyAny–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Stage TNM GradeTumor LocationDescriptionIllustration
    IVAT4a, N2, M0AnyAny–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T4b, N0–2, M0AnyAny–T4b = Tumor invades other adjacent structures, such as the aorta, vertebral body, or airway.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Any T, N3, M0AnyAnyAny T = See Table 1.
    N3 = Metastasis in seven or more regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    IVBAny T, Any N, M1AnyAnyAny T = See Table 1.
    Any N = See Table 1.
    M1 = Distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.
    Table 6. Definitions of pTNM Stages IVA and IVB for Squamous Cell Carcinoma of the EsophagusCurrent Clinical Trials
    Stage TNM GradeTumor LocationDescriptionIllustration
    IVAT4a, N2, M0AnyAny–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T4b, N0–2, M0AnyAny–T4b = Tumor invades other adjacent structures, such as the aorta, vertebral body, or airway.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    Any T, N3, M0AnyAnyAny T = See Table 1.
    N3 = Metastasis in seven or more regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    IVBAny T, Any N, M1AnyAnyAny T = See Table 1.
    Any N = See Table 1.
    M1 = Distant metastasis.
    Any G = See Table 1.
    Any L = See Table 1.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; L = tumor location; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    bLocation is defined by the position of the epicenter of the tumor in the esophagus.

    Current Clinical Trials

    Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.

    Staging for adenocarcinoma of the esophagus

    Table 7. Definitions of pTNM Stage 0 for Adenocarcinoma of the EsophagusTable 8. Definitions of pTNM Stages IA, IB, and IC for Adenocarcinoma of the EsophagusTable 9. Definitions of pTNM Stages IIA and IIB for Adenocarcinoma of the EsophagusTable 10. Definitions of pTNM Stages IIIA and IIIB for Adenocarcinoma of the EsophagusTable 11. Definitions of pTNM Stages IVA and IVB for Adenocarcinoma of the Esophagus
    StageTNMGrade DescriptionIllustration
    StageTNM GradeDescriptionIllustration
    StageTNMGrade DescriptionIllustration
    StageTNMGradeDescriptionIllustration
    StageTNMGradeDescriptionIllustration
    0Tis, N0, M0N/ATis = High-grade dysplasia, defined as malignant cells confined to the epithelium by the basement membrane.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    StageTNM GradeDescriptionIllustration
    IAT1a, N0, M0G1–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    T1a, N0, M0GX–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    IBT1a, N0, M0G2–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G2 = Moderately differentiated.
    T1b, N0, M0G1–2–T1b = Tumor invades the submucosa.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    G2 = Moderately differentiated.
    T1b, N0, M0GX–T1b = Tumor invades the submucosa.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    ICT1, N0, M0G3T1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G3 = Poorly differentiated, undifferentiated.
    T2, N0, M0G1–2T2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    G2 = Moderately differentiated.
    StageTNMGrade DescriptionIllustration
    IIAT2, N0, M0G3T2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G3 = Poorly differentiated, undifferentiated.
    T2, N0, M0GXT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    IIBT1, N1, M0AnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T3, N0, M0AnyT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    Any G = See Table 1.
    StageTNMGradeDescriptionIllustration
    IIIAT1, N2, M0AnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T2, N1, M0AnyT2 = Tumor invades the muscularis propria.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    IIIBT2, N2, M0AnyT2 = Tumor invades the muscularis propria.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T3, N1–2, M0AnyT3 = Tumor invades adventitia.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T4a, N0–1, M0Any–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    StageTNMGradeDescriptionIllustration
    IVAT4a, N2, M0Any–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T4b, N0–2, M0Any–T4b = Tumor invades other adjacent structures, such as the aorta, vertebral body, or airway.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any T, N3, M0AnyAny T = See Table 1.
    N3 = Metastasis in seven or more regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    IVBAny T, Any N, M1AnyAny T = See Table 1.
    Any N = See Table 1.
    M1 = Distant metastasis.
    Any G = See Table 1.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; N/A = not applicable; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    Table 8. Definitions of pTNM Stages IA, IB, and IC for Adenocarcinoma of the EsophagusTable 9. Definitions of pTNM Stages IIA and IIB for Adenocarcinoma of the EsophagusTable 10. Definitions of pTNM Stages IIIA and IIIB for Adenocarcinoma of the EsophagusTable 11. Definitions of pTNM Stages IVA and IVB for Adenocarcinoma of the Esophagus
    StageTNM GradeDescriptionIllustration
    StageTNMGrade DescriptionIllustration
    StageTNMGradeDescriptionIllustration
    StageTNMGradeDescriptionIllustration
    IAT1a, N0, M0G1–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    T1a, N0, M0GX–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    IBT1a, N0, M0G2–T1a = Tumor invades the lamina propria or muscularis mucosae.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G2 = Moderately differentiated.
    T1b, N0, M0G1–2–T1b = Tumor invades the submucosa.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    G2 = Moderately differentiated.
    T1b, N0, M0GX–T1b = Tumor invades the submucosa.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    ICT1, N0, M0G3T1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G3 = Poorly differentiated, undifferentiated.
    T2, N0, M0G1–2T2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G1 = Well differentiated.
    G2 = Moderately differentiated.
    StageTNMGrade DescriptionIllustration
    IIAT2, N0, M0G3T2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G3 = Poorly differentiated, undifferentiated.
    T2, N0, M0GXT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    IIBT1, N1, M0AnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T3, N0, M0AnyT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    Any G = See Table 1.
    StageTNMGradeDescriptionIllustration
    IIIAT1, N2, M0AnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T2, N1, M0AnyT2 = Tumor invades the muscularis propria.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    IIIBT2, N2, M0AnyT2 = Tumor invades the muscularis propria.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T3, N1–2, M0AnyT3 = Tumor invades adventitia.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T4a, N0–1, M0Any–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    StageTNMGradeDescriptionIllustration
    IVAT4a, N2, M0Any–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T4b, N0–2, M0Any–T4b = Tumor invades other adjacent structures, such as the aorta, vertebral body, or airway.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any T, N3, M0AnyAny T = See Table 1.
    N3 = Metastasis in seven or more regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    IVBAny T, Any N, M1AnyAny T = See Table 1.
    Any N = See Table 1.
    M1 = Distant metastasis.
    Any G = See Table 1.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    Table 9. Definitions of pTNM Stages IIA and IIB for Adenocarcinoma of the EsophagusTable 10. Definitions of pTNM Stages IIIA and IIIB for Adenocarcinoma of the EsophagusTable 11. Definitions of pTNM Stages IVA and IVB for Adenocarcinoma of the Esophagus
    StageTNMGrade DescriptionIllustration
    StageTNMGradeDescriptionIllustration
    StageTNMGradeDescriptionIllustration
    IIAT2, N0, M0G3T2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    G3 = Poorly differentiated, undifferentiated.
    T2, N0, M0GXT2 = Tumor invades the muscularis propria.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    GX = Grade cannot be assessed.
    IIBT1, N1, M0AnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T3, N0, M0AnyT3 = Tumor invades adventitia.
    N0 = No regional lymph node metastasis.
    M0 = No distant metastasis.
    Any G = See Table 1.
    StageTNMGradeDescriptionIllustration
    IIIAT1, N2, M0AnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T2, N1, M0AnyT2 = Tumor invades the muscularis propria.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    IIIBT2, N2, M0AnyT2 = Tumor invades the muscularis propria.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T3, N1–2, M0AnyT3 = Tumor invades adventitia.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T4a, N0–1, M0Any–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    StageTNMGradeDescriptionIllustration
    IVAT4a, N2, M0Any–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T4b, N0–2, M0Any–T4b = Tumor invades other adjacent structures, such as the aorta, vertebral body, or airway.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any T, N3, M0AnyAny T = See Table 1.
    N3 = Metastasis in seven or more regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    IVBAny T, Any N, M1AnyAny T = See Table 1.
    Any N = See Table 1.
    M1 = Distant metastasis.
    Any G = See Table 1.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    Table 10. Definitions of pTNM Stages IIIA and IIIB for Adenocarcinoma of the EsophagusTable 11. Definitions of pTNM Stages IVA and IVB for Adenocarcinoma of the Esophagus
    StageTNMGradeDescriptionIllustration
    StageTNMGradeDescriptionIllustration
    IIIAT1, N2, M0AnyT1 = Tumor invades the lamina propria, muscularis mucosae, or submucosa.
    –T1a = Tumor invades the lamina propria or muscularis mucosae.
    –T1b = Tumor invades the submucosa.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T2, N1, M0AnyT2 = Tumor invades the muscularis propria.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    IIIBT2, N2, M0AnyT2 = Tumor invades the muscularis propria.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T3, N1–2, M0AnyT3 = Tumor invades adventitia.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T4a, N0–1, M0Any–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    StageTNMGradeDescriptionIllustration
    IVAT4a, N2, M0Any–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T4b, N0–2, M0Any–T4b = Tumor invades other adjacent structures, such as the aorta, vertebral body, or airway.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any T, N3, M0AnyAny T = See Table 1.
    N3 = Metastasis in seven or more regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    IVBAny T, Any N, M1AnyAny T = See Table 1.
    Any N = See Table 1.
    M1 = Distant metastasis.
    Any G = See Table 1.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.
    Table 11. Definitions of pTNM Stages IVA and IVB for Adenocarcinoma of the Esophagus
    StageTNMGradeDescriptionIllustration
    IVAT4a, N2, M0Any–T4a = Tumor invades the pleura, pericardium, azygos vein, diaphragm, or peritoneum.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    T4b, N0–2, M0Any–T4b = Tumor invades other adjacent structures, such as the aorta, vertebral body, or airway.
    N0 = No regional lymph node metastasis.
    N1 = Metastasis in one or two regional lymph nodes.
    N2 = Metastasis in three to six regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    Any T, N3, M0AnyAny T = See Table 1.
    N3 = Metastasis in seven or more regional lymph nodes.
    M0 = No distant metastasis.
    Any G = See Table 1.
    IVBAny T, Any N, M1AnyAny T = See Table 1.
    Any N = See Table 1.
    M1 = Distant metastasis.
    Any G = See Table 1.
    T = primary tumor; N = regional lymph nodes; M = distant metastasis; G = grade; p = pathological.
    aReprinted with permission from AJCC: Esophageal and esophagogastric junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 185–202.

    ReferenceSection

  • Ziegler K, Sanft C, Zeitz M, et al.: Evaluation of endosonography in TN staging of oesophageal cancer. Gut 32 (1): 16-20, 1991.
  • Tio TL, Coene PP, den Hartog Jager FC, et al.: Preoperative TNM classification of esophageal carcinoma by endosonography. Hepatogastroenterology 37 (4): 376-81, 1990.
  • Vazquez-Sequeiros E, Norton ID, Clain JE, et al.: Impact of EUS-guided fine-needle aspiration on lymph node staging in patients with esophageal carcinoma. Gastrointest Endosc 53 (7): 751-7, 2001.
  • Bonavina L, Incarbone R, Lattuada E, et al.: Preoperative laparoscopy in management of patients with carcinoma of the esophagus and of the esophagogastric junction. J Surg Oncol 65 (3): 171-4, 1997.
  • Sugarbaker DJ, Jaklitsch MT, Liptay MJ: Thoracoscopic staging and surgical therapy for esophageal cancer. Chest 107 (6 Suppl): 218S-223S, 1995.
  • Luketich JD, Schauer P, Landreneau R, et al.: Minimally invasive surgical staging is superior to endoscopic ultrasound in detecting lymph node metastases in esophageal cancer. J Thorac Cardiovasc Surg 114 (5): 817-21; discussion 821-3, 1997.
  • Krasna MJ, Reed CE, Nedzwiecki D, et al.: CALGB 9380: a prospective trial of the feasibility of thoracoscopy/laparoscopy in staging esophageal cancer. Ann Thorac Surg 71 (4): 1073-9, 2001.
  • Flamen P, Lerut A, Van Cutsem E, et al.: Utility of positron emission tomography for the staging of patients with potentially operable esophageal carcinoma. J Clin Oncol 18 (18): 3202-10, 2000.
  • Flamen P, Van Cutsem E, Lerut A, et al.: Positron emission tomography for assessment of the response to induction radiochemotherapy in locally advanced oesophageal cancer. Ann Oncol 13 (3): 361-8, 2002.
  • Weber WA, Ott K, Becker K, et al.: Prediction of response to preoperative chemotherapy in adenocarcinomas of the esophagogastric junction by metabolic imaging. J Clin Oncol 19 (12): 3058-65, 2001.
  • van Westreenen HL, Westerterp M, Bossuyt PM, et al.: Systematic review of the staging performance of 18F-fluorodeoxyglucose positron emission tomography in esophageal cancer. J Clin Oncol 22 (18): 3805-12, 2004.
  • Meyers BF, Downey RJ, Decker PA, et al.: The utility of positron emission tomography in staging of potentially operable carcinoma of the thoracic esophagus: results of the American College of Surgeons Oncology Group Z0060 trial. J Thorac Cardiovasc Surg 133 (3): 738-45, 2007.
  • Rice TW, Kelsen D, Blackstone EH, et al.: Esophagus and Esophagogastric Junction. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 185-202.
  • Korst RJ, Rusch VW, Venkatraman E, et al.: Proposed revision of the staging classification for esophageal cancer. J Thorac Cardiovasc Surg 115 (3): 660-69; discussion 669-70, 1998.
  • 食管癌的临床治疗(成人)(PDQ®)

    食管癌治疗方法概述

    针对微小可切除的食管癌,单独手术切除有可能取得治愈。与此相反,用于局部晚期可手术切除食管癌的治疗手段近年来得到显著改善。由于远处转移和局部复发的风险,化疗、放疗及手术切除相结合的多学科治疗已成为标准治疗。

    多种治疗方法的联合应用正处于临床评估阶段,包括:

  • 单独手术治疗
  • 化疗
  • 放疗
  • 采用以下联合治疗后,部分患者得到有效缓解,如

  • 手术
  • 化疗
  • 放疗
  • 支架
  • 光动力治疗
  • 内镜联合 Nd:YAG激光
  • 表12.食管癌的标准治疗方法手术手术(巴雷特食管)
    分期(TNM分期标准)治疗方法
    手术
    内镜下切除
    术前放化疗加手术
    单独手术治疗
    术前放化疗加手术
    单独手术治疗
    术前化疗加手术
    术前化疗加手术
    术前放化疗加手术
    术前化疗加手术
    术前化疗加手术
    术前放化疗加手术(用于IVA期食管癌 )
    化疗用于转移性下段食管腺癌可以部分缓解。
    Nd:YAG腔内肿瘤破坏或电凝
    内镜下置入支架可缓解吞咽困难
    放疗联合/不联合腔内置管和扩张术
    腔内近距离放射治疗可缓解吞咽困难
    任何标准治疗的缓和应用,包括支持治疗。

    手术

    手术(巴雷特食管)

    食管腺癌的巴雷特化生的发病率提示,巴雷特食管是一类癌前病变。巴雷特化生患者应行内镜监测,有助于早期发现腺癌,即更有可能进行治愈性性手术切除。对高级别不典型增生的巴雷特化生患者,强烈推荐手术切除。

    手术(食管癌)

    食管癌患者的生存率较低。可手术切除的食管癌患者采用手术,其5年生存率为5%-30%。早期食管癌患者的生存率稍高。

    位于食管黏膜层和黏膜下层无症状的小肿瘤只会偶然发现。这些小肿瘤应采用手术切除。一旦出现症状(如多数患者出现吞咽困难),食管癌已侵犯或侵透固有肌层,可能伴淋巴结转移或其他脏器转移

    部分食管梗阻的患者,可置入膨胀式金属支架缓解吞咽困难。

    或者,如果患者有弥漫性病变或不宜采用手术,可采用放疗。也曾有缓解吞咽困难的替代疗法报道,包括激光治疗、电凝用来破坏腔内肿瘤。

    完全性食管梗阻,但未出现全身转移的临床迹象,手术切除并上提胃来代替食管是缓解吞咽困难的传统方法。

    食管癌根治切除术的最佳切除方法还不得而知。一种方法提倡采用经膈肌裂孔切除吻合术,将胃与颈段食管相吻合。第二种方法是移动腹腔内胃,经胸切除食管,将胃与胸上段食管或颈段食管进行吻合。有项研究结果显示,经膈肌裂孔食管切除术的术后并发症发生率低于经胸食管切除术伴扩大完整淋巴清扫术。然而,中位总体无疾病生存期和质量调整生存无明显差异。

    同样,采用经过验证的生活质量(QOL)评分系统计算的长期生活质量未见差异。

    近期,微创方法治疗具有切口小、降低术中出血量、术后并发症更少、住院时间缩短等优势。然而,这种方法在手术切缘阴性、淋巴结切除的准确性、长期临床转归等方面的效果并未完全明确。

    在美国,食管癌患者的中位诊断年龄为67岁。

    一项关于某个外科团队长达17年共505例连续患者的回顾性研究结果显示,年龄超过70岁的患者同70岁以下患者相比,二组患者在围手术期死亡率、中位生存期、食管癌切除术治疗吞咽困难方面并无差异。

    [证据级别:3iiA和3iiB]在本项研究中,所有患者均在考虑手术风险后行手术治疗。单纯年龄一个因素,并不是潜在手术切除癌症患者选择治疗方法的决定性因素。

    食管癌采用手术治疗的围术期死亡率不足10%。

    在放化疗联合应用降低围手术期死亡率及缓解吞咽困难方面,已有相关报道。

    术前放化疗

    基于多项随机临床试验的研究结果,术前放化疗已成为IB、II、III、IVA期食管癌的标准治疗。

    已有III 期临床试验将术前同步放化疗与单独手术用于食管癌患者进行对比研究。

    [证据级别:1iiA]由于与早期随机研究结果相悖,新辅助放化疗的临床获益一直存有争议。

    然而,食管癌的术前放化疗研究(CROSS)结果明确显示,与单独手术切除相比,术前放化疗可显著改善局部晚期食管癌患者的生存期。

    对于术前放化疗用于早期肿瘤的观点一直存有争议。尽管CROSS研究纳入了早期病例,法国国家消化道癌症研究所(FFCD) 9901研究(NCT00047112),

    该研究仅纳入早期( 临床I或II期)病例,该组患者的生存期并未明显改善。

    证据(术前放化疗):

  • CROSS研究随机挑选366例有手术可能的食管癌和胃-食管交界处癌病例,一组采取单独手术切除,另一组临床给予卡铂周疗(滴定剂量至AUC(药时曲线下面积),2 mg/mL/分钟)联合紫杉醇(按体表面积计算,50 mg/m2),同步放疗(剂量41.4 Gy,分23次),持续用药5周。本项CROSS研究招募的患者多为腺癌(75%)。
  • [证据级别:2iiA]
  • 中位随诊时间为84个月,结果显示术前放化疗有助于延长中位总体生存期,单独手术组为24个月,联合治疗组48.6个月(风险比[HR]0.68,95%可信区间[CI]0.53-0.88,P=.003)。食管鳞状细胞癌联合治疗组的中位总体生存期是81.6个月,单独手术组21.1个月(HR 0.48,95%CI0.28-0.83,log-rank P=.008)。食管腺癌联合治疗组的中位总体生存期是43.2个月,单独手术组27.1个月(HR 0.73,95%CI0.55-0.98,log-rank P=.038)。
  • 此外,术前放化疗可改善R0切除率(92% vs. 69%,P<.001)。R0是指肿瘤完全切除且切缘1mm以内。
  • 术前放化疗组患者的病理学完全缓解率为29%。腺癌患者的病理学完全缓解率为23%,鳞癌患者为49%(P=.008)。
  • 两组的术后并发症和住院期间死亡率无差异。放化疗组最常见的不良反应是白细胞减少症(6%)和中性粒细胞减少症(2%)。最常见的其他不良反应是厌食(5%)和疲劳(3%)。
  • 中位随诊84个月,5年无进展生存率(PFS)术前放化疗组(44%)优于单独手术组(27%)(HR, 0.61 [0.47–0.78])。术前放化疗将局部区域性复发从34%降至14%(P<.001),腹膜转移14%降至4%(P<.001)。术前放化疗组的血行转移略优于单独手术组((35% vs. 29%; P=.025),但具有统计学意义。
  • [证据级别:1iiDii]
  • 一项多中心前瞻性随机试验将食管鳞癌患者分为术前化疗(如顺铂)联合放疗(剂量37Gy,每次3.7Gy)组和单独手术组进行对比研究。
  • [证据级别:1iiA]
  • 研究结果显示总体生存期无差异,联合治疗组的围手术期死亡率明显高于对照组(12% vs. 4%)。
  • 一项单中心III期临床试验对食管腺癌进行了研究,一组患者采用联合治疗,即氟尿嘧啶(5-FU)与顺铂联合放疗(40Gy,每次2.67Gy)加手术治疗。另一组单独手术治疗。
  • [证据级别:1iiA]
  • 结果显示,联合治疗组的生存期为16个月,单独手术组为11个月。
  • 还有一项单中心试验,随机将患者(75%食管腺癌)分为两组,一组患者给予5-FU、顺铂、长春碱联合放疗(每次1.5Gy,每日两次,总剂量45 Gy),联合手术治疗。对照组仅行食管癌切除术。
  • [证据级别:1iiA]
  • 中位随诊时间超过8年,联合治疗组和单独手术组的中位生存期(17.6个月 vs. 16.9个月)、3年总体生存率(16% vs. 30%)或3年无疾病进展生存率(16% vs. 28%)无显著差异。
  • 一项组间试验(CALGB-9781 [NCT00003118])纳入475例可切除的胸段食管鳞癌和腺癌患者,随机分为两组,一组给予术前放化疗(5-FU、顺铂,放疗剂量50.4 Gy)并行食管切除和淋巴清扫术,另一组单独行手术治疗。由于疗效不佳,该临床试验终止。然而,中位随诊6年后发现仍有56例患者存活。
  • [证据级别:1iiA]
  • 联合治疗组的中位生存期是4.48年(95% CI;范围, 2.4年未进行评估),单独手术组1.79年(95% CI, 1.41–2.59年),P=0.002。联合治疗组的5年生存率是39%(95% CI, 21%–57%),单独手术组为16%(95% CI, 5%–33%)。
  • 为了进一步评估新辅助放化疗对早期食管癌的影响,FFCD 9901将195例临床I和II期食管癌患者进行了随机分配,一组给予单独手术治疗,另一组给予新辅助放化疗(放疗剂量45Gy,分为25次;化疗5-FU[800 mg/m2]联合顺铂[75 mg/m2])联合手术治疗。
  • [证据级别:1iiA]
  • 中期分析显示,由于无效,该试验提早终止。
  • 中位随诊时间94个月,放化疗组的3年总体生存率与对照组无明显差别(48% vs. 53%,P=.94)。而围手术期死亡率高于对照组(11.1% vs. 3.4%,P=.049)。
  • 术前化疗

    一些术前化疗的随机试验正处于研究阶段。多项研究结果显示术前化疗患者的生存期优于单独手术组。

    然而,一项大规模随机研究,并未证实术前化疗能延长生存期。

    与单独术前化疗相比,术前放化疗可提高病理缓解率并改善临床转归。

    证据(术前化疗):

  • 一项组间研究(NCT00525785)随机将440例局灶部可切除的食管癌患者(含各种病理类型)进行分组,一组采用化疗联合手术治疗,即术前给予3个周期的5-FU和顺铂治疗,并在手术后继续采用2个周期的化疗,另一组采用单独手术切除。
  • [证据级别:1iiA]
  • 中位随诊55个月后,化疗联合手术组的中位生存期(14.9个月)与单独手术组(16.1个月)无明显差异。两组患者的2年生存率分别为35%和37%。
  • 联用化疗不会增加术后并发症风险。
  • 医学研究委员会的食管癌工作小组随机选择802例可切除的食管癌患者(含各种病理类型),将患者分为两组,一组给予术前5-FU联合顺铂治疗2周期,另一组行单独手术切除。
  • [证据级别:1iiA]
  • 中位随诊时间37个月,术前化疗组的中位生存期比单独手术组延长3.5个月(16.8个月 vs. 13.3个月,95% CI,1.0–6.5个月)。2年总体生存率分别为43%和34%,联合治疗组比单独手术组高9%(95% CI,3–14个月)。
  • 该组间研究和术前化疗的研究结果面临挑战,因为未见T和N分期报道,同时主治医生未提出预随机方案和放疗等信息

  • 日本临床肿瘤协作组将330例临床II和III期(除T4外)鳞癌患者随机分组,一组术前给予顺铂联合5-FU化疗2个周期,另一组术后给予同样的化疗方案。在患者自然累积后,进行中期分析。尽管未达到PFS的主要终点,术前化疗组的总体生存期仍具有明显优势(P=.01)。最终结果是本项研究被数据与安全监测委员会提早叫停。
  • [证据级别:1iiC]
  • 中位随诊时间61个月,术前化疗组的5年生存率为55%,术后化疗组为43%(P=.04)。两组患者的5年无进展生存率无明显差异,分别为39%和44%(P=.22)。
  • 此外,两组患者的术后并发症和治疗相关的毒性反应无明显差异。
  • Fédération Nationale des Centres de Lutte contre le Cancer和FFCD随机将224例可切除的食管下段腺癌、食管胃结合部腺癌和胃腺癌患者进行分组,一组给予围手术期化疗联合手术治疗(n=113),对照组行单独手术切除(n=111)。化疗包括术前2-3个周期的顺铂静脉输注(100 mg/m2),D1;5-FU持续静脉给药(800 mg/m2),D1-5,Q28d。术后给予同一方案化疗3-4个周期。
  • [证据级别:1iiA]
  • 围手术期化疗可改善患者的5年总体生存率(5年总体生存率, 38% vs. 24%;HR, 0.69;P=.02)。
  • 38%围手术期化疗的患者出现3-4级毒性反应,术后并发症发生率未见升高。
  • 食管胃结合部腺癌的术前化疗或放化疗试验(POET)对放疗联用化疗的额外临床获益进行了评估。患者随机分组,一组给予诱导性化疗(15周)再给予手术,另一组先给予化疗(12周)再给予放化疗(3周)和手术治疗。
  • [证据级别:1iiA]
  • 由于效果不佳?,研究提早结束。共126例患者随机进行了分组。
  • 术前放疗组的3年生存率为27%-47%(log-rank,P=.07)。放化疗组的术后并发症发生率未见明显升高(10.2% vs. 3.8%, P=.26)。
  • 根治性放化疗

    针对不可手术或不可切除的患者,根治性放化疗的临床疗效已得到大量随机对照试验的证实。

    针对食管鳞癌,单独同步放化疗与术前放化疗联合手术的临床疗效无差异。

    证据(根治性放化疗)

  • 一项放疗肿瘤工作组试验(RTOG-8501)随机将患者进行分配,一组给予放化疗联合治疗,另一组给予单独放疗。单独放疗组(总剂量64Gy,32次);放化疗联合组,放疗(总剂量50Gy,25次),同步给予化疗顺铂(75 mg/m2),第1和5周持续输注5-FU(1,000 mg/m2,D1-4),间隔3周后再行2个周期化疗。
  • [证据级别:1iiA]
  • 联合治疗组的5年生存率改善(27% vs. 0%)。
  • 8年随诊结果显示,放化疗患者的总体生存率为22%。
  • 组间-0123(RTOG-9405[NCT00002631])研究的目的是改善RTOG-8501的结果。组间-0123将236例局限期食管癌患者进行随机分组,一组采用高剂量放疗(64.8 Gy)及4个周期的顺铂联合5-FU治疗,另一组采用常规剂量(50.4 Gy)放疗联合同一化疗方案。
  • [证据级别:1iiA]
  • 患者不断增加至298例,由于中期分析显示采用高剂量放疗很有可能无明显优势,因此本项试验于1999年结束。
  • 中位随诊2年后,高剂量组和常规剂量组的中位生存期(13个月 vs. 18个月)、2年生存率(31% vs. 40%)和局部或区域复发(56% vs. 52%)方面未见明显差异。
  • 高剂量放疗组的死亡率明显高于常规剂量组(9% vs. 2%)。高剂量组死亡的11例患者中,有7例患者已接受的放疗剂量为50.4Gy或不足50.4Gy。
  • 一项东部肿瘤协作组织的临床试验(EST-1282)对135例患者进行了评估。
  • [证据级别:1iiA]
  • 试验结果显示化疗联用放疗的2年生存率高于单独放疗组,这一结果与组间研究一致。
  • PRODIGE5/ACCORD17 (NCT00861094) 试验旨在对奥沙利铂、氟尿嘧啶、亚叶酸钙(FLOFOX)与以5-FU,顺铂为基础的化疗用于根治性放化疗治疗局限期食管癌进行对比研究。在本项多中心、随机II和III期临床试验中,267例患者随机分为两组,一组给予6个周期的FLOFOX方案(3个周期同步放疗),奥沙利铂(85 mg/m2)、亚叶酸钙(200 mg/m2)、5-FU囊注(400 mg/m2)及5-Fu持续输注(1,600 mg/m2,输注46小时);另一组给予4个周期5-FU(1,000 mg/m2,D1-4)和顺铂(75 mg/m2,D1)。两组患者均行同一种放疗方案(总剂量50Gy,25次)。
  • [证据级别:1iiDiii]
  • 中位随诊时间25.3个月,FOLFOX治疗组与5-FU联合顺铂化疗组的无疾病进展生存期(PFS)无明显差异,分别为9.7个月和9.4个月(P=.64)。
  • FOLFOX治疗组出现1例毒性反应导致的死亡病例。5-FU联合顺铂化疗组有6例。
  • 两组患者的3和4级不良反应事件无明显差异。各类毒性反应中,感觉异常、神经性病变,AST和ALT升高多发于FOLFOX治疗组。而血清肌酐升高、脱发症多发于5-FU联合顺铂化疗组。
  • 一项德国III期临床试验针对T3和T4食管鳞癌患者的治疗进行了对比研究,一组(A组)给予诱导性化疗(3个疗程的5-FU囊注、亚叶酸钙、依托泊苷和顺铂),其后采用同步放化疗(顺铂、依托泊苷,放疗剂量40Gy),之后行手术治疗。另一组(B组)采用诱导性化疗,其后给予同步放化疗(至少65 Gy),但未予手术治疗。总体生存期是主要结果。
  • [证据级别:1iiA]
  • 对172个随机分配的病例资料进行分析,结果显示,两组患者的2年总生存率无显著差异。(A组 39.9%;95% CI, 29.4%–50.4%;B组 35.4%;95% CI,25.2%–45.6%;log-rank检测结果为0.15,P<.007)。
  • 手术组的2年局部无进展生存率为64.3%(95% CI, 52.1%–76.5%),放化疗组为40.7%(95% CI, 28.9%–52.5%)。两者具有显著性差异(HR,B组:A组 2.1;95% CI,1.3–3.5;P<.003)。
  • 手术组的治疗相关死亡率高于放化疗组,分别为12.8%和3.5%(P<.03)。
  • FFCD 9102 (NCT00416858)将259例T3N0–1M0胸段食管癌患者随机分为两组,一组给予2个周期的5-FU和顺铂联合治疗(D1-5和D22-26),一组给予常规放疗(总剂量46Gy,4.5周)或分期放疗(15Gy,D1-5和D22-26)。根据临床疗效随机分为手术组(A组)和继续放化疗组(B组,3个周期的5-FU和顺铂联合常规放疗(20Gy)或分期放疗(15Gy))。
  • [证据级别:1iiA]
  • 在259例随机病例中,鳞癌230例(89%),腺癌29例(11%)。
  • 患者随机分配,手术组的2年总体生存率为34%,放化疗组40%(HR, 0.90;P=.44)。两组患者的中位生存期分别为17.7个月和19.3个月。
  • 手术组的3个月死亡率为9.3%,放化疗组为0.8%(P=.002)。
  • 术后放疗

    两项随机临床试验的结果显示,与单独手术相比,术后放疗对总体生存期无临床意义。

    [证据级别:1iiA]。新诊病例应积极接受治疗,或参加各种对照研究的临床试验。处于研究阶段的临床试验的相关信息可登录NCI官网进行查询。

    参考文献

  • Tietjen TG, Pasricha PJ, Kalloo AN: Management of malignant esophageal stricture with esophageal dilation and esophageal stents. Gastrointest Endosc Clin N Am 4 (4): 851-62, 1994.
  • Lightdale CJ, Heier SK, Marcon NE, et al.: Photodynamic therapy with porfimer sodium versus thermal ablation therapy with Nd:YAG laser for palliation of esophageal cancer: a multicenter randomized trial. Gastrointest Endosc 42 (6): 507-12, 1995.
  • Kubba AK: Role of photodynamic therapy in the management of gastrointestinal cancer. Digestion 60 (1): 1-10, 1999 Jan-Feb.
  • Heier SK, Heier LM: Tissue sensitizers. Gastrointest Endosc Clin N Am 4 (2): 327-52, 1994.
  • Bourke MJ, Hope RL, Chu G, et al.: Laser palliation of inoperable malignant dysphagia: initial and at death. Gastrointest Endosc 43 (1): 29-32, 1996.
  • Lerut T, Coosemans W, Van Raemdonck D, et al.: Surgical treatment of Barrett's carcinoma. Correlations between morphologic findings and prognosis. J Thorac Cardiovasc Surg 107 (4): 1059-65; discussion 1065-6, 1994.
  • Kelsen DP, Bains M, Burt M: Neoadjuvant chemotherapy and surgery of cancer of the esophagus. Semin Surg Oncol 6 (5): 268-73, 1990.
  • Saxon RR, Morrison KE, Lakin PC, et al.: Malignant esophageal obstruction and esophagorespiratory fistula: palliation with a polyethylene-covered Z-stent. Radiology 202 (2): 349-54, 1997.
  • Campbell WR, Taylor SA, Pierce GE, et al.: Therapeutic alternatives in patients with esophageal cancer. Am J Surg 150 (6): 665-8, 1985.
  • Mellow MH, Pinkas H: Endoscopic therapy for esophageal carcinoma with Nd:YAG laser: prospective evaluation of efficacy, complications, and survival. Gastrointest Endosc 30 (6): 334-9, 1984.
  • Karlin DA, Fisher RS, Krevsky B: Prolonged survival and effective palliation in patients with squamous cell carcinoma of the esophagus following endoscopic laser therapy. Cancer 59 (11): 1969-72, 1987.
  • Hulscher JB, van Sandick JW, de Boer AG, et al.: Extended transthoracic resection compared with limited transhiatal resection for adenocarcinoma of the esophagus. N Engl J Med 347 (21): 1662-9, 2002.
  • de Boer AG, van Lanschot JJ, van Sandick JW, et al.: Quality of life after transhiatal compared with extended transthoracic resection for adenocarcinoma of the esophagus. J Clin Oncol 22 (20): 4202-8, 2004.
  • Santillan AA, Farma JM, Meredith KL, et al.: Minimally invasive surgery for esophageal cancer. J Natl Compr Canc Netw 6 (9): 879-84, 2008.
  • Ginsberg RJ: Cancer treatment in the elderly. J Am Coll Surg 187 (4): 427-8, 1998.
  • Ellis FH, Williamson WA, Heatley GJ: Cancer of the esophagus and cardia: does age influence treatment selection and surgical outcomes? J Am Coll Surg 187 (4): 345-51, 1998.
  • Bosset JF, Gignoux M, Triboulet JP, et al.: Chemoradiotherapy followed by surgery compared with surgery alone in squamous-cell cancer of the esophagus. N Engl J Med 337 (3): 161-7, 1997.
  • Walsh TN, Noonan N, Hollywood D, et al.: A comparison of multimodal therapy and surgery for esophageal adenocarcinoma. N Engl J Med 335 (7): 462-7, 1996.
  • Urba SG, Orringer MB, Turrisi A, et al.: Randomized trial of preoperative chemoradiation versus surgery alone in patients with locoregional esophageal carcinoma. J Clin Oncol 19 (2): 305-13, 2001.
  • Tepper J, Krasna MJ, Niedzwiecki D, et al.: Phase III trial of trimodality therapy with cisplatin, fluorouracil, radiotherapy, and surgery compared with surgery alone for esophageal cancer: CALGB 9781. J Clin Oncol 26 (7): 1086-92, 2008.
  • van Hagen P, Hulshof MC, van Lanschot JJ, et al.: Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 366 (22): 2074-84, 2012.
  • Oppedijk V, van der Gaast A, van Lanschot JJ, et al.: Patterns of recurrence after surgery alone versus preoperative chemoradiotherapy and surgery in the CROSS trials. J Clin Oncol 32 (5): 385-91, 2014.
  • Mariette C, Dahan L, Mornex F, et al.: Surgery alone versus chemoradiotherapy followed by surgery for stage I and II esophageal cancer: final analysis of randomized controlled phase III trial FFCD 9901. J Clin Oncol 32 (23): 2416-22, 2014.
  • Shapiro J, van Lanschot JJ, Hulshof MC, et al.: Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or junctional cancer (CROSS): long-term results of a randomised controlled trial. Lancet Oncol 16 (9): 1090-8, 2015.
  • Medical Research Council Oesophageal Cancer Working Group: Surgical resection with or without preoperative chemotherapy in oesophageal cancer: a randomised controlled trial. Lancet 359 (9319): 1727-33, 2002.
  • Ando N, Kato H, Igaki H, et al.: A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus (JCOG9907). Ann Surg Oncol 19 (1): 68-74, 2012.
  • Ychou M, Boige V, Pignon JP, et al.: Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol 29 (13): 1715-21, 2011.
  • Kelsen DP, Ginsberg R, Pajak TF, et al.: Chemotherapy followed by surgery compared with surgery alone for localized esophageal cancer. N Engl J Med 339 (27): 1979-84, 1998.
  • Stahl M, Walz MK, Stuschke M, et al.: Phase III comparison of preoperative chemotherapy compared with chemoradiotherapy in patients with locally advanced adenocarcinoma of the esophagogastric junction. J Clin Oncol 27 (6): 851-6, 2009.
  • Cooper JS, Guo MD, Herskovic A, et al.: Chemoradiotherapy of locally advanced esophageal cancer: long-term follow-up of a prospective randomized trial (RTOG 85-01). Radiation Therapy Oncology Group. JAMA 281 (17): 1623-7, 1999.
  • Minsky BD, Pajak TF, Ginsberg RJ, et al.: INT 0123 (Radiation Therapy Oncology Group 94-05) phase III trial of combined-modality therapy for esophageal cancer: high-dose versus standard-dose radiation therapy. J Clin Oncol 20 (5): 1167-74, 2002.
  • Stahl M, Stuschke M, Lehmann N, et al.: Chemoradiation with and without surgery in patients with locally advanced squamous cell carcinoma of the esophagus. J Clin Oncol 23 (10): 2310-7, 2005.
  • Bedenne L, Michel P, Bouché O, et al.: Chemoradiation followed by surgery compared with chemoradiation alone in squamous cancer of the esophagus: FFCD 9102. J Clin Oncol 25 (10): 1160-8, 2007.
  • Smith TJ, Ryan LM, Douglass HO, et al.: Combined chemoradiotherapy vs. radiotherapy alone for early stage squamous cell carcinoma of the esophagus: a study of the Eastern Cooperative Oncology Group. Int J Radiat Oncol Biol Phys 42 (2): 269-76, 1998.
  • Conroy T, Galais MP, Raoul JL, et al.: Definitive chemoradiotherapy with FOLFOX versus fluorouracil and cisplatin in patients with oesophageal cancer (PRODIGE5/ACCORD17): final results of a randomised, phase 2/3 trial. Lancet Oncol 15 (3): 305-14, 2014.
  • Ténière P, Hay JM, Fingerhut A, et al.: Postoperative radiation therapy does not increase survival after curative resection for squamous cell carcinoma of the middle and lower esophagus as shown by a multicenter controlled trial. French University Association for Surgical Research. Surg Gynecol Obstet 173 (2): 123-30, 1991.
  • Fok M, Sham JS, Choy D, et al.: Postoperative radiotherapy for carcinoma of the esophagus: a prospective, randomized controlled study. Surgery 113 (2): 138-47, 1993.
  • Esophageal Cancer Treatment (Adult) (PDQ®)

    Treatment Option Overview for Esophageal Cancer

    For patients with minimally invasive resectable esophageal cancer, surgical resection alone offers the potential for cure. In contrast, therapeutic management for patients with locally advanced resectable esophageal cancer has evolved significantly over the last few decades. Because of the risk of distant metastases and local relapse, multimodality therapy with integration of chemotherapy, radiation therapy, and surgical resection has become the standard of care.

    Combined modality therapies are under clinical evaluation and include the following:

  • Surgery alone.
  • Chemotherapy.
  • Radiation therapy.
  • Effective palliation may be obtained in individual cases with various combinations of the following:

  • Surgery.
  • Chemotherapy.
  • Radiation therapy.
  • Stents.
  • Photodynamic therapy.
  • Endoscopic therapy with Nd:YAG laser.
  • Table 12. Standard Treatment Options for Esophageal CancerSurgerySurgery (Barrett esophagus)
    Stage (TNM Staging Criteria)Treatment Options
    Surgery
    Endoscopic resection
    Chemoradiation therapy followed by surgery
    Surgery alone
    Chemoradiation followed by surgery
    Surgery alone
    Chemotherapy followed by surgery
    Definitive chemoradiation
    Chemoradiation followed by surgery
    Preoperative chemotherapy followed by surgery
    Definitive chemoradiation
    Chemoradiation followed by surgery (for patients with stage IVA disease)
    Chemotherapy, which has provided partial responses for patients with metastatic distal esophageal adenocarcinomas
    Nd:YAG endoluminal tumor destruction or electrocoagulation
    Endoscopic-placed stents to provide palliation of dysphagia
    Radiation therapy with or without intraluminal intubation and dilation
    Intraluminal brachytherapy to provide palliation of dysphagia
    Palliative use of any of the standard therapies, including supportive care

    Surgery

    Surgery (Barrett esophagus)

    The prevalence of Barrett metaplasia in adenocarcinoma of the esophagus suggests that Barrett esophagus is a premalignant condition. Endoscopic surveillance of patients with Barrett metaplasia may detect adenocarcinoma at an earlier stage that is more amenable to curative resection. Strong consideration should be given to resection in patients with high-grade dysplasia in the setting of Barrett metaplasia.

    Surgery (esophageal cancer)

    The survival rate of patients with esophageal cancer is poor. Surgical treatment of resectable esophageal cancers results in 5-year survival rates of 5% to 30%, with higher survival rates in patients with early-stage cancers.

    Asymptomatic small tumors confined to the esophageal mucosa or submucosa are detected only by chance. Surgery is the treatment of choice for these small tumors. Once symptoms are present (e.g., dysphagia, in most cases), esophageal cancers have usually invaded the muscularis propria or beyond and may have metastasized to lymph nodes or other organs.

    In some patients with partial esophageal obstruction, dysphagia may be relieved by placement of an expandable metallic stent

    or by radiation therapy if the patient has disseminated disease or is not a candidate for surgery. Alternative methods of relieving dysphagia have been reported, including laser therapy and electrocoagulation to destroy intraluminal tumor.

    In the presence of complete esophageal obstruction without clinical evidence of systemic metastasis, surgical excision of the tumor with mobilization of the stomach to replace the esophagus has been the traditional means of relieving the dysphagia.

    The optimal surgical approach for radical resection of esophageal cancer is not known. One approach advocates transhiatal esophagectomy with anastomosis of the stomach to the cervical esophagus. A second approach advocates abdominal mobilization of the stomach and transthoracic excision of the esophagus with anastomosis of the stomach to the upper thoracic esophagus or the cervical esophagus. One study concluded that transhiatal esophagectomy was associated with lower morbidity than was transthoracic esophagectomy with extended en bloc lymphadenectomy; however, median overall disease-free and quality-adjusted survival did not differ significantly.

    Similarly, no differences in long-term quality of life (QOL) using validated QOL instruments have been reported.

    More recently, minimally invasive approaches that offer potential advantages of smaller incisions, decreased intraoperative blood loss, fewer postoperative complications, and shorter hospital stays have emerged. However, the ability to obtain negative surgical margins, the adequacy of lymph node dissection, and long-term outcomes have not been fully established with this approach.

    In the United States, the median age of patients who present with esophageal cancer is 67 years.

    The results of a retrospective review of 505 consecutive patients who were operated on by a single surgical team over 17 years found no difference in the perioperative mortality, median survival, or palliative benefit of esophagectomy on dysphagia when the patients older than 70 years were compared with their younger peers.

    [Levels of evidence: 3iiA and 3iiB] All of the patients in this series were selected for surgery on the basis of potential operative risk. Age alone does not determine therapy for patients with potentially resectable disease.

    Surgical treatment of esophageal cancer is associated with an operative mortality rate of less than 10%.

    In an attempt to avoid perioperative mortality and to relieve dysphagia, definitive radiation therapy in combination with chemotherapy has been studied.

    Preoperative Chemoradiation Therapy

    On the basis of several randomized trial results, chemoradiation followed by surgery is a standard treatment option for patients with stages IB, II, III, and IVA esophageal cancer.

    Phase III trials have compared preoperative concurrent chemoradiation therapy with surgery alone for patients with esophageal cancer.

    [Level of evidence: 1iiA] The benefit of neoadjuvant chemoradiation has been controversial because of contradictory results of early randomized studies.

    However, the Chemoradiotherapy for Oesophageal Cancer Followed by Surgery Study (CROSS) has definitively demonstrated a survival benefit for preoperative chemoradiation compared with surgery alone in locally advanced esophageal cancer.

    For early-stage tumors, the role of preoperative chemoradiation remains controversial. Although the CROSS study included early-stage patients, the Francophone de Cancérologie Digestive (FFCD) 9901 study (NCT00047112)

    , which included only early-stage (stage I or II) patients, failed to demonstrate a survival advantage in this group of patients.

    Evidence (preoperative chemoradiation therapy):

  • The CROSS study randomly assigned 366 patients with resectable esophageal or junctional cancers to receive either surgery alone or weekly administration of carboplatin (dose titrated to achieve an AUC [area under the curve] of 2 mg/mL/minute) and paclitaxel (50 mg/m2 of BSA [body surface area]) and concurrent radiation therapy (41.4 Gy in 23 fractions) administered over 5 weeks. Most patients enrolled in the CROSS trial (75%) had adenocarcinoma.
  • [Level of evidence: 1iiA]
  • With a median follow-up of 84 months, preoperative chemoradiation was found to improve median overall survival (OS) from 24 months in the surgery-alone group to 48.6 months (hazard ratio [HR], 0.68; 95% confidence interval [CI], 0.53–0.88; P = .003). Median OS for patients with squamous cell carcinomas was 81.6 months in the preoperative chemoradiation group compared with 21.1 months in the surgery-alone group (HR, 0.48; 95% CI, 0.28–0.83; log rank P = .008); for patients with adenocarcinomas, median OS was 43.2 months in the preoperative chemoradiation group compared with 27.1 months in the surgery-alone group (HR, 0.73; 95% CI, 0.55–0.98; log rank P = .038).
  • Additionally, preoperative chemoradiation improved the rate of R0 resections (92% vs. 69%, P < .001). R0 is defined as complete resection with no tumor within 1 mm of resection margins.
  • A complete pathologic response was achieved in 29% of patients who underwent resection after chemoradiation therapy. A pathologic complete response was observed in 23% of patients with adenocarcinoma compared with 49% of patients with squamous cell carcinoma (P = .008).
  • Postoperative complications and in-hospital mortality were equivalent in both groups. The most common hematologic side effects in the chemoradiation group were leukopenia (6%) and neutropenia (2%). The most common nonhematologic side effects were anorexia (5%) and fatigue (3%).
  • With a median follow-up of 84 months, the 5-year progression-free survival (PFS) was 44% in the preoperative chemoradiation group compared with 27% in the surgery-alone group (HR, 0.61 [0.47–0.78]). Preoperative chemoradiation therapy reduced locoregional recurrence from 34% to 14% (P < .001) and peritoneal carcinomatosis from 14% to 4% (P < .001). There was a small but significant effect on hematogenous dissemination in favor of the chemoradiation therapy group (35% vs. 29%; P = .025).
  • [Level of evidence: 1iiDii]
  • A multicenter prospective randomized trial compared preoperative combined chemotherapy (i.e., cisplatin) and radiation therapy (37 Gy in 3.7-Gy fractions) versus surgery alone in patients with squamous cell carcinoma.
  • [Level of evidence: 1iiA]
  • The study showed no improvement in OS and a significantly higher postoperative mortality (12% vs. 4%) in the combined-modality arm.
  • In patients with adenocarcinoma of the esophagus, a single-institution phase III trial was conducted in patients treated with induction chemoradiation therapy consisting of 5-fluorouracil (5-FU), cisplatin, and 40 Gy (in 2.67-Gy fractions) plus surgery compared with resection alone.
  • [Level of evidence: 1iiA]
  • The results demonstrated a modest survival benefit of 16 months for combined modality therapy versus 11 months for surgery alone.
  • A subsequent single-institution trial randomly assigned patients (75% with adenocarcinoma) to 5-FU, cisplatin, vinblastine, and radiation therapy (1.5 Gy twice daily to a total of 45 Gy) plus resection versus esophagectomy alone.
  • [Level of evidence: 1iiA]
  • At a median follow-up of more than 8 years, there was no significant difference between the surgery alone and combined modality therapy with respect to median survival (17.6 months vs. 16.9 months), OS (16% vs. 30% at 3 years), or disease-free survival (16% vs. 28% at 3 years).
  • An intergroup trial (CALGB-9781 [NCT00003118]) planned to randomly assign 475 patients with resectable squamous cell or adenocarcinoma of the thoracic esophagus to treatment with preoperative chemoradiation therapy (5-FU, cisplatin, and 50.4 Gy) followed by esophagectomy and nodal dissection or surgery alone. The trial was closed as a result of poor patient accrual; however, results from the 56 enrolled patients, with a median follow-up of 6 years, were reported.
  • [Level of evidence: 1iiA]
  • The median survival was 4.48 years (95% CI; range, 2.4 years to not estimable) for trimodality therapy versus 1.79 years (95% CI, 1.41–2.59 years) for surgery alone (P = .002), with 5-year OS of 39% for trimodality therapy (95% CI, 21%–57%) versus 16% for surgery alone (95% CI, 5%–33%).
  • To further evaluate the impact of neoadjuvant chemoradiation therapy for early-stage disease, FFCD 9901 randomly assigned 195 patients with stage I or stage II esophageal cancer to receive surgery alone or neoadjuvant chemoradiation therapy (45 Gy in 25 fractions administered with two courses of 5-FU [800 mg/m2] and cisplatin [75 mg/m2]) followed by surgery.
  • [Level of evidence: 1iiA]
  • At interim analysis, accrual to the study was stopped early because of futility.
  • With a median follow-up of 94 months, there was no significant improvement in 3-year OS with chemoradiation (48% vs. 53%, P = .94); there was a significantly higher postoperative mortality rate of 11.1% versus 3.4% (P = .049).
  • Preoperative Chemotherapy

    The effects of preoperative chemotherapy are being evaluated in randomized trials. Several studies have demonstrated a survival benefit with preoperative chemotherapy compared with surgery alone.

    However, one large randomized study failed to confirm a survival benefit with preoperative chemotherapy.

    Compared with preoperative chemotherapy alone, preoperative chemoradiation therapy improves pathologic response and may improve outcomes.

    Evidence (preoperative chemotherapy):

  • An intergroup trial (NCT00525785) randomly assigned 440 patients with local and operable esophageal cancer of any cell type to three cycles of preoperative 5-FU and cisplatin followed by surgery and two additional cycles of chemotherapy versus surgery alone.
  • [Level of evidence: 1iiA]
  • After a median follow-up of 55 months, there were no significant differences in median survival between the chemotherapy-plus-surgery group (14.9 months) and the surgery-alone group (16.1 months); median survival at 2 years was 35% for the chemotherapy-plus-surgery group and 37% for the surgery-alone group.
  • The addition of chemotherapy did not increase the morbidity associated with surgery.
  • The Medical Research Council Oesophageal Cancer Working Party randomly assigned 802 patients with resectable esophageal cancer, also of any cell type, to two cycles of preoperative 5-FU and cisplatin followed by surgery versus surgery alone.
  • [Level of evidence: 1iiA]
  • At a median follow-up of 37 months, median survival was significantly improved in the preoperative chemotherapy arm (16.8 months vs. 13.3 months with surgery alone; difference, 3.5 months; 95% CI, 1.0–6.5 months), as was 2-year OS (43% in the preoperative chemotherapy arm and 34% in the surgery-alone arm; difference, 9%; 95% CI, 3–14 months).
  • The interpretation of the results from the intergroup and preoperative chemotherapy trials is challenging because T or N staging was not reported, and prerandomization and radiation could be offered at the discretion of the treating oncologist.

  • The Japanese Clinical Oncology Group randomly assigned 330 patients with clinical stage II or III, excluding T4, squamous cell carcinomas to receive either two cycles of preoperative cisplatin and 5-FU followed by surgery or surgery followed by postoperative chemotherapy of the same regimen. A planned interim analysis was conducted after patient accrual; although the primary endpoint of PFS was not met, there was a significant benefit in OS among patients treated with preoperative chemotherapy (P = .01). As a result of these findings, the Data and Safety Monitoring Committee recommended early closure of the study.
  • [Level of evidence: 1iiC]
  • With a median follow-up of 61 months, the 5-year OS was 55% among patients treated with preoperative chemotherapy compared with 43% among patients treated with postoperative chemotherapy (P = .04). However, there was no significant difference between groups with respect to PFS (5-year PFS, 39% vs. 44%; P = .22).
  • Additionally, there were no significant differences between the two groups with respect to postoperative complications or treatment-related toxicities.
  • The Fédération Nationale des Centres de Lutte contre le Cancer and the FFCD randomly assigned 224 patients with resectable adenocarcinoma of the lower esophagus, gastroesophageal junction, or stomach to receive either perioperative chemotherapy and surgery (n = 113) or surgery alone (n = 111). Chemotherapy consisted of two or three preoperative cycles of intravenous (IV) cisplatin (100 mg/m2) on day 1 and continuous IV infusion of 5-FU (800 mg/m2) for 5 consecutive days (day 1–5) every 28 days, and three or four postoperative cycles of the same regimen.
  • [Level of evidence: 1iiA]
  • Perioperative chemotherapy was associated with improved 5-year OS (5–year OS, 38% vs. 24%; HR, 0.69; P = .02).
  • Grade 3 and 4 toxicity occurred in 38% of patients treated with perioperative chemotherapy, but there was no increase in postoperative morbidity.
  • The Preoperative Chemotherapy or Radiochemotherapy in Esophago-gastric Adenocarcinoma Trial (POET) sought to evaluate the additional benefit of radiation therapy to preoperative chemotherapy. Patients were randomly assigned to receive either induction chemotherapy (15 weeks) followed by surgery or chemotherapy (12 weeks) followed by chemoradiation therapy (3 weeks) followed by surgery.
  • [Level of evidence: 1iiA]
  • The study was closed early because of poor accrual. In total, 126 patients were randomly assigned.
  • Preoperative radiation therapy yielded 3-year survival of 27% to 47% (log rank, P = .07). Postoperative mortality was nonsignificantly increased in the chemoradiation therapy group (10.2% vs. 3.8%, P = .26).
  • Definitive Chemoradiation

    For patients who are deemed either medically inoperable or have tumors that are unresectable, the efficacy of definitive chemoradiation has been established in numerous randomized controlled trials.

    For patients with squamous cell carcinomas of the esophagus, definitive chemoradiation may offer equivalent outcomes compared with preoperative chemoradiation followed by surgical resection.

    Evidence (definitive chemoradiation):

  • A Radiation Therapy Oncology Group trial (RTOG-8501) randomly assigned patients to chemotherapy and radiation therapy versus radiation therapy alone. Patients were randomly assigned to receive radiation therapy alone (64 Gy in 32 fractions) or chemoradiation (50 Gy in 25 fractions) with concurrent cisplatin (75 mg/m2) and continuous-infusion 5-FU (1,000 mg/m2 on days 1 to 4 in weeks 1 and 5 followed by two additional cycles of chemotherapy administered 3 weeks apart).
  • [Level of evidence: 1iiA]
  • There was an improvement in 5-year survival for the combined modality group (27% vs. 0%).
  • An 8-year follow-up of this trial demonstrated an OS rate of 22% for patients receiving chemoradiation therapy.
  • Intergroup-0123 (RTOG-9405 [NCT00002631]) was conducted in an attempt to improve upon the results of RTOG-8501. Intergroup-0123 randomly assigned 236 patients with localized esophageal tumors to undergo chemoradiation with high-dose radiation therapy (64.8 Gy) and four monthly cycles of 5-FU and cisplatin versus conventional-dose radiation therapy (50.4 Gy) and the same chemotherapy schedule.
  • [Level of evidence: 1iiA]
  • Although originally designed to accrue 298 patients, this trial was closed in 1999 after a planned interim analysis showed that it was statistically unlikely that there would be any advantage to using high-dose radiation.
  • At a 2-year median follow-up, no statistically significant differences in median survival were observed between the high-dose and conventional-dose radiation therapy arms (13 months vs. 18 months), 2-year survival (31% vs. 40%), or local and regional failures (56% vs. 52%).
  • There was a higher treatment mortality in the higher-dose arm (9% vs. 2%); however, 7 of 11 deaths in the high-dose arm occurred in patients who had received 50.4 Gy or less.
  • An Eastern Cooperative Oncology Group trial (EST-1282) evaluated 135 patients.
  • [Level of evidence: 1iiA]
  • This trial showed that chemotherapy plus radiation therapy provided a better 2-year survival rate than did radiation therapy alone, similar to results from the intergroup trial.
  • The PRODIGE5/ACCORD17 (NCT00861094) trial sought to evaluate and compare the efficacy and safety of oxaliplatin, fluorouracil, and leucovorin calcium (FOLFOX) versus 5-FU and cisplatin as the chemotherapy backbone among patients treated with definitive chemoradiation for localized esophageal cancer. In this multicenter, randomized, phase II and III trial, 267 patients were randomly assigned to receive either six cycles of FOLFOX (three cycles concomitant with radiation therapy), oxaliplatin (85 mg/m2), leucovorin (200 mg/m2), bolus 5-FU (400 mg/m2) and infusion 5-FU (1,600 mg/m2 over 46 hours) or four cycles of 5-FU (1,000 mg/m2 for 4 days) and cisplatin (75 mg/m2 on day 1). All patients received radiation therapy (50 Gy in 25 fractions).
  • [Level of evidence: 1iiDiii]
  • With a median follow-up of 25.3 months, there was no significant difference in PFS (9.7 months with FOLFOX vs. 9.4 months with 5-FU and cisplatin; P = .64).
  • There was one death caused by toxicity in the FOLFOX group versus six deaths in the 5-FU and cisplatin arm (P = .066).
  • There were no significant differences in grade 3 or 4 adverse events between treatment groups. Among toxicities of all grades, paresthesia, sensory neuropathy, and increases in aspartate transaminase and alanine transaminase were more common in the FOLFOX group; whereas, increases in serum creatinine, mucositis, and alopecia were more common in the 5-FU and cisplatin group.
  • A phase III German trial also compared induction chemotherapy (three courses of bolus 5-FU, leucovorin, etoposide, and cisplatin) followed by chemoradiation therapy (cisplatin, etoposide, and 40 Gy) followed by surgery (arm A), or the same induction chemotherapy followed by chemoradiation therapy (at least 65 Gy) without surgery (arm B) for patients with T3 or T4 squamous cell carcinoma of the esophagus. OS was the primary outcome.
  • [Level of evidence: 1iiA]
  • The analysis of 172 eligible, randomly assigned patients showed that OS at 2 years was not statistically significantly different between the two treatment groups (arm A, 39.9%; 95% CI, 29.4%–50.4%; arm B, 35.4%; 95% CI, 25.2%–45.6%; log-rank test for equivalence with 0.15, P < .007).
  • Local PFS was higher in the surgery group (2-year PFS, 64.3%; 95% CI, 52.1%–76.5%) than in the chemoradiation therapy group (2-year PFS, 40.7%; 95% CI, 28.9%–52.5%; HR for arm B vs. arm A, 2.1; 95% CI, 1.3–3.5; P < .003).
  • Treatment-related mortality was higher in the surgery group (12.8%) than in the chemoradiation therapy group (3.5%) (P < .03).
  • FFCD 9102 (NCT00416858) randomly assigned 259 patients with T3N0–1M0 thoracic esophageal cancer to receive either two cycles of 5-FU and cisplatin (days 1–5 and 22–26) and either conventional radiation therapy (46 Gy in 4.5 weeks) or split course (15 Gy, days 1–5 and 22–26). Patients with response were then randomly assigned to receive either surgical resection (arm A) or continuation of chemoradiation (arm B: three cycles of 5-FU plus cisplatin and either conventional 20 Gy or split-course 15 Gy radiation therapy).
  • [Level of evidence: 1iiA]
  • Of the 259 randomly assigned patients, 230 (89%) had squamous cell carcinoma, and 29 patients (11%) had adenocarcinomas.
  • The 2-year OS was 34% in patients randomly assigned to receive surgery versus 40% in patients randomly assigned to receive definitive chemoradiation (HR, 0.90; P = .44). Median survival was 17.7 months for surgery and 19.3 months for definitive chemoradiation.
  • The 3-month mortality rate was 9.3% in the surgery arm compared with 0.8% in the chemoradiation arm (P = .002).
  • Postoperative Radiation Therapy

    Two randomized trials have shown no significant OS benefit for postoperative radiation therapy compared with surgery alone.

    [Level of evidence: 1iiA] All newly diagnosed patients should be considered candidates for therapies and clinical trials comparing various treatment modalities. Information about ongoing clinical trials is available from the NCI website.

    ReferenceSection

  • Tietjen TG, Pasricha PJ, Kalloo AN: Management of malignant esophageal stricture with esophageal dilation and esophageal stents. Gastrointest Endosc Clin N Am 4 (4): 851-62, 1994.
  • Lightdale CJ, Heier SK, Marcon NE, et al.: Photodynamic therapy with porfimer sodium versus thermal ablation therapy with Nd:YAG laser for palliation of esophageal cancer: a multicenter randomized trial. Gastrointest Endosc 42 (6): 507-12, 1995.
  • Kubba AK: Role of photodynamic therapy in the management of gastrointestinal cancer. Digestion 60 (1): 1-10, 1999 Jan-Feb.
  • Heier SK, Heier LM: Tissue sensitizers. Gastrointest Endosc Clin N Am 4 (2): 327-52, 1994.
  • Bourke MJ, Hope RL, Chu G, et al.: Laser palliation of inoperable malignant dysphagia: initial and at death. Gastrointest Endosc 43 (1): 29-32, 1996.
  • Lerut T, Coosemans W, Van Raemdonck D, et al.: Surgical treatment of Barrett's carcinoma. Correlations between morphologic findings and prognosis. J Thorac Cardiovasc Surg 107 (4): 1059-65; discussion 1065-6, 1994.
  • Kelsen DP, Bains M, Burt M: Neoadjuvant chemotherapy and surgery of cancer of the esophagus. Semin Surg Oncol 6 (5): 268-73, 1990.
  • Saxon RR, Morrison KE, Lakin PC, et al.: Malignant esophageal obstruction and esophagorespiratory fistula: palliation with a polyethylene-covered Z-stent. Radiology 202 (2): 349-54, 1997.
  • Campbell WR, Taylor SA, Pierce GE, et al.: Therapeutic alternatives in patients with esophageal cancer. Am J Surg 150 (6): 665-8, 1985.
  • Mellow MH, Pinkas H: Endoscopic therapy for esophageal carcinoma with Nd:YAG laser: prospective evaluation of efficacy, complications, and survival. Gastrointest Endosc 30 (6): 334-9, 1984.
  • Karlin DA, Fisher RS, Krevsky B: Prolonged survival and effective palliation in patients with squamous cell carcinoma of the esophagus following endoscopic laser therapy. Cancer 59 (11): 1969-72, 1987.
  • Hulscher JB, van Sandick JW, de Boer AG, et al.: Extended transthoracic resection compared with limited transhiatal resection for adenocarcinoma of the esophagus. N Engl J Med 347 (21): 1662-9, 2002.
  • de Boer AG, van Lanschot JJ, van Sandick JW, et al.: Quality of life after transhiatal compared with extended transthoracic resection for adenocarcinoma of the esophagus. J Clin Oncol 22 (20): 4202-8, 2004.
  • Santillan AA, Farma JM, Meredith KL, et al.: Minimally invasive surgery for esophageal cancer. J Natl Compr Canc Netw 6 (9): 879-84, 2008.
  • Ginsberg RJ: Cancer treatment in the elderly. J Am Coll Surg 187 (4): 427-8, 1998.
  • Ellis FH, Williamson WA, Heatley GJ: Cancer of the esophagus and cardia: does age influence treatment selection and surgical outcomes? J Am Coll Surg 187 (4): 345-51, 1998.
  • Bosset JF, Gignoux M, Triboulet JP, et al.: Chemoradiotherapy followed by surgery compared with surgery alone in squamous-cell cancer of the esophagus. N Engl J Med 337 (3): 161-7, 1997.
  • Walsh TN, Noonan N, Hollywood D, et al.: A comparison of multimodal therapy and surgery for esophageal adenocarcinoma. N Engl J Med 335 (7): 462-7, 1996.
  • Urba SG, Orringer MB, Turrisi A, et al.: Randomized trial of preoperative chemoradiation versus surgery alone in patients with locoregional esophageal carcinoma. J Clin Oncol 19 (2): 305-13, 2001.
  • Tepper J, Krasna MJ, Niedzwiecki D, et al.: Phase III trial of trimodality therapy with cisplatin, fluorouracil, radiotherapy, and surgery compared with surgery alone for esophageal cancer: CALGB 9781. J Clin Oncol 26 (7): 1086-92, 2008.
  • van Hagen P, Hulshof MC, van Lanschot JJ, et al.: Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 366 (22): 2074-84, 2012.
  • Oppedijk V, van der Gaast A, van Lanschot JJ, et al.: Patterns of recurrence after surgery alone versus preoperative chemoradiotherapy and surgery in the CROSS trials. J Clin Oncol 32 (5): 385-91, 2014.
  • Mariette C, Dahan L, Mornex F, et al.: Surgery alone versus chemoradiotherapy followed by surgery for stage I and II esophageal cancer: final analysis of randomized controlled phase III trial FFCD 9901. J Clin Oncol 32 (23): 2416-22, 2014.
  • Shapiro J, van Lanschot JJ, Hulshof MC, et al.: Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or junctional cancer (CROSS): long-term results of a randomised controlled trial. Lancet Oncol 16 (9): 1090-8, 2015.
  • Medical Research Council Oesophageal Cancer Working Group: Surgical resection with or without preoperative chemotherapy in oesophageal cancer: a randomised controlled trial. Lancet 359 (9319): 1727-33, 2002.
  • Ando N, Kato H, Igaki H, et al.: A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus (JCOG9907). Ann Surg Oncol 19 (1): 68-74, 2012.
  • Ychou M, Boige V, Pignon JP, et al.: Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol 29 (13): 1715-21, 2011.
  • Kelsen DP, Ginsberg R, Pajak TF, et al.: Chemotherapy followed by surgery compared with surgery alone for localized esophageal cancer. N Engl J Med 339 (27): 1979-84, 1998.
  • Stahl M, Walz MK, Stuschke M, et al.: Phase III comparison of preoperative chemotherapy compared with chemoradiotherapy in patients with locally advanced adenocarcinoma of the esophagogastric junction. J Clin Oncol 27 (6): 851-6, 2009.
  • Cooper JS, Guo MD, Herskovic A, et al.: Chemoradiotherapy of locally advanced esophageal cancer: long-term follow-up of a prospective randomized trial (RTOG 85-01). Radiation Therapy Oncology Group. JAMA 281 (17): 1623-7, 1999.
  • Minsky BD, Pajak TF, Ginsberg RJ, et al.: INT 0123 (Radiation Therapy Oncology Group 94-05) phase III trial of combined-modality therapy for esophageal cancer: high-dose versus standard-dose radiation therapy. J Clin Oncol 20 (5): 1167-74, 2002.
  • Stahl M, Stuschke M, Lehmann N, et al.: Chemoradiation with and without surgery in patients with locally advanced squamous cell carcinoma of the esophagus. J Clin Oncol 23 (10): 2310-7, 2005.
  • Bedenne L, Michel P, Bouché O, et al.: Chemoradiation followed by surgery compared with chemoradiation alone in squamous cancer of the esophagus: FFCD 9102. J Clin Oncol 25 (10): 1160-8, 2007.
  • Smith TJ, Ryan LM, Douglass HO, et al.: Combined chemoradiotherapy vs. radiotherapy alone for early stage squamous cell carcinoma of the esophagus: a study of the Eastern Cooperative Oncology Group. Int J Radiat Oncol Biol Phys 42 (2): 269-76, 1998.
  • Conroy T, Galais MP, Raoul JL, et al.: Definitive chemoradiotherapy with FOLFOX versus fluorouracil and cisplatin in patients with oesophageal cancer (PRODIGE5/ACCORD17): final results of a randomised, phase 2/3 trial. Lancet Oncol 15 (3): 305-14, 2014.
  • Ténière P, Hay JM, Fingerhut A, et al.: Postoperative radiation therapy does not increase survival after curative resection for squamous cell carcinoma of the middle and lower esophagus as shown by a multicenter controlled trial. French University Association for Surgical Research. Surg Gynecol Obstet 173 (2): 123-30, 1991.
  • Fok M, Sham JS, Choy D, et al.: Postoperative radiotherapy for carcinoma of the esophagus: a prospective, randomized controlled study. Surgery 113 (2): 138-47, 1993.
  • 食管癌的临床治疗(成人)(PDQ®)

    0期食管癌的治疗

    0期食管癌的标准治疗方法

    在美国,0期食管鳞癌较为罕见,但曾有手术病例。

    早期微小浸润食管癌,手术和内镜切除的治愈率较高。

  • 手术
  • 内镜下切除
  • 当前的临床试验

    采用我们先进的临床试验搜索引擎查找正在进行患者招募的NCI支持的癌症临床试验。搜索引擎可按试验所在地、治疗类型、药物名称和其他标准进行设置。还能查询有关临床试验的基本信息

    参考文献

  • Rusch VW, Levine DS, Haggitt R, et al.: The management of high grade dysplasia and early cancer in Barrett's esophagus. A multidisciplinary problem. Cancer 74 (4): 1225-9, 1994.
  • Heitmiller RF, Redmond M, Hamilton SR: Barrett's esophagus with high-grade dysplasia. An indication for prophylactic esophagectomy. Ann Surg 224 (1): 66-71, 1996.
  • Pech O, Bollschweiler E, Manner H, et al.: Comparison between endoscopic and surgical resection of mucosal esophageal adenocarcinoma in Barrett's esophagus at two high-volume centers. Ann Surg 254 (1): 67-72, 2011.
  • Prasad GA, Wu TT, Wigle DA, et al.: Endoscopic and surgical treatment of mucosal (T1a) esophageal adenocarcinoma in Barrett's esophagus. Gastroenterology 137 (3): 815-23, 2009.
  • Esophageal Cancer Treatment (Adult) (PDQ®)

    Stage 0 Esophageal Cancer Treatment

    Standard Treatment Options for Stage 0 Esophageal Cancer

    Stage 0 squamous cell esophageal cancer is rarely seen in the United States, but surgery has been used.

    For early-stage minimally invasive esophageal cancer, surgical and endoscopic techniques offer high rates of cure.

  • Surgery.
  • Endoscopic resection.
  • Current Clinical Trials

    Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.

    ReferenceSection

  • Rusch VW, Levine DS, Haggitt R, et al.: The management of high grade dysplasia and early cancer in Barrett's esophagus. A multidisciplinary problem. Cancer 74 (4): 1225-9, 1994.
  • Heitmiller RF, Redmond M, Hamilton SR: Barrett's esophagus with high-grade dysplasia. An indication for prophylactic esophagectomy. Ann Surg 224 (1): 66-71, 1996.
  • Pech O, Bollschweiler E, Manner H, et al.: Comparison between endoscopic and surgical resection of mucosal esophageal adenocarcinoma in Barrett's esophagus at two high-volume centers. Ann Surg 254 (1): 67-72, 2011.
  • Prasad GA, Wu TT, Wigle DA, et al.: Endoscopic and surgical treatment of mucosal (T1a) esophageal adenocarcinoma in Barrett's esophagus. Gastroenterology 137 (3): 815-23, 2009.
  • 食管癌的临床治疗(成人)(PDQ®)

    临床I期食管癌的治疗

    I期食管癌的标准治疗方法

    I期食管癌的标准治疗方法,包括:

  • 术前放化疗联合手术。
  • 单独手术治疗。
  • 当前的临床试验

    采用我们先进的临床试验搜索引擎查找正在进行患者招募的NCI支持的癌症临床试验。搜索引擎可按试验所在地、治疗类型、药物名称和其他标准进行设置。还能查询有关临床试验的基本信息

    参考文献

  • Bosset JF, Gignoux M, Triboulet JP, et al.: Chemoradiotherapy followed by surgery compared with surgery alone in squamous-cell cancer of the esophagus. N Engl J Med 337 (3): 161-7, 1997.
  • Walsh TN, Noonan N, Hollywood D, et al.: A comparison of multimodal therapy and surgery for esophageal adenocarcinoma. N Engl J Med 335 (7): 462-7, 1996.
  • Urba SG, Orringer MB, Turrisi A, et al.: Randomized trial of preoperative chemoradiation versus surgery alone in patients with locoregional esophageal carcinoma. J Clin Oncol 19 (2): 305-13, 2001.
  • Tepper J, Krasna MJ, Niedzwiecki D, et al.: Phase III trial of trimodality therapy with cisplatin, fluorouracil, radiotherapy, and surgery compared with surgery alone for esophageal cancer: CALGB 9781. J Clin Oncol 26 (7): 1086-92, 2008.
  • van Hagen P, Hulshof MC, van Lanschot JJ, et al.: Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 366 (22): 2074-84, 2012.
  • Esophageal Cancer Treatment (Adult) (PDQ®)

    Stage I Esophageal Cancer Treatment

    Standard Treatment Options for Stage I Esophageal Cancer

    Standard treatment options for stage I esophageal cancer include the following:

  • Chemoradiation followed by surgery.
  • Surgery alone.
  • Current Clinical Trials

    Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.

    ReferenceSection

  • Bosset JF, Gignoux M, Triboulet JP, et al.: Chemoradiotherapy followed by surgery compared with surgery alone in squamous-cell cancer of the esophagus. N Engl J Med 337 (3): 161-7, 1997.
  • Walsh TN, Noonan N, Hollywood D, et al.: A comparison of multimodal therapy and surgery for esophageal adenocarcinoma. N Engl J Med 335 (7): 462-7, 1996.
  • Urba SG, Orringer MB, Turrisi A, et al.: Randomized trial of preoperative chemoradiation versus surgery alone in patients with locoregional esophageal carcinoma. J Clin Oncol 19 (2): 305-13, 2001.
  • Tepper J, Krasna MJ, Niedzwiecki D, et al.: Phase III trial of trimodality therapy with cisplatin, fluorouracil, radiotherapy, and surgery compared with surgery alone for esophageal cancer: CALGB 9781. J Clin Oncol 26 (7): 1086-92, 2008.
  • van Hagen P, Hulshof MC, van Lanschot JJ, et al.: Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 366 (22): 2074-84, 2012.
  • 食管癌的临床治疗(成人)(PDQ®)

    II期食管癌的治疗

    II期食管癌的标准治疗方法

    II期食管癌的标准治疗方法,包括:

  • 术前放化疗联合手术。
  • 单独手术治疗。
  • 术前化疗联合手术。
  • 根治性放化疗。
  • 当前的临床试验

    采用我们先进的临床试验搜索引擎查找正在进行患者招募的NCI支持的癌症临床试验。搜索引擎可按试验所在地、治疗类型、药物名称和其他标准进行设置。还能查询有关临床试验的基本信息

    参考文献

  • Walsh TN, Noonan N, Hollywood D, et al.: A comparison of multimodal therapy and surgery for esophageal adenocarcinoma. N Engl J Med 335 (7): 462-7, 1996.
  • Tepper J, Krasna MJ, Niedzwiecki D, et al.: Phase III trial of trimodality therapy with cisplatin, fluorouracil, radiotherapy, and surgery compared with surgery alone for esophageal cancer: CALGB 9781. J Clin Oncol 26 (7): 1086-92, 2008.
  • van Hagen P, Hulshof MC, van Lanschot JJ, et al.: Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 366 (22): 2074-84, 2012.
  • Urba SG, Orringer MB, Turrisi A, et al.: Randomized trial of preoperative chemoradiation versus surgery alone in patients with locoregional esophageal carcinoma. J Clin Oncol 19 (2): 305-13, 2001.
  • Bosset JF, Gignoux M, Triboulet JP, et al.: Chemoradiotherapy followed by surgery compared with surgery alone in squamous-cell cancer of the esophagus. N Engl J Med 337 (3): 161-7, 1997.
  • Conroy T, Galais MP, Raoul JL, et al.: Definitive chemoradiotherapy with FOLFOX versus fluorouracil and cisplatin in patients with oesophageal cancer (PRODIGE5/ACCORD17): final results of a randomised, phase 2/3 trial. Lancet Oncol 15 (3): 305-14, 2014.
  • Mariette C, Dahan L, Mornex F, et al.: Surgery alone versus chemoradiotherapy followed by surgery for stage I and II esophageal cancer: final analysis of randomized controlled phase III trial FFCD 9901. J Clin Oncol 32 (23): 2416-22, 2014.
  • Medical Research Council Oesophageal Cancer Working Group: Surgical resection with or without preoperative chemotherapy in oesophageal cancer: a randomised controlled trial. Lancet 359 (9319): 1727-33, 2002.
  • Ando N, Kato H, Igaki H, et al.: A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus (JCOG9907). Ann Surg Oncol 19 (1): 68-74, 2012.
  • Ychou M, Boige V, Pignon JP, et al.: Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol 29 (13): 1715-21, 2011.
  • Stahl M, Stuschke M, Lehmann N, et al.: Chemoradiation with and without surgery in patients with locally advanced squamous cell carcinoma of the esophagus. J Clin Oncol 23 (10): 2310-7, 2005.
  • Bedenne L, Michel P, Bouché O, et al.: Chemoradiation followed by surgery compared with chemoradiation alone in squamous cancer of the esophagus: FFCD 9102. J Clin Oncol 25 (10): 1160-8, 2007.
  • Esophageal Cancer Treatment (Adult) (PDQ®)

    Stage II Esophageal Cancer Treatment

    Standard Treatment Options for Stage II Esophageal Cancer

    Standard treatment options for stage II esophageal cancer include the following:

  • Chemoradiation followed by surgery.
  • Surgery alone.
  • Chemotherapy followed by surgery.
  • Definitive chemoradiation.
  • Current Clinical Trials

    Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.

    ReferenceSection

  • Walsh TN, Noonan N, Hollywood D, et al.: A comparison of multimodal therapy and surgery for esophageal adenocarcinoma. N Engl J Med 335 (7): 462-7, 1996.
  • Tepper J, Krasna MJ, Niedzwiecki D, et al.: Phase III trial of trimodality therapy with cisplatin, fluorouracil, radiotherapy, and surgery compared with surgery alone for esophageal cancer: CALGB 9781. J Clin Oncol 26 (7): 1086-92, 2008.
  • van Hagen P, Hulshof MC, van Lanschot JJ, et al.: Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 366 (22): 2074-84, 2012.
  • Urba SG, Orringer MB, Turrisi A, et al.: Randomized trial of preoperative chemoradiation versus surgery alone in patients with locoregional esophageal carcinoma. J Clin Oncol 19 (2): 305-13, 2001.
  • Bosset JF, Gignoux M, Triboulet JP, et al.: Chemoradiotherapy followed by surgery compared with surgery alone in squamous-cell cancer of the esophagus. N Engl J Med 337 (3): 161-7, 1997.
  • Conroy T, Galais MP, Raoul JL, et al.: Definitive chemoradiotherapy with FOLFOX versus fluorouracil and cisplatin in patients with oesophageal cancer (PRODIGE5/ACCORD17): final results of a randomised, phase 2/3 trial. Lancet Oncol 15 (3): 305-14, 2014.
  • Mariette C, Dahan L, Mornex F, et al.: Surgery alone versus chemoradiotherapy followed by surgery for stage I and II esophageal cancer: final analysis of randomized controlled phase III trial FFCD 9901. J Clin Oncol 32 (23): 2416-22, 2014.
  • Medical Research Council Oesophageal Cancer Working Group: Surgical resection with or without preoperative chemotherapy in oesophageal cancer: a randomised controlled trial. Lancet 359 (9319): 1727-33, 2002.
  • Ando N, Kato H, Igaki H, et al.: A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus (JCOG9907). Ann Surg Oncol 19 (1): 68-74, 2012.
  • Ychou M, Boige V, Pignon JP, et al.: Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol 29 (13): 1715-21, 2011.
  • Stahl M, Stuschke M, Lehmann N, et al.: Chemoradiation with and without surgery in patients with locally advanced squamous cell carcinoma of the esophagus. J Clin Oncol 23 (10): 2310-7, 2005.
  • Bedenne L, Michel P, Bouché O, et al.: Chemoradiation followed by surgery compared with chemoradiation alone in squamous cancer of the esophagus: FFCD 9102. J Clin Oncol 25 (10): 1160-8, 2007.
  • 食管癌的临床治疗(成人)(PDQ®)

    III期食管癌的治疗

    III期食管癌的标准治疗方法

    III 期食管癌的标准治疗方法,包括:

  • 术前放化疗联合手术。
  • 术前化疗联合手术。
  • 根治性放化疗。
  • 当前的临床试验

    采用我们先进的临床试验搜索引擎查找正在进行患者招募的NCI支持的癌症临床试验。搜索引擎可按试验所在地、治疗类型、药物名称和其他标准进行设置。还能查询有关临床试验的基本信息

    参考文献

  • Walsh TN, Noonan N, Hollywood D, et al.: A comparison of multimodal therapy and surgery for esophageal adenocarcinoma. N Engl J Med 335 (7): 462-7, 1996.
  • Tepper J, Krasna MJ, Niedzwiecki D, et al.: Phase III trial of trimodality therapy with cisplatin, fluorouracil, radiotherapy, and surgery compared with surgery alone for esophageal cancer: CALGB 9781. J Clin Oncol 26 (7): 1086-92, 2008.
  • van Hagen P, Hulshof MC, van Lanschot JJ, et al.: Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 366 (22): 2074-84, 2012.
  • Medical Research Council Oesophageal Cancer Working Group: Surgical resection with or without preoperative chemotherapy in oesophageal cancer: a randomised controlled trial. Lancet 359 (9319): 1727-33, 2002.
  • Ando N, Kato H, Igaki H, et al.: A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus (JCOG9907). Ann Surg Oncol 19 (1): 68-74, 2012.
  • Ychou M, Boige V, Pignon JP, et al.: Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol 29 (13): 1715-21, 2011.
  • Conroy T, Galais MP, Raoul JL, et al.: Definitive chemoradiotherapy with FOLFOX versus fluorouracil and cisplatin in patients with oesophageal cancer (PRODIGE5/ACCORD17): final results of a randomised, phase 2/3 trial. Lancet Oncol 15 (3): 305-14, 2014.
  • Stahl M, Stuschke M, Lehmann N, et al.: Chemoradiation with and without surgery in patients with locally advanced squamous cell carcinoma of the esophagus. J Clin Oncol 23 (10): 2310-7, 2005.
  • Bedenne L, Michel P, Bouché O, et al.: Chemoradiation followed by surgery compared with chemoradiation alone in squamous cancer of the esophagus: FFCD 9102. J Clin Oncol 25 (10): 1160-8, 2007.
  • Esophageal Cancer Treatment (Adult) (PDQ®)

    Stage III Esophageal Cancer Treatment

    Standard Treatment Options for Stage III Esophageal Cancer

    Standard treatment options for stage III esophageal cancer include the following:

  • Chemoradiation followed by surgery.
  • Preoperative chemotherapy followed by surgery.
  • Definitive chemoradiation.
  • Current Clinical Trials

    Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.

    ReferenceSection

  • Walsh TN, Noonan N, Hollywood D, et al.: A comparison of multimodal therapy and surgery for esophageal adenocarcinoma. N Engl J Med 335 (7): 462-7, 1996.
  • Tepper J, Krasna MJ, Niedzwiecki D, et al.: Phase III trial of trimodality therapy with cisplatin, fluorouracil, radiotherapy, and surgery compared with surgery alone for esophageal cancer: CALGB 9781. J Clin Oncol 26 (7): 1086-92, 2008.
  • van Hagen P, Hulshof MC, van Lanschot JJ, et al.: Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med 366 (22): 2074-84, 2012.
  • Medical Research Council Oesophageal Cancer Working Group: Surgical resection with or without preoperative chemotherapy in oesophageal cancer: a randomised controlled trial. Lancet 359 (9319): 1727-33, 2002.
  • Ando N, Kato H, Igaki H, et al.: A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus (JCOG9907). Ann Surg Oncol 19 (1): 68-74, 2012.
  • Ychou M, Boige V, Pignon JP, et al.: Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol 29 (13): 1715-21, 2011.
  • Conroy T, Galais MP, Raoul JL, et al.: Definitive chemoradiotherapy with FOLFOX versus fluorouracil and cisplatin in patients with oesophageal cancer (PRODIGE5/ACCORD17): final results of a randomised, phase 2/3 trial. Lancet Oncol 15 (3): 305-14, 2014.
  • Stahl M, Stuschke M, Lehmann N, et al.: Chemoradiation with and without surgery in patients with locally advanced squamous cell carcinoma of the esophagus. J Clin Oncol 23 (10): 2310-7, 2005.
  • Bedenne L, Michel P, Bouché O, et al.: Chemoradiation followed by surgery compared with chemoradiation alone in squamous cancer of the esophagus: FFCD 9102. J Clin Oncol 25 (10): 1160-8, 2007.
  • 食管癌的临床治疗(成人)(PDQ®)

    IV期食管癌的治疗

    IV期食管癌的治疗方法

    约50%食管癌患者在确诊时伴随转移癌,建议采用姑息治疗。

    IV期食管癌的治疗方法包括:

  • 术前放化疗联合手术(针对临床IVA期患者)。
  • 转移性食管下段腺癌患者采用化疗,可取得部分缓解。
  • Nd:YAG激光治疗的腔内肿瘤破坏或电凝治疗。
  • 内镜下置入支架可缓解吞咽困难。
  • 放疗联合/不联合腔内置管和扩张术。
  • 腔内近距离放射治疗可缓解吞咽困难
  • 食管癌可采用多种抗癌药治疗。多篇文献报道,以铂类为基础联合5-FU、紫杉类、拓扑异构酶抑制剂、羟基脲、长春瑞滨的治疗,其客观缓解率约为30%-60%,中位生存期不足1年。

    曲妥珠单抗联合化疗可用于过度表达HER2-neu的食管癌患者。

    [证据级别:1iiA]

  • 目前有多项临床试验对单药和联合化疗进行研究。
  • 当前的临床试验

    采用我们先进的临床试验搜索引擎查找正在进行患者招募的NCI支持的癌症临床试验。搜索引擎可按试验所在地、治疗类型、药物名称和其他标准进行设置。还能查询有关临床试验的基本信息

    参考文献

  • Enzinger PC, Ilson DH, Kelsen DP: Chemotherapy in esophageal cancer. Semin Oncol 26 (5 Suppl 15): 12-20, 1999.
  • Waters JS, Norman A, Cunningham D, et al.: Long-term survival after epirubicin, cisplatin and fluorouracil for gastric cancer: results of a randomized trial. Br J Cancer 80 (1-2): 269-72, 1999.
  • Ross P, Nicolson M, Cunningham D, et al.: Prospective randomized trial comparing mitomycin, cisplatin, and protracted venous-infusion fluorouracil (PVI 5-FU) With epirubicin, cisplatin, and PVI 5-FU in advanced esophagogastric cancer. J Clin Oncol 20 (8): 1996-2004, 2002.
  • Taïeb J, Artru P, Baujat B, et al.: Optimisation of 5-fluorouracil (5-FU)/cisplatin combination chemotherapy with a new schedule of hydroxyurea, leucovorin, 5-FU and cisplatin (HLFP regimen) for metastatic oesophageal cancer. Eur J Cancer 38 (5): 661-6, 2002.
  • Bourke MJ, Hope RL, Chu G, et al.: Laser palliation of inoperable malignant dysphagia: initial and at death. Gastrointest Endosc 43 (1): 29-32, 1996.
  • Baron TH: Expandable metal stents for the treatment of cancerous obstruction of the gastrointestinal tract. N Engl J Med 344 (22): 1681-7, 2001.
  • Sur RK, Levin CV, Donde B, et al.: Prospective randomized trial of HDR brachytherapy as a sole modality in palliation of advanced esophageal carcinoma--an International Atomic Energy Agency study. Int J Radiat Oncol Biol Phys 53 (1): 127-33, 2002.
  • Gaspar LE, Nag S, Herskovic A, et al.: American Brachytherapy Society (ABS) consensus guidelines for brachytherapy of esophageal cancer. Clinical Research Committee, American Brachytherapy Society, Philadelphia, PA. Int J Radiat Oncol Biol Phys 38 (1): 127-32, 1997.
  • Conroy T, Etienne PL, Adenis A, et al.: Vinorelbine and cisplatin in metastatic squamous cell carcinoma of the oesophagus: response, toxicity, quality of life and survival. Ann Oncol 13 (5): 721-9, 2002.
  • Bang YJ, Van Cutsem E, Feyereislova A, et al.: Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376 (9742): 687-97, 2010.
  • Esophageal Cancer Treatment (Adult) (PDQ®)

    Stage IV Esophageal Cancer Treatment

    Treatment Options for Stage IV Esophageal Cancer

    At diagnosis, approximately 50% of patients with esophageal cancer will have metastatic disease and will be candidates for palliative therapy.

    Treatment options for stage IV esophageal cancer include the following:

  • Chemoradiation followed by surgery (for patients with stage IVA disease).
  • Chemotherapy, which has provided partial responses for patients with metastatic distal esophageal adenocarcinomas.
  • Nd:YAG endoluminal tumor destruction or electrocoagulation.
  • Endoscopic-placed stents to provide palliation of dysphagia.
  • Radiation therapy with or without intraluminal intubation and dilation.
  • Intraluminal brachytherapy to provide palliation of dysphagia.
  • Esophageal cancer responds to many anticancer agents. Objective response rates of 30% to 60% and median survivals of less than 1 year are commonly reported with platinum-based combination regimens with 5-fluorouracil, taxanes, topoisomerase inhibitors, hydroxyurea, or vinorelbine.

    Trastuzumab may be effective in combination with chemotherapy among patients with tumors that overexpress HER2-neu.

    [Level of evidence: 1iiA]

  • Clinical trials evaluating single-agent or combination chemotherapy.
  • Current Clinical Trials

    Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.

    ReferenceSection

  • Enzinger PC, Ilson DH, Kelsen DP: Chemotherapy in esophageal cancer. Semin Oncol 26 (5 Suppl 15): 12-20, 1999.
  • Waters JS, Norman A, Cunningham D, et al.: Long-term survival after epirubicin, cisplatin and fluorouracil for gastric cancer: results of a randomized trial. Br J Cancer 80 (1-2): 269-72, 1999.
  • Ross P, Nicolson M, Cunningham D, et al.: Prospective randomized trial comparing mitomycin, cisplatin, and protracted venous-infusion fluorouracil (PVI 5-FU) With epirubicin, cisplatin, and PVI 5-FU in advanced esophagogastric cancer. J Clin Oncol 20 (8): 1996-2004, 2002.
  • Taïeb J, Artru P, Baujat B, et al.: Optimisation of 5-fluorouracil (5-FU)/cisplatin combination chemotherapy with a new schedule of hydroxyurea, leucovorin, 5-FU and cisplatin (HLFP regimen) for metastatic oesophageal cancer. Eur J Cancer 38 (5): 661-6, 2002.
  • Bourke MJ, Hope RL, Chu G, et al.: Laser palliation of inoperable malignant dysphagia: initial and at death. Gastrointest Endosc 43 (1): 29-32, 1996.
  • Baron TH: Expandable metal stents for the treatment of cancerous obstruction of the gastrointestinal tract. N Engl J Med 344 (22): 1681-7, 2001.
  • Sur RK, Levin CV, Donde B, et al.: Prospective randomized trial of HDR brachytherapy as a sole modality in palliation of advanced esophageal carcinoma--an International Atomic Energy Agency study. Int J Radiat Oncol Biol Phys 53 (1): 127-33, 2002.
  • Gaspar LE, Nag S, Herskovic A, et al.: American Brachytherapy Society (ABS) consensus guidelines for brachytherapy of esophageal cancer. Clinical Research Committee, American Brachytherapy Society, Philadelphia, PA. Int J Radiat Oncol Biol Phys 38 (1): 127-32, 1997.
  • Conroy T, Etienne PL, Adenis A, et al.: Vinorelbine and cisplatin in metastatic squamous cell carcinoma of the oesophagus: response, toxicity, quality of life and survival. Ann Oncol 13 (5): 721-9, 2002.
  • Bang YJ, Van Cutsem E, Feyereislova A, et al.: Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376 (9742): 687-97, 2010.
  • 食管癌的临床治疗(成人)(PDQ®)

    复发性食管癌的治疗

    对复发性食管癌患者来讲,姑息治疗通常意味着困难重重。所有患者均应考虑参加临床试验,这一点已在本篇食管癌治疗方法的概述中有所论述。

  • 标准治疗的缓和应用,包括支持治疗。
  • 当前的临床试验

    采用我们先进的临床试验搜索引擎查找正在进行患者招募的NCI支持的癌症临床试验。搜索引擎可按试验所在地、治疗类型、药物名称和其他标准进行设置。还能查询有关临床试验的基本信息

    Esophageal Cancer Treatment (Adult) (PDQ®)

    Recurrent Esophageal Cancer Treatment

    Palliation presents difficult problems for all patients with recurrent esophageal cancer. All patients should be considered candidates for clinical trials as outlined in the Treatment Option Overview for Esophageal Cancer section of this summary.

  • Palliative use of any of the standard therapies, including supportive care.
  • Current Clinical Trials

    Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.

    食管癌的临床治疗(成人)(PDQ®)

    最新更新日期:2020.01.22

    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)。

    2020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。

    Esophageal Cancer Treatment (Adult) (PDQ®)

    Changes to This Summary (01/22/2020)

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

    Updated statistics with estimated new cases and deaths for 2020 (cited American Cancer Society as reference 1).

    This 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.

    食管癌的临床治疗(成人)(PDQ®)

    About This PDQ Summary

    Purpose of This Summary

    This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about treatment of adult esophageal 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.

    Reviewers and Updates

    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:

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

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

    Levels of Evidence

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

    Permission to Use This Summary

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

    The preferred citation for this PDQ summary is:

    PDQ® Adult Treatment Editorial Board. PDQ Esophageal Cancer Treatment (Adult). Bethesda, MD: National Cancer Institute. Updated . Available at: https://www.cancer.gov/types/esophageal/hp/esophageal-treatment-pdq. Accessed . [PMID: 26389338]

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

    Disclaimer

    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.

    Contact Us

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

    Esophageal Cancer Treatment (Adult) (PDQ®)

    About This PDQ Summary

    Purpose of This Summary

    This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about treatment of adult esophageal 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.

    Reviewers and Updates

    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:

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

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

    Levels of Evidence

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

    Permission to Use This Summary

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

    The preferred citation for this PDQ summary is:

    PDQ® Adult Treatment Editorial Board. PDQ Esophageal Cancer Treatment (Adult). Bethesda, MD: National Cancer Institute. Updated . Available at: https://www.cancer.gov/types/esophageal/hp/esophageal-treatment-pdq. Accessed . [PMID: 26389338]

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

    Disclaimer

    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.

    Contact Us

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

    < 1 2 3 4 5 6 7 8 9 10 11 12 >
    目录
    章 节
    有关食管癌的基本信息 食管癌的细胞学分类 食管癌的分期信息 食管癌治疗方法概述 0期食管癌的治疗 临床I期食管癌的治疗 II期食管癌的治疗 III期食管癌的治疗 IV期食管癌的治疗 复发性食管癌的治疗 最新更新日期:2020.01.22 About This PDQ Summary