NSCLC是除小细胞肺癌(SCLC)之外的其余上皮性肺癌。 最常见的NSCLC为鳞状细胞癌、大细胞癌和腺癌,但也有其他发病率较低的类型,所有类型都可以发生少见的组织学变异。 虽然NSCLC与吸烟相关,但从不吸烟的人群也会发生腺癌。与SCLC相比,NSCLC对化疗和放疗的敏感性相对较低。可切除性肿瘤患者可以通过手术或手术加化疗治愈。很多不可切除性肿瘤患者可通过放疗达到局部控制效果,但只有少数患者能治愈。局部晚期不可切除性肿瘤患者可通过放疗联合化疗长期生存。晚期转移性肿瘤患者可通过化疗、靶向药物与其他支持性治疗延长生存期、缓解症状。
据估计,2020年美国肺癌(NSCLC与SCLC)新发病例和死亡病例数分别为:
在美国,肺癌是癌症相关死亡的首要原因。
从1995年到2001年,肺癌患者的5年相对生存率为15.7%。患者5年相对生存率与诊断分期有显著关联性,局部、区域和远端转移肿瘤的5年相对生存率分别为49%、16%与2%。
据估计,2015年中国肺癌新发病例和死亡病例分别为:
在中国,肺癌是首位恶性肿瘤死亡原因。
从2012年到2015年,中国肺癌患者年龄标化的5年生存率为19.7%。
NSCLC来源于中心支气管到终末肺泡的肺上皮细胞。 NSCLC的组织学类型与原发部位相关,反映了支气管到肺泡的呼吸道上皮变化。 鳞状细胞癌通常来源于中心支气管附近。腺癌和支气管肺泡癌通常来源于周围肺组织。
吸烟相关肺癌变是一个多步骤过程。鳞状细胞癌与腺癌有明确的癌前病变。在具有侵袭性之前,肺上皮可能经过下列形态变化:
异型增生和原位癌是主要的癌前病变,因为它们更容易进展为浸润性癌,自行消退的可能性较小。
此外,肺癌切除术后,每年患者有1%到2%几率再次罹患肺癌。
NSCLC具有多种不同的组织学类型。最常见的组织学类型包括:
由于诊断、分期、预后和治疗方法相似,这些组织学类型通常一同分类。
年龄增长是大多数癌症发生的最重要危险因素。肺癌的其他危险因素包括:
肺癌发生的一个最重要的危险因素是吸烟。吸烟者的肺癌风险平均为终生非吸烟者(终生吸烟<100支)的10倍。随着吸烟数量和吸烟时间增加、开始吸烟年龄减小,肺癌危险也增加。
戒烟引起癌前病变减少,发生肺癌的风险也下降。既往吸烟者在戒烟后数年肺癌风险仍偏高。石棉暴露与吸烟对肺癌风险可能具有协同效应。
许多吸烟相关肺癌治愈患者可能再次罹患恶性肿瘤。在美国肺癌研究组试验907例T1N0可切除性肿瘤患者中,每年非肺部第二癌症发生率为1.8%,每年新肺癌发生率为1.6%。
其他研究曾报道过长期生存者第二肿瘤风险增高,其中第二肺癌发生率为10%,所有第二癌症总发生率为20%。
由于有吸烟史的患者肺癌复发的风险持续较高,许多随机对照临床研究为此评估了多种化疗预防方案。但在目前的III期临床研究中,尚无关于β胡萝卜素、视黄醇、13-顺式维甲酸、α生育酚、N-乙酰半胱氨酸或乙酰水杨酸有阳性、可重复性的结果。
[证据等级:1iiA]针对早期的肺癌患者,预防上呼吸消化道出现第二原发性肿瘤的化疗预防方案正在进行临床评估。
(如需了解更多信息,请参见PDQ肺癌预防总结。)
在被认为是肺癌高危患者的人群中,早期发现的唯一筛查方法是低剂量螺旋CT扫描。
胸部影像和痰细胞学筛查肺癌研究未能证明筛查能降低肺癌死亡率。
(如需了解更多信息,请参见PDQ肺癌筛查总结中的低剂量螺旋计算机断层扫描筛查。)
肺癌可能出现症状或在胸部影像学上偶然发现。 症状与体征的产生可能来自原发性局部侵犯或对邻近胸腔结构的压迫、远端转移或副肿瘤综合征。发病时最常见的症状为咳嗽加重或胸痛。其他症状包括:
症状产生的原因可能是局部侵犯或对邻近胸腔结构的压迫,例如压迫食管引起吞咽困难,压迫喉神经引起声音嘶哑,压迫上腔静脉引起面部水肿、头颈部浅静脉扩张等。也可能出现远端转移症状,例如脑转移引起神经源性损害或人格变化,或骨转移引起骨痛。少数患者可能出现副肿瘤疾病症状与体征,例如肥大性骨关节病合并杵状指,或甲状旁腺激素相关蛋白引起高钙血症。体格检查可能发现锁骨上淋巴结肿大、胸腔积液或肺不张、迁延性肺炎、或慢性阻塞性肺病或肺纤维化等合并症相关体征。
对怀疑NSCLC的患者,检查的主要目的是确诊、判断病变受累范围。患者治疗方法的选择取决于组织学、分期、一般情况和合并症等。
确定是否为癌症的方法包括:
在患者开始肺癌治疗之前,必须由一位经验丰富的肺癌病理科医师审阅病理材料。这一点十分关键,因为SCLC对化疗反应良好,通常不进行手术治疗,在显微镜检查中可能与NSCLC混淆。
免疫组化与电镜对诊断与分类的作用有限,多数肺部肿瘤可以通过光镜标准分类。
(关于分期检查与步骤的更多信息,请参见本总结的分期评估章节。)
肺癌基因突变的发现促进了分子靶向治疗的发展,以改善转移性癌症患者的生存期。
尤其是腺癌,人们已经发现编码表皮生长因子受体(EGFR)和下游的丝裂原活化蛋白激酶(MAPK)与磷脂酰肌醇3激酶(PI3K)信号通路组成成分的基因发生特定突变。这些突变可能能够解释药物敏感性与对激酶抑制剂的原发性或获得性耐药机制。
与治疗决策相关的其他遗传异常包括对间变性淋巴瘤激酶(ALK)抑制剂敏感的间变性淋巴瘤激酶(ALK)-酪氨酸激酶受体易位和编码肝细胞生长因子受体的MET(间质上皮转换因子)扩增。MET扩增导致对EGFR酪氨酸激酶抑制剂的继发性耐药。
多项研究曾试图鉴别很多临床病理因素对预后的意义。
与预后不良相关的因素包括:
对于手术不能治疗的肿瘤患者,一般情况较差和体重下降超过10%对预后有不良影响。在积极综合干预的临床试验中排除了这些患者。
在对临床试验数据的多项回顾性分析中,未发现高龄影响治疗的反应或生存期。
(如需了解预后的更多信息,请参见本总结中各分期NSCLC对应的治疗章节。)
因为几乎所有NSCLC患者的治疗效果均不理想,可考虑将合适的患者纳入临床试验。关于正在进行的临床试验信息,请访问NCI网站。
其他包含与肺癌相关信息的PDQ总结包括:
NSCLC is any type of epithelial lung cancer other than small cell lung cancer (SCLC). The most common types of NSCLC are squamous cell carcinoma, large cell carcinoma, and adenocarcinoma, but there are several other types that occur less frequently, and all types can occur in unusual histologic variants. Although NSCLCs are associated with cigarette smoke, adenocarcinomas may be found in patients who have never smoked. As a class, NSCLCs are relatively insensitive to chemotherapy and radiation therapy compared with SCLC. Patients with resectable disease may be cured by surgery or surgery followed by chemotherapy. Local control can be achieved with radiation therapy in a large number of patients with unresectable disease, but cure is seen only in a small number of patients. Patients with locally advanced unresectable disease may achieve long-term survival with radiation therapy combined with chemotherapy. Patients with advanced metastatic disease may achieve improved survival and palliation of symptoms with chemotherapy, targeted agents, and other supportive measures.
Estimated new cases and deaths from lung cancer (NSCLC and SCLC combined) in the United States in 2020:
Lung cancer is the leading cause of cancer-related mortality in the United States.
The 5-year relative survival rate from 1995 to 2001 for patients with lung cancer was 15.7%. The 5-year relative survival rate for patients with local-stage (49%), regional-stage (16%), and distant-stage (2%) disease varies markedly, depending on the stage at diagnosis.
NSCLC arises from the epithelial cells of the lung of the central bronchi to terminal alveoli. The histological type of NSCLC correlates with site of origin, reflecting the variation in respiratory tract epithelium of the bronchi to alveoli. Squamous cell carcinoma usually starts near a central bronchus. Adenocarcinoma and bronchioloalveolar carcinoma usually originate in peripheral lung tissue.
Smoking-related lung carcinogenesis is a multistep process. Squamous cell carcinoma and adenocarcinoma have defined premalignant precursor lesions. Before becoming invasive, lung epithelium may undergo morphological changes that include the following:
Dysplasia and carcinoma in situ are considered the principal premalignant lesions because they are more likely to progress to invasive cancer and less likely to spontaneously regress.
In addition, after resection of a lung cancer, there is a 1% to 2% risk per patient per year that a second lung cancer will occur.
NSCLC is a heterogeneous aggregate of histologies. The most common histologies include the following:
These histologies are often classified together because approaches to diagnosis, staging, prognosis, and treatment are similar.
Increasing age is the most important risk factor for most cancers. Other risk factors for lung cancer include the following:
The single most important risk factor for the development of lung cancer is smoking. For smokers, the risk for lung cancer is on average tenfold higher than in lifetime nonsmokers (defined as a person who has smoked <100 cigarettes in his or her lifetime). The risk increases with the quantity of cigarettes, duration of smoking, and starting age.
Smoking cessation results in a decrease in precancerous lesions and a reduction in the risk of developing lung cancer. Former smokers continue to have an elevated risk of lung cancer for years after quitting. Asbestos exposure may exert a synergistic effect of cigarette smoking on the lung cancer risk.
A significant number of patients cured of their smoking-related lung cancer may develop a second malignancy. In the Lung Cancer Study Group trial of 907 patients with stage T1, N0 resected tumors, the rate was 1.8% per year for nonpulmonary second cancers and 1.6% per year for new lung cancers.
Other studies have reported even higher risks of second tumors in long-term survivors, including rates of 10% for second lung cancers and 20% for all second cancers.
Because of the persistent risk of developing second lung cancers in former smokers, various chemoprevention strategies have been evaluated in randomized control trials. None of the phase III trials using the agents beta carotene, retinol, 13-cis-retinoic acid, [alpha]-tocopherol, N-acetylcysteine, or acetylsalicylic acid has demonstrated beneficial, reproducible results.
[Level of evidence: 1iiA] Chemoprevention of second primary cancers of the upper aerodigestive tract is undergoing clinical evaluation in patients with early-stage lung cancer.
(Refer to the PDQ summary on Lung Cancer Prevention for more information.)
In patients considered at high risk for developing lung cancer, the only screening modality for early detection that has been shown to alter mortality is low-dose helical CT scanning.
Studies of lung cancer screening with chest radiography and sputum cytology have failed to demonstrate that screening lowers lung cancer mortality rates.
(Refer to the Screening by low-dose helical computed tomography subsection in the PDQ summary on Lung Cancer Screening for more information.)
Lung cancer may present with symptoms or be found incidentally on chest imaging. Symptoms and signs may result from the location of the primary local invasion or compression of adjacent thoracic structures, distant metastases, or paraneoplastic phenomena. The most common symptoms at presentation are worsening cough or chest pain. Other presenting symptoms include the following:
Symptoms may result from local invasion or compression of adjacent thoracic structures such as compression involving the esophagus causing dysphagia, compression involving the laryngeal nerves causing hoarseness, or compression involving the superior vena cava causing facial edema and distension of the superficial veins of the head and neck. Symptoms from distant metastases may also be present and include neurological defect or personality change from brain metastases or pain from bone metastases. Infrequently, patients may present with symptoms and signs of paraneoplastic diseases such as hypertrophic osteoarthropathy with digital clubbing or hypercalcemia from parathyroid hormone-related protein. Physical examination may identify enlarged supraclavicular lymphadenopathy, pleural effusion or lobar collapse, unresolved pneumonia, or signs of associated disease such as chronic obstructive pulmonary disease or pulmonary fibrosis.
Investigations of patients with suspected NSCLC focus on confirming the diagnosis and determining the extent of the disease. Treatment options for patients are determined by histology, stage, and general health and comorbidities of the patient.
The procedures used to determine the presence of cancer include the following:
Before a patient begins lung cancer treatment, an experienced lung cancer pathologist must review the pathologic material. This is critical because SCLC, which responds well to chemotherapy and is generally not treated surgically, can be confused on microscopic examination with NSCLC.
Immunohistochemistry and electron microscopy are invaluable techniques for diagnosis and subclassification, but most lung tumors can be classified by light microscopic criteria.
(Refer to the Staging Evaluation section of this summary for more information on tests and procedures used for staging.)
The identification of mutations in lung cancer has led to the development of molecularly targeted therapy to improve the survival of subsets of patients with metastatic disease.
In particular, subsets of adenocarcinoma now can be defined by specific mutations in genes encoding components of the epidermal growth factor receptor (EGFR) and downstream mitogen-activated protein kinases (MAPK) and phosphatidylinositol 3-kinases (PI3K) signaling pathways. These mutations may define mechanisms of drug sensitivity and primary or acquired resistance to kinase inhibitors.
Other genetic abnormalities of potential relevance to treatment decisions include translocations involving the anaplastic lymphoma kinase (ALK)-tyrosine kinase receptor, which are sensitive to ALK inhibitors, and amplification of MET (mesenchymal epithelial transition factor), which encodes the hepatocyte growth factor receptor. MET amplification has been associated with secondary resistance to EGFR tyrosine kinase inhibitors.
Multiple studies have attempted to identify the prognostic importance of a variety of clinicopathologic factors.
Factors that have correlated with adverse prognosis include the following:
For patients with inoperable disease, prognosis is adversely affected by poor performance status and weight loss of more than 10%. These patients have been excluded from clinical trials evaluating aggressive multimodality interventions.
In multiple retrospective analyses of clinical trial data, advanced age alone has not been shown to influence response or survival with therapy.
(Refer to the separate treatment sections for each stage of NSCLC in this summary for more information about prognosis.)
Because treatment is not satisfactory for almost all patients with NSCLC, eligible patients should be considered for clinical trials. Information about ongoing clinical trials is available from the NCI website.
Other PDQ summaries containing information related to lung cancer include the following:
在非小细胞肺癌(NSCLC)中,分期确定对治疗和预后具有重要意义。仔细进行初步诊断评估确定部位与原发及转移性肿瘤受累情况对患者的适当治疗至关重要。
一般通过症状、体征、实验室检查或远端转移风险认知评估是否存在远端转移。如果初步评估提示转移或III期肿瘤患者考虑积极的局部或综合治疗,应进行其他检查,例如骨扫描和颅脑计算机断层扫描(CT)/磁共振成像(MRI)。
分期对治疗选择至关重要。疾病分期是很多临床因素和病理因素的综合结果。
在评估生存期结局报告时应注意临床分期与病理分期之间的区别。
确定疾病分期所用的方法包括:
用于获得组织标本的操作包括支气管镜、纵隔镜检查或前纵隔切开术。NSCLC的病理分期依据包括:
预后与治疗决策基于以下一些因素:
诊断时可根据疾病范围和治疗方法将NSCLC患者分为三组:
如需准确评估淋巴结情况以确定治疗方法,手术纵隔淋巴结分期是评估标准。
准确的纵隔淋巴结分期能够提供重要的预后信息。
证据(淋巴结情况):
CT扫描主要用于确定肿瘤大小。 CT扫描可以向下延伸,以便显现肝和肾上腺区域。胸部与上腹部MRI扫描并不优于CT扫描。
证据(CT扫描):
18F-FDG PET扫描逐渐普及,其应用于分期评估改变了纵隔淋巴结及远端转移的分期评估方法。
一些随机试验评估了18F-FDG PET扫描在潜在可切除的NSCLC中的应用,就非治愈性开胸手术数量是否相对减少的结果不一。
虽然当前证据存在冲突,18F-FDG PET扫描可能识别无法通过手术切除、术前分期检查不能发现的转移灶证据,从而改善早期肺癌患者的结局。
证据(18F-FDG PET扫描):
决策分析发现18F-FDG PET扫描可能降低医疗总支出,因其能发现纵隔淋巴结或其他未检测到的转移灶CT扫描假阴性患者。
一些研究认为假阳性率高得令人无法接受,故18F-FDG PET阳性纵隔病变因省略纵隔镜检查而节省支出并不合理。
一项随机研究发现除常用分期之外增加18F-FDG PET扫描可显著减少开胸手术。
另一项随机研究评估了18F-FDG PET扫描对临床治疗的影响,发现18F-FDG PET扫描可提供相应的分期相关信息,但并未显著减少开胸手术。
CT成像联合18F-FDG PET扫描的灵敏度与特异性均高于单行CT成像。
证据(CT/18F-FDG PET扫描):
对于临床上可切除的NSCLC患者,建议对胸部CT扫描或18F-FDG PET扫描结果阳性的最短径大于1 cm的纵隔淋巴结行活检。而影像学上增大的纵隔淋巴结即便18F-FDG PET扫描结果阴性,也不能完全排除活检可能。如果CT与18F-FDG PET结果不匹配,需要行纵隔镜检测纵隔淋巴结是否癌变。
有脑转移风险的患者可行CT或MRI扫描明确分期。一项研究将332例有切除可能且并无神经系统症状的NSCLC患者进行肺癌术前随机分组,分别接受脑CT或MRI成像以判断有无隐匿的脑转移情况。MRI的术前检出率高于CT扫描(P=0.069),从治疗前到术后12个月的总检出率约为7%。
I期或II期患者的检出率为4%(即200例患者中检出8例),但III期患者的检出率为11.4%(即132例患者中检出15例)。MRI组脑转移的平均最大径显著小于CT组。目前尚不明确MRI的检出率较高能否改善预后。并非所有患者都能耐受MRI,对于不能耐受MRI的患者,可选择对比剂增强CT扫描作为替代检查。
大量非随机、前瞻性或回顾性研究发现18F-FDG PET扫描与常用影像学检查相比,对远端转移灶行分期具有更多诊断学优势,但标准的18F-FDG PET扫描亦有其局限性。18F-FDG PET扫描范围无法延伸至盆腔以下,故可能无法检测到下肢长骨的骨转移。因脑和泌尿系可有18F-FDG PET的代谢示踪剂累积,18F-FDG PET扫描对检测这些部位的转移亦不可靠。
2010年,美国癌症联合委员会(AJCC)和国际抗癌联盟采纳了修订后的国际肺癌分期方法,该方法基于5000多名患者的临床数据库信息。
这些修订内容为不同患者群提供了更多的预后特异性信息,但分期与预后之间的相关性早在PET成像被广泛应用前已经建立。
AJCC对NSCLC进行了TMN(肿瘤、结节、转移灶)分期定义。
分期 | TNM | 描述 | ||
---|---|---|---|---|
分期 | TNM | 描述 | ||
分期 | TNM | 描述 | 论证 | |
分期 | TNM | 描述 | 论证 | |
分期 | TNM | 描述 | 论证 | |
分期 | TNM | 描述 | 论证 | |
隐匿性癌 | TX, N0, M0 | TX=原发性肿瘤无法评估,或痰液、支气管灌洗液中找到恶性细胞,但影像学和支气管镜未发现肿瘤。 | ||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
分期 | TNM | 描述 | ||
0 | Tis, N0, M0 | Tis = 原位癌;SCIS = 原位鳞状细胞癌;AIS:原位腺癌;单纯附壁型腺癌,最大径≤3cm。 | ||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
分期 | TNM | 描述 | 论证 | |
IA1 | T1mi, N0, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
T1a, N0, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IA2 | T1b, N0, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IA3 | T1c, N0, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IB | T2a, N0, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
分期 | TNM | 描述 | 论证 | |
IIA | T2b, N0, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IIB | T1a, N1, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T1b, N1, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T1c, N1, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T2a, N1, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T2b, N1, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T3, N0, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内的独立肿瘤。 | |||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
分期 | TNM | 描述 | 论证 | |
IIIA | T1a, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T1b, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T1c, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T2a, N2, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T2b, N2, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T3, N1, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内的独立肺肿瘤。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T4, N0, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
T4, N1, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
IIIB | T1a, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T1b, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T1c, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T2a, N3, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T2b, N3, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T3, N2, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T4, N2, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
N2=转移至纵隔和/或隆突下淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IIIC | T3, N3, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | ||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T4, N3, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
分期 | TNM | 描述 | 论证 | |
IV | Any T, Any N, M1 | TX=原发性肿瘤无法评估,或痰液、支气管灌洗液中找到恶性细胞,但影像学和支气管镜未发现肿瘤。 | ||
T0 = 无原发性肿瘤的证据。 | ||||
Tis = 原位癌;SCIS = 原位鳞状细胞癌;AIS=原位腺癌:附壁型为主,最大径≤3cm。 | ||||
T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | ||||
T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | ||||
NX = 区域淋巴结无法评估。 | ||||
N0 = 无区域淋巴结转移。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M1 = 远端转移。 | ||||
IVA | Any T, Any N, M1a | Any T = 请参见上文Any T、Any N、M1中的T描述。 | ||
Any N = 请参见上面Any T、Any N、M1中的N描述。 | ||||
M1 = 远端转移。 | ||||
-M1a=独立的肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | ||||
Any T, Any N, M1b | Any T = 请参见上文Any T、Any N、M1中的T描述。 | |||
Any N = 请参见上面Any T、Any N、M1中的N描述。 | ||||
M1 = 远端转移。 | ||||
-M1a=独立肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | ||||
-M1b =单个器官中的单个胸外转移(包括单个非区域性淋巴结受累)。 | ||||
IVB | Any T, Any N, M1c | Any T = 请参见上文Any T、Any N、M1中的T描述。 | ||
Any N = 请参见上面Any T、Any N、M1中的N描述。 | ||||
M1 = 远端转移。 | ||||
-M1a=独立肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | ||||
-M1b =单个器官中的单个胸外转移(包括单个非区域性淋巴结受累)。 | ||||
–M1c =单个器官或多个器官中的多个胸外转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. |
分期 | TNM | 描述 | ||
---|---|---|---|---|
分期 | TNM | 描述 | 论证 | |
分期 | TNM | 描述 | 论证 | |
分期 | TNM | 描述 | 论证 | |
分期 | TNM | 描述 | 论证 | |
0 | Tis, N0, M0 | Tis = 原位癌;SCIS = 原位鳞状细胞癌;AIS:原位腺癌;单纯附壁型腺癌,最大径≤3cm。 | ||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
分期 | TNM | 描述 | 论证 | |
IA1 | T1mi, N0, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
T1a, N0, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IA2 | T1b, N0, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IA3 | T1c, N0, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IB | T2a, N0, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
分期 | TNM | 描述 | 论证 | |
IIA | T2b, N0, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IIB | T1a, N1, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T1b, N1, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T1c, N1, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T2a, N1, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T2b, N1, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T3, N0, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内的独立肿瘤。 | |||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
分期 | TNM | 描述 | 论证 | |
IIIA | T1a, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T1b, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T1c, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T2a, N2, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T2b, N2, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T3, N1, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内的独立肺肿瘤。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T4, N0, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
T4, N1, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
IIIB | T1a, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T1b, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T1c, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T2a, N3, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T2b, N3, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T3, N2, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T4, N2, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
N2=转移至纵隔和/或隆突下淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IIIC | T3, N3, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | ||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T4, N3, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
分期 | TNM | 描述 | 论证 | |
IV | Any T, Any N, M1 | TX=原发性肿瘤无法评估,或痰液、支气管灌洗液中找到恶性细胞,但影像学和支气管镜未发现肿瘤。 | ||
T0 = 无原发性肿瘤的证据。 | ||||
Tis = 原位癌;SCIS = 原位鳞状细胞癌;AIS=原位腺癌:附壁型为主,最大径≤3cm。 | ||||
T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | ||||
T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | ||||
NX = 区域淋巴结无法评估。 | ||||
N0 = 无区域淋巴结转移。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M1 = 远端转移。 | ||||
IVA | Any T, Any N, M1a | Any T = 请参见上文Any T、Any N、M1中的T描述。 | ||
Any N = 请参见上面Any T、Any N、M1中的N描述。 | ||||
M1 = 远端转移。 | ||||
-M1a=独立的肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | ||||
Any T, Any N, M1b | Any T = 请参见上文Any T、Any N、M1中的T描述。 | |||
Any N = 请参见上面Any T、Any N、M1中的N描述。 | ||||
M1 = 远端转移。 | ||||
-M1a=独立肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | ||||
-M1b =单个器官中的单个胸外转移(包括单个非区域性淋巴结受累)。 | ||||
IVB | Any T, Any N, M1c | Any T = 请参见上文Any T、Any N、M1中的T描述。 | ||
Any N = 请参见上面Any T、Any N、M1中的N描述。 | ||||
M1 = 远端转移。 | ||||
-M1a=独立肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | ||||
-M1b =单个器官中的单个胸外转移(包括单个非区域性淋巴结受累)。 | ||||
–M1c =单个器官或多个器官中的多个胸外转移。 | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. |
分期 | TNM | 描述 | 论证 | |
---|---|---|---|---|
分期 | TNM | 描述 | 论证 | |
分期 | TNM | 描述 | 论证 | |
分期 | TNM | 描述 | 论证 | |
IA1 | T1mi, N0, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
T1a, N0, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IA2 | T1b, N0, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IA3 | T1c, N0, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IB | T2a, N0, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
分期 | TNM | 描述 | 论证 | |
IIA | T2b, N0, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IIB | T1a, N1, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T1b, N1, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T1c, N1, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T2a, N1, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T2b, N1, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T3, N0, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内的独立肿瘤。 | |||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
分期 | TNM | 描述 | 论证 | |
IIIA | T1a, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T1b, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T1c, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T2a, N2, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T2b, N2, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T3, N1, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内的独立肺肿瘤。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
T4, N0, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
N0 = 无区域淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
T4, N1, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
M0 = 无远端转移。 | ||||
IIIB | T1a, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T1b, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T1c, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T2a, N3, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T2b, N3, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T3, N2, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T4, N2, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
N2=转移至纵隔和/或隆突下淋巴结转移。 | ||||
M0 = 无远端转移。 | ||||
IIIC | T3, N3, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | ||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
T4, N3, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M0 = 无远端转移。 | ||||
分期 | TNM | 描述 | 论证 | |
IV | Any T, Any N, M1 | TX=原发性肿瘤无法评估,或痰液、支气管灌洗液中找到恶性细胞,但影像学和支气管镜未发现肿瘤。 | ||
T0 = 无原发性肿瘤的证据。 | ||||
Tis = 原位癌;SCIS = 原位鳞状细胞癌;AIS=原位腺癌:附壁型为主,最大径≤3cm。 | ||||
T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | ||||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | ||||
–T1b = 肿瘤最大径>1cm但≤2cm。 | ||||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | ||||
T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | ||||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | ||||
T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | ||||
T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | ||||
NX = 区域淋巴结无法评估。 | ||||
N0 = 无区域淋巴结转移。 | ||||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | ||||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | ||||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | ||||
M1 = 远端转移。 | ||||
IVA | Any T, Any N, M1a | Any T = 请参见上文Any T、Any N、M1中的T描述。 | ||
Any N = 请参见上面Any T、Any N、M1中的N描述。 | ||||
M1 = 远端转移。 | ||||
-M1a=独立的肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | ||||
Any T, Any N, M1b | Any T = 请参见上文Any T、Any N、M1中的T描述。 | |||
Any N = 请参见上面Any T、Any N、M1中的N描述。 | ||||
M1 = 远端转移。 | ||||
-M1a=独立肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | ||||
-M1b =单个器官中的单个胸外转移(包括单个非区域性淋巴结受累)。 | ||||
IVB | Any T, Any N, M1c | Any T = 请参见上文Any T、Any N、M1中的T描述。 | ||
Any N = 请参见上面Any T、Any N、M1中的N描述。 | ||||
M1 = 远端转移。 | ||||
-M1a=独立肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | ||||
-M1b =单个器官中的单个胸外转移(包括单个非区域性淋巴结受累)。 | ||||
–M1c =单个器官或多个器官中的多个胸外转移。 | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | ||||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | ||||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. |
分期 | TNM | 描述 | 论证 |
---|---|---|---|
分期 | TNM | 描述 | 论证 |
分期 | TNM | 描述 | 论证 |
IIA | T2b, N0, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | |||
N0 = 无区域淋巴结转移。 | |||
M0 = 无远端转移。 | |||
IIB | T1a, N1, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | |||
M0 = 无远端转移。 | |||
T1b, N1, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
–T1b = 肿瘤最大径>1cm但≤2cm。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | |||
M0 = 无远端转移。 | |||
T1c, N1, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
–T1b = 肿瘤最大径>1cm但≤2cm。 | |||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | |||
M0 = 无远端转移。 | |||
T2a, N1, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | |||
M0 = 无远端转移。 | |||
T2b, N1, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | |||
M0 = 无远端转移。 | |||
T3, N0, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内的独立肿瘤。 | ||
N0 = 无区域淋巴结转移。 | |||
M0 = 无远端转移。 | |||
分期 | TNM | 描述 | 论证 |
IIIA | T1a, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
M0 = 无远端转移。 | |||
T1b, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
–T1b = 肿瘤最大径>1cm但≤2cm。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
M0 = 无远端转移。 | |||
T1c, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
–T1b = 肿瘤最大径>1cm但≤2cm。 | |||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
M0 = 无远端转移。 | |||
T2a, N2, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
M0 = 无远端转移。 | |||
T2b, N2, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
M0 = 无远端转移。 | |||
T3, N1, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内的独立肺肿瘤。 | ||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | |||
M0 = 无远端转移。 | |||
T4, N0, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | ||
N0 = 无区域淋巴结转移。 | |||
M0 = 无远端转移。 | |||
T4, N1, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | ||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | |||
M0 = 无远端转移。 | |||
IIIB | T1a, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
T1b, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
–T1b = 肿瘤最大径>1cm但≤2cm。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
T1c, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
–T1b = 肿瘤最大径>1cm但≤2cm。 | |||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
T2a, N3, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
T2b, N3, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
T3, N2, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | ||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
M0 = 无远端转移。 | |||
T4, N2, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | ||
N2=转移至纵隔和/或隆突下淋巴结转移。 | |||
M0 = 无远端转移。 | |||
IIIC | T3, N3, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | |
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
T4, N3, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | ||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
分期 | TNM | 描述 | 论证 |
IV | Any T, Any N, M1 | TX=原发性肿瘤无法评估,或痰液、支气管灌洗液中找到恶性细胞,但影像学和支气管镜未发现肿瘤。 | |
T0 = 无原发性肿瘤的证据。 | |||
Tis = 原位癌;SCIS = 原位鳞状细胞癌;AIS=原位腺癌:附壁型为主,最大径≤3cm。 | |||
T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
–T1b = 肿瘤最大径>1cm但≤2cm。 | |||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | |||
T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | |||
T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | |||
T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
NX = 区域淋巴结无法评估。 | |||
N0 = 无区域淋巴结转移。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M1 = 远端转移。 | |||
IVA | Any T, Any N, M1a | Any T = 请参见上文Any T、Any N、M1中的T描述。 | |
Any N = 请参见上面Any T、Any N、M1中的N描述。 | |||
M1 = 远端转移。 | |||
-M1a=独立的肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | |||
Any T, Any N, M1b | Any T = 请参见上文Any T、Any N、M1中的T描述。 | ||
Any N = 请参见上面Any T、Any N、M1中的N描述。 | |||
M1 = 远端转移。 | |||
-M1a=独立肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | |||
-M1b =单个器官中的单个胸外转移(包括单个非区域性淋巴结受累)。 | |||
IVB | Any T, Any N, M1c | Any T = 请参见上文Any T、Any N、M1中的T描述。 | |
Any N = 请参见上面Any T、Any N、M1中的N描述。 | |||
M1 = 远端转移。 | |||
-M1a=独立肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | |||
-M1b =单个器官中的单个胸外转移(包括单个非区域性淋巴结受累)。 | |||
–M1c =单个器官或多个器官中的多个胸外转移。 | |||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | |||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | |||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | |||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | |||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | |||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. |
分期 | TNM | 描述 | 论证 |
---|---|---|---|
分期 | TNM | 描述 | 论证 |
IIIA | T1a, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
M0 = 无远端转移。 | |||
T1b, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
–T1b = 肿瘤最大径>1cm但≤2cm。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
M0 = 无远端转移。 | |||
T1c, N2, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
–T1b = 肿瘤最大径>1cm但≤2cm。 | |||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
M0 = 无远端转移。 | |||
T2a, N2, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
M0 = 无远端转移。 | |||
T2b, N2, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
M0 = 无远端转移。 | |||
T3, N1, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内的独立肺肿瘤。 | ||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | |||
M0 = 无远端转移。 | |||
T4, N0, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | ||
N0 = 无区域淋巴结转移。 | |||
M0 = 无远端转移。 | |||
T4, N1, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | ||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | |||
M0 = 无远端转移。 | |||
IIIB | T1a, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
T1b, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
–T1b = 肿瘤最大径>1cm但≤2cm。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
T1c, N3, M0 | T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | ||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
–T1b = 肿瘤最大径>1cm但≤2cm。 | |||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
T2a, N3, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
T2b, N3, M0 | T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | ||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
T3, N2, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | ||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
M0 = 无远端转移。 | |||
T4, N2, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | ||
N2=转移至纵隔和/或隆突下淋巴结转移。 | |||
M0 = 无远端转移。 | |||
IIIC | T3, N3, M0 | T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | |
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
T4, N3, M0 | T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | ||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M0 = 无远端转移。 | |||
分期 | TNM | 描述 | 论证 |
IV | Any T, Any N, M1 | TX=原发性肿瘤无法评估,或痰液、支气管灌洗液中找到恶性细胞,但影像学和支气管镜未发现肿瘤。 | |
T0 = 无原发性肿瘤的证据。 | |||
Tis = 原位癌;SCIS = 原位鳞状细胞癌;AIS=原位腺癌:附壁型为主,最大径≤3cm。 | |||
T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
–T1b = 肿瘤最大径>1cm但≤2cm。 | |||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | |||
T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | |||
T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | |||
T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
NX = 区域淋巴结无法评估。 | |||
N0 = 无区域淋巴结转移。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M1 = 远端转移。 | |||
IVA | Any T, Any N, M1a | Any T = 请参见上文Any T、Any N、M1中的T描述。 | |
Any N = 请参见上面Any T、Any N、M1中的N描述。 | |||
M1 = 远端转移。 | |||
-M1a=独立的肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | |||
Any T, Any N, M1b | Any T = 请参见上文Any T、Any N、M1中的T描述。 | ||
Any N = 请参见上面Any T、Any N、M1中的N描述。 | |||
M1 = 远端转移。 | |||
-M1a=独立肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | |||
-M1b =单个器官中的单个胸外转移(包括单个非区域性淋巴结受累)。 | |||
IVB | Any T, Any N, M1c | Any T = 请参见上文Any T、Any N、M1中的T描述。 | |
Any N = 请参见上面Any T、Any N、M1中的N描述。 | |||
M1 = 远端转移。 | |||
-M1a=独立肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | |||
-M1b =单个器官中的单个胸外转移(包括单个非区域性淋巴结受累)。 | |||
–M1c =单个器官或多个器官中的多个胸外转移。 | |||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | |||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. | |||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | |||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. |
分期 | TNM | 描述 | 论证 |
---|---|---|---|
IV | Any T, Any N, M1 | TX=原发性肿瘤无法评估,或痰液、支气管灌洗液中找到恶性细胞,但影像学和支气管镜未发现肿瘤。 | |
T0 = 无原发性肿瘤的证据。 | |||
Tis = 原位癌;SCIS = 原位鳞状细胞癌;AIS=原位腺癌:附壁型为主,最大径≤3cm。 | |||
T1 = 肿瘤最大径≤3cm,被肺或脏层胸膜包绕,支气管镜检查无侵及叶支气管近端的证据(未侵主支气管)。 | |||
–T1mi =微浸润性腺癌:腺癌(最大径≤3 cm),附壁型为主,最大径≤5 mm。 | |||
–T1a =肿瘤最大径≤1cm。如果任意大小的肿瘤出现少见的表浅扩散,侵犯局限于支气管壁,可能向近端延伸到主支气管,亦被分类为T1a。 | |||
–T1b = 肿瘤最大径>1cm但≤2cm。 | |||
–T1c = 肿瘤最大径>2 cm但≤3 cm。 | |||
T2=肿瘤最大径>3cm但≤5cm,或肿瘤具有下列特征:侵犯主支气管,但距隆突远端一定距离,侵犯脏层胸膜(PL1或PL2),或伴肺不张或阻塞性肺炎波及肺门区域,但未累及一侧全肺。对于具有这些特征的T2肿瘤,如果≤4cm或无法确定大小,则分类为T2a;如果> 4 cm但≤5cm,则分类为T2b。 | |||
–T2a = 肿瘤最大径>3 cm但≤4 cm。 | |||
–T2b = 肿瘤最大径>4 cm但≤5 cm。 | |||
T3=肿瘤最大径>5 cm但≤7 cm,或直接侵及下列任一器官:壁层胸膜(PL3)、胸壁(含肺上沟瘤)、膈神经、壁层心包;或与原发肿瘤位于同一肺叶内独立的结节。 | |||
T4=肿瘤径>7 cm或任何大小的肿瘤侵犯下列一个或多个结构:隔膜、纵隔、心脏、大血管、气管、喉返神经、食管、椎体、或隆突;位于原发性肿瘤同侧的不同肺叶内独立的结节。 | |||
NX = 区域淋巴结无法评估。 | |||
N0 = 无区域淋巴结转移。 | |||
N1=转移至同侧支气管旁和(或)同侧肺门淋巴结与肺内淋巴结,包括直接侵犯。 | |||
N2=转移至同侧纵隔和/或隆突下淋巴结。 | |||
N3=转移至对侧纵隔、对侧肺门、同侧或对侧斜角肌淋巴结或锁骨上淋巴结。 | |||
M1 = 远端转移。 | |||
IVA | Any T, Any N, M1a | Any T = 请参见上文Any T、Any N、M1中的T描述。 | |
Any N = 请参见上面Any T、Any N、M1中的N描述。 | |||
M1 = 远端转移。 | |||
-M1a=独立的肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | |||
Any T, Any N, M1b | Any T = 请参见上文Any T、Any N、M1中的T描述。 | ||
Any N = 请参见上面Any T、Any N、M1中的N描述。 | |||
M1 = 远端转移。 | |||
-M1a=独立肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | |||
-M1b =单个器官中的单个胸外转移(包括单个非区域性淋巴结受累)。 | |||
IVB | Any T, Any N, M1c | Any T = 请参见上文Any T、Any N、M1中的T描述。 | |
Any N = 请参见上面Any T、Any N、M1中的N描述。 | |||
M1 = 远端转移。 | |||
-M1a=独立肿瘤结节位于对侧肺叶内;伴有胸膜或心包结节或出现恶性胸水或心包积液。多数肺癌患者的胸膜(心包)积液是肿瘤引起。但也有一些患者的胸膜(心包)积液的细胞病理学检查发现肿瘤细胞阴性,积液非血性、非渗出液。如果出现这些情况且临床评估认为积液与肿瘤无关,则积液不作为分期依据。 | |||
-M1b =单个器官中的单个胸外转移(包括单个非区域性淋巴结受累)。 | |||
–M1c =单个器官或多个器官中的多个胸外转移。 | |||
T = 原发性肿瘤;N = 区域淋巴结;M = 远端转移。 | |||
经AJCC授权使用:肺。参考:Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual.8th ed. New York, NY: Springer, 2017, pp 431-56. |
In non-small cell lung cancer (NSCLC), the determination of stage has important therapeutic and prognostic implications. Careful initial diagnostic evaluation to define the location and to determine the extent of primary and metastatic tumor involvement is critical for the appropriate care of patients.
In general, symptoms, physical signs, laboratory findings, or perceived risk of distant metastasis lead to an evaluation for distant metastatic disease. Additional tests such as bone scans and computed tomography (CT)/magnetic resonance imaging (MRI) of the brain may be performed if initial assessments suggest metastases or if patients with stage III disease are under consideration for aggressive local and combined modality treatments.
Stage has a critical role in the selection of therapy. The stage of disease is based on a combination of clinical factors and pathological factors.
The distinction between clinical stage and pathological stage should be considered when evaluating reports of survival outcome.
Procedures used to determine staging include the following:
Procedures used to obtain tissue samples include bronchoscopy, mediastinoscopy, or anterior mediastinotomy. Pathological staging of NSCLC requires the following:
Prognostic and treatment decisions are based on some of the following factors:
At diagnosis, patients with NSCLC can be divided into the following three groups that reflect both the extent of the disease and the treatment approach:
Surgical staging of the mediastinum is considered standard if accurate evaluation of the nodal status is needed to determine therapy.
Accurate staging of the mediastinal lymph nodes provides important prognostic information.
Evidence (nodal status):
CT scanning is primarily used for determining the size of the tumor. The CT scan should extend inferiorly to include the liver and adrenal glands. MRI scans of the thorax and upper abdomen do not appear to yield advantages over CT scans.
Evidence (CT scan):
The wider availability and use of 18F-FDG PET scanning for staging has modified the approach to staging mediastinal lymph nodes and distant metastases.
Randomized trials evaluating the utility of 18F-FDG PET scanning in potentially resectable NSCLC report conflicting results in terms of the relative reduction in the number of noncurative thoracotomies.
Although the current evidence is conflicting, 18F-FDG PET scanning may improve results of early-stage lung cancer by identifying patients who have evidence of metastatic disease that is beyond the scope of surgical resection and that is not evident by standard preoperative staging procedures.
Evidence (18F-FDG PET scan):
Decision analyses demonstrate that 18F-FDG PET scanning may reduce the overall costs of medical care by identifying patients with falsely negative CT scans in the mediastinum or otherwise undetected sites of metastases.
Studies concluded that the money saved by forgoing mediastinoscopy in 18F-FDG PET-positive mediastinal lesions was not justified because of the unacceptably high number of false-positive results.
A randomized study found that the addition of 18F-FDG PET scanning to conventional staging was associated with significantly fewer thoracotomies.
A second randomized trial evaluating the impact of 18F-FDG PET scanning on clinical management found that 18F-FDG PET scanning provided additional information regarding appropriate stage but did not lead to significantly fewer thoracotomies.
The combination of CT imaging and 18F-FDG PET scanning has greater sensitivity and specificity than CT imaging alone.
Evidence (CT/18F-FDG PET scan):
For patients with clinically operable NSCLC, the recommendation is for a biopsy of mediastinal lymph nodes that were found to be larger than 1 cm in shortest transverse axis on chest CT scan or were found to be positive on 18F-FDG PET scan. Negative 18F-FDG PET scanning does not preclude biopsy of radiographically enlarged mediastinal lymph nodes. Mediastinoscopy is necessary for the detection of cancer in mediastinal lymph nodes when the results of the CT scan and 18F-FDG PET scan do not corroborate each other.
Patients at risk for brain metastases may be staged with CT or MRI scans. One study randomly assigned 332 patients with potentially operable NSCLC and no neurological symptoms to brain CT or MRI imaging to detect occult brain metastasis before lung surgery. MRI showed a trend towards a higher preoperative detection rate than CT scan (P = .069), with an overall detection rate of approximately 7% from pretreatment to 12 months after surgery.
Patients with stage I or stage II disease had a detection rate of 4% (i.e., eight detections out of 200 patients); however, individuals with stage III disease had a detection rate of 11.4% (i.e., 15 detections out of 132 patients). The mean maximal diameter of the brain metastases was significantly smaller in the MRI group. Whether the improved detection rate of MRI translates into improved outcome remains unknown. Not all patients are able to tolerate MRI, and for these patients contrast-enhanced CT scan is a reasonable substitute.
Numerous nonrandomized, prospective, and retrospective studies have demonstrated that 18F-FDG PET scanning seems to offer diagnostic advantages over conventional imaging in staging distant metastatic disease; however, standard 18F-FDG PET scans have limitations. 18F-FDG PET scans may not extend below the pelvis and may not detect bone metastases in the long bones of the lower extremities. Because the metabolic tracer used in 18F-FDG PET scanning accumulates in the brain and urinary tract, 18F-FDG PET scanning is not reliable for detection of metastases in these sites.
The Revised International System for Staging Lung Cancer, based on information from a clinical database of more than 5,000 patients, was adopted in 2010 by the American Joint Committee on Cancer (AJCC) and the Union Internationale Contre le Cancer.
These revisions provide greater prognostic specificity for patient groups; however, the correlation between stage and prognosis predates the widespread availability of PET imaging.
The AJCC has designated staging by TNM (tumor, node, metastasis) classification to define NSCLC.
Stage | TNM | Description | ||
---|---|---|---|---|
Stage | TNM | Description | ||
Stage | TNM | Description | Illustration | |
Stage | TNM | Description | Illustration | |
Stage | TNM | Description | Illustration | |
Stage | TNM | Description | Illustration | |
Occult carcinoma | TX, N0, M0 | TX = Primary tumor cannot be assessed, or tumor proven by the presence of malignant cells in sputum or bronchial washings but not visualized by imaging or bronchoscopy. | ||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
Stage | TNM | Description | ||
0 | Tis, N0, M0 | Tis = Carcinoma in situ; SCIS =Squamous cell carcinoma in situ; AIS: Adenocarcinoma in situ; Adenocarcinoma with pure lepidic pattern, ≤3 cm in greatest dimension. | ||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
Stage | TNM | Description | Illustration | |
IA1 | T1mi, N0, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1mi = Minimally invasive adenocarcinoma: Adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
T1a, N0, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1mi = Minimally invasive adenocarcinoma: adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
IA2 | T1b, N0, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1mi = Minimally invasive adenocarcinoma: adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
IA3 | T1c, N0, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1mi = Minimally invasive adenocarcinoma: adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
IB | T2a, N0, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
Stage | TNM | Description | Illustration | |
IIA | T2b, N0, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
IIB | T1a, N1, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T1b, N1, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T1c, N1, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T2a, N1, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T2b, N1, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T3, N0, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (Including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
Stage | TNM | Description | Illustration | |
IIIA | T1a, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T1b, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T1c, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T2a, N2, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T2b, N2, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T3, N1, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T4, N0, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
T4, N1, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
IIIB | T1a, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T1b, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T1c, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T2a, N3, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T2b, N3, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T3, N2, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T4, N2, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
IIIC | T3, N3, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | ||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T4, N3, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
Stage | TNM | Description | Illustration | |
IV | Any T, Any N, M1 | TX = Primary tumor cannot be assessed, or tumor proven by the presence of malignant cells in sputum or bronchial washings but not visualized by imaging or bronchoscopy. | ||
T0 = No evidence of primary tumor. | ||||
Tis = carcinoma in situ; SCIS = squamous cell carcinoma in situ; AIS = adenocarcinoma in situ: Adenocarcinoma with pure lepidic pattern, ≤3 cm in greatest dimension. | ||||
T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||||
–T1mi = Minimally invasive adenocarcinoma: Adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | ||||
T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | ||||
NX = Regional lymph nodes cannot be assessed. | ||||
N0 = No regional lymph node metastasis. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M1 = Distant metastasis. | ||||
IVA | Any T, Any N, M1a | Any T = See T descriptions above in Any T, Any N, M1. | ||
Any N = See N descriptions above in Any T, Any N, M1. | ||||
M1 = Distant metastasis. | ||||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | ||||
Any T, Any N, M1b | Any T = See T descriptions above in Any T, Any N, M1. | |||
Any N = See N descriptions above in Any T, Any N, M1. | ||||
M1 = Distant metastasis. | ||||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | ||||
–M1b = Single extrathoracic metastases in a single organ (including involvement of a single nonregional node). | ||||
IVB | Any T, Any N, M1c | Any T = See T descriptions above in Any T, Any N, M1. | ||
Any N = See N descriptions above in Any T, Any N, M1. | ||||
M1 = Distant metastasis. | ||||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | ||||
–M1b = Single extrathoracic metastases in a single organ (including involvement of a single nonregional node). | ||||
–M1c = Multiple extrathoracic metastases in a single organ or in multiple organs. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. |
Stage | TNM | Description | ||
---|---|---|---|---|
Stage | TNM | Description | Illustration | |
Stage | TNM | Description | Illustration | |
Stage | TNM | Description | Illustration | |
Stage | TNM | Description | Illustration | |
0 | Tis, N0, M0 | Tis = Carcinoma in situ; SCIS =Squamous cell carcinoma in situ; AIS: Adenocarcinoma in situ; Adenocarcinoma with pure lepidic pattern, ≤3 cm in greatest dimension. | ||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
Stage | TNM | Description | Illustration | |
IA1 | T1mi, N0, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1mi = Minimally invasive adenocarcinoma: Adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
T1a, N0, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1mi = Minimally invasive adenocarcinoma: adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
IA2 | T1b, N0, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1mi = Minimally invasive adenocarcinoma: adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
IA3 | T1c, N0, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1mi = Minimally invasive adenocarcinoma: adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
IB | T2a, N0, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
Stage | TNM | Description | Illustration | |
IIA | T2b, N0, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
IIB | T1a, N1, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T1b, N1, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T1c, N1, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T2a, N1, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T2b, N1, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T3, N0, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (Including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
Stage | TNM | Description | Illustration | |
IIIA | T1a, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T1b, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T1c, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T2a, N2, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T2b, N2, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T3, N1, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T4, N0, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
T4, N1, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
IIIB | T1a, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T1b, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T1c, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T2a, N3, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T2b, N3, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T3, N2, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T4, N2, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
IIIC | T3, N3, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | ||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T4, N3, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
Stage | TNM | Description | Illustration | |
IV | Any T, Any N, M1 | TX = Primary tumor cannot be assessed, or tumor proven by the presence of malignant cells in sputum or bronchial washings but not visualized by imaging or bronchoscopy. | ||
T0 = No evidence of primary tumor. | ||||
Tis = carcinoma in situ; SCIS = squamous cell carcinoma in situ; AIS = adenocarcinoma in situ: Adenocarcinoma with pure lepidic pattern, ≤3 cm in greatest dimension. | ||||
T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||||
–T1mi = Minimally invasive adenocarcinoma: Adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | ||||
T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | ||||
NX = Regional lymph nodes cannot be assessed. | ||||
N0 = No regional lymph node metastasis. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M1 = Distant metastasis. | ||||
IVA | Any T, Any N, M1a | Any T = See T descriptions above in Any T, Any N, M1. | ||
Any N = See N descriptions above in Any T, Any N, M1. | ||||
M1 = Distant metastasis. | ||||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | ||||
Any T, Any N, M1b | Any T = See T descriptions above in Any T, Any N, M1. | |||
Any N = See N descriptions above in Any T, Any N, M1. | ||||
M1 = Distant metastasis. | ||||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | ||||
–M1b = Single extrathoracic metastases in a single organ (including involvement of a single nonregional node). | ||||
IVB | Any T, Any N, M1c | Any T = See T descriptions above in Any T, Any N, M1. | ||
Any N = See N descriptions above in Any T, Any N, M1. | ||||
M1 = Distant metastasis. | ||||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | ||||
–M1b = Single extrathoracic metastases in a single organ (including involvement of a single nonregional node). | ||||
–M1c = Multiple extrathoracic metastases in a single organ or in multiple organs. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. |
Stage | TNM | Description | Illustration | |
---|---|---|---|---|
Stage | TNM | Description | Illustration | |
Stage | TNM | Description | Illustration | |
Stage | TNM | Description | Illustration | |
IA1 | T1mi, N0, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1mi = Minimally invasive adenocarcinoma: Adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
T1a, N0, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1mi = Minimally invasive adenocarcinoma: adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
IA2 | T1b, N0, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1mi = Minimally invasive adenocarcinoma: adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
IA3 | T1c, N0, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1mi = Minimally invasive adenocarcinoma: adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
IB | T2a, N0, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
Stage | TNM | Description | Illustration | |
IIA | T2b, N0, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
IIB | T1a, N1, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T1b, N1, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T1c, N1, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T2a, N1, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T2b, N1, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T3, N0, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (Including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
Stage | TNM | Description | Illustration | |
IIIA | T1a, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T1b, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T1c, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T2a, N2, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T2b, N2, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T3, N1, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
T4, N0, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
N0 = No regional lymph node metastasis. | ||||
M0 = No distant metastasis. | ||||
T4, N1, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
M0 = No distant metastasis. | ||||
IIIB | T1a, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T1b, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T1c, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T2a, N3, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T2b, N3, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T3, N2, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T4, N2, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
M0 = No distant metastasis. | ||||
IIIC | T3, N3, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | ||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
T4, N3, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M0 = No distant metastasis. | ||||
Stage | TNM | Description | Illustration | |
IV | Any T, Any N, M1 | TX = Primary tumor cannot be assessed, or tumor proven by the presence of malignant cells in sputum or bronchial washings but not visualized by imaging or bronchoscopy. | ||
T0 = No evidence of primary tumor. | ||||
Tis = carcinoma in situ; SCIS = squamous cell carcinoma in situ; AIS = adenocarcinoma in situ: Adenocarcinoma with pure lepidic pattern, ≤3 cm in greatest dimension. | ||||
T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||||
–T1mi = Minimally invasive adenocarcinoma: Adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | ||||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | ||||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | ||||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | ||||
T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | ||||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | ||||
T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | ||||
T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | ||||
NX = Regional lymph nodes cannot be assessed. | ||||
N0 = No regional lymph node metastasis. | ||||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | ||||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | ||||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | ||||
M1 = Distant metastasis. | ||||
IVA | Any T, Any N, M1a | Any T = See T descriptions above in Any T, Any N, M1. | ||
Any N = See N descriptions above in Any T, Any N, M1. | ||||
M1 = Distant metastasis. | ||||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | ||||
Any T, Any N, M1b | Any T = See T descriptions above in Any T, Any N, M1. | |||
Any N = See N descriptions above in Any T, Any N, M1. | ||||
M1 = Distant metastasis. | ||||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | ||||
–M1b = Single extrathoracic metastases in a single organ (including involvement of a single nonregional node). | ||||
IVB | Any T, Any N, M1c | Any T = See T descriptions above in Any T, Any N, M1. | ||
Any N = See N descriptions above in Any T, Any N, M1. | ||||
M1 = Distant metastasis. | ||||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | ||||
–M1b = Single extrathoracic metastases in a single organ (including involvement of a single nonregional node). | ||||
–M1c = Multiple extrathoracic metastases in a single organ or in multiple organs. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | ||||
T = primary tumor; N = regional lymph node; M = distant metastasis. | ||||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. |
Stage | TNM | Description | Illustration |
---|---|---|---|
Stage | TNM | Description | Illustration |
Stage | TNM | Description | Illustration |
IIA | T2b, N0, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | |||
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis. | |||
IIB | T1a, N1, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | |||
M0 = No distant metastasis. | |||
T1b, N1, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | |||
M0 = No distant metastasis. | |||
T1c, N1, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | |||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | |||
M0 = No distant metastasis. | |||
T2a, N1, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | |||
M0 = No distant metastasis. | |||
T2b, N1, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | |||
M0 = No distant metastasis. | |||
T3, N0, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (Including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | ||
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis. | |||
Stage | TNM | Description | Illustration |
IIIA | T1a, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
T1b, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
T1c, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | |||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
T2a, N2, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
T2b, N2, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
T3, N1, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | ||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | |||
M0 = No distant metastasis. | |||
T4, N0, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | ||
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis. | |||
T4, N1, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | ||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | |||
M0 = No distant metastasis. | |||
IIIB | T1a, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
T1b, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
T1c, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | |||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
T2a, N3, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
T2b, N3, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
T3, N2, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | ||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
T4, N2, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | ||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
IIIC | T3, N3, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
T4, N3, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | ||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
Stage | TNM | Description | Illustration |
IV | Any T, Any N, M1 | TX = Primary tumor cannot be assessed, or tumor proven by the presence of malignant cells in sputum or bronchial washings but not visualized by imaging or bronchoscopy. | |
T0 = No evidence of primary tumor. | |||
Tis = carcinoma in situ; SCIS = squamous cell carcinoma in situ; AIS = adenocarcinoma in situ: Adenocarcinoma with pure lepidic pattern, ≤3 cm in greatest dimension. | |||
T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1mi = Minimally invasive adenocarcinoma: Adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | |||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | |||
T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | |||
T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |||
T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
NX = Regional lymph nodes cannot be assessed. | |||
N0 = No regional lymph node metastasis. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M1 = Distant metastasis. | |||
IVA | Any T, Any N, M1a | Any T = See T descriptions above in Any T, Any N, M1. | |
Any N = See N descriptions above in Any T, Any N, M1. | |||
M1 = Distant metastasis. | |||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | |||
Any T, Any N, M1b | Any T = See T descriptions above in Any T, Any N, M1. | ||
Any N = See N descriptions above in Any T, Any N, M1. | |||
M1 = Distant metastasis. | |||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | |||
–M1b = Single extrathoracic metastases in a single organ (including involvement of a single nonregional node). | |||
IVB | Any T, Any N, M1c | Any T = See T descriptions above in Any T, Any N, M1. | |
Any N = See N descriptions above in Any T, Any N, M1. | |||
M1 = Distant metastasis. | |||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | |||
–M1b = Single extrathoracic metastases in a single organ (including involvement of a single nonregional node). | |||
–M1c = Multiple extrathoracic metastases in a single organ or in multiple organs. | |||
T = primary tumor; N = regional lymph node; M = distant metastasis. | |||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | |||
T = primary tumor; N = regional lymph node; M = distant metastasis. | |||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | |||
T = primary tumor; N = regional lymph node; M = distant metastasis. | |||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. |
Stage | TNM | Description | Illustration |
---|---|---|---|
Stage | TNM | Description | Illustration |
IIIA | T1a, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
T1b, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
T1c, N2, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | |||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
T2a, N2, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
T2b, N2, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
T3, N1, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | ||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | |||
M0 = No distant metastasis. | |||
T4, N0, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | ||
N0 = No regional lymph node metastasis. | |||
M0 = No distant metastasis. | |||
T4, N1, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | ||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | |||
M0 = No distant metastasis. | |||
IIIB | T1a, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
T1b, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
T1c, N3, M0 | T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | ||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | |||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
T2a, N3, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
T2b, N3, M0 | T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | ||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
T3, N2, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | ||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
T4, N2, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | ||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
M0 = No distant metastasis. | |||
IIIC | T3, N3, M0 | T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
T4, N3, M0 | T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | ||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M0 = No distant metastasis. | |||
Stage | TNM | Description | Illustration |
IV | Any T, Any N, M1 | TX = Primary tumor cannot be assessed, or tumor proven by the presence of malignant cells in sputum or bronchial washings but not visualized by imaging or bronchoscopy. | |
T0 = No evidence of primary tumor. | |||
Tis = carcinoma in situ; SCIS = squamous cell carcinoma in situ; AIS = adenocarcinoma in situ: Adenocarcinoma with pure lepidic pattern, ≤3 cm in greatest dimension. | |||
T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1mi = Minimally invasive adenocarcinoma: Adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | |||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | |||
T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | |||
T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |||
T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
NX = Regional lymph nodes cannot be assessed. | |||
N0 = No regional lymph node metastasis. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M1 = Distant metastasis. | |||
IVA | Any T, Any N, M1a | Any T = See T descriptions above in Any T, Any N, M1. | |
Any N = See N descriptions above in Any T, Any N, M1. | |||
M1 = Distant metastasis. | |||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | |||
Any T, Any N, M1b | Any T = See T descriptions above in Any T, Any N, M1. | ||
Any N = See N descriptions above in Any T, Any N, M1. | |||
M1 = Distant metastasis. | |||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | |||
–M1b = Single extrathoracic metastases in a single organ (including involvement of a single nonregional node). | |||
IVB | Any T, Any N, M1c | Any T = See T descriptions above in Any T, Any N, M1. | |
Any N = See N descriptions above in Any T, Any N, M1. | |||
M1 = Distant metastasis. | |||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | |||
–M1b = Single extrathoracic metastases in a single organ (including involvement of a single nonregional node). | |||
–M1c = Multiple extrathoracic metastases in a single organ or in multiple organs. | |||
T = primary tumor; N = regional lymph node; M = distant metastasis. | |||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. | |||
T = primary tumor; N = regional lymph node; M = distant metastasis. | |||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. |
Stage | TNM | Description | Illustration |
---|---|---|---|
IV | Any T, Any N, M1 | TX = Primary tumor cannot be assessed, or tumor proven by the presence of malignant cells in sputum or bronchial washings but not visualized by imaging or bronchoscopy. | |
T0 = No evidence of primary tumor. | |||
Tis = carcinoma in situ; SCIS = squamous cell carcinoma in situ; AIS = adenocarcinoma in situ: Adenocarcinoma with pure lepidic pattern, ≤3 cm in greatest dimension. | |||
T1 = Tumor ≤3 cm in greatest dimension, surrounded by lung or visceral pleura, without bronchoscopic evidence of invasion more proximal than the lobar bronchus (i.e., not in the main bronchus). | |||
–T1mi = Minimally invasive adenocarcinoma: Adenocarcinoma (≤3 cm in greatest dimension) with a predominantly lepidic pattern and ≤5 mm invasion in greatest dimension. | |||
–T1a = Tumor ≤1 cm in greatest dimension. A superficial, spreading tumor of any size whose invasive component is limited to the bronchial wall and may extend proximal to the main bronchus also is classified as T1a, but these tumors are uncommon. | |||
–T1b = Tumor >1 cm but ≤2 cm in greatest dimension. | |||
–T1c = Tumor >2 cm but ≤3 cm in greatest dimension. | |||
T2 = Tumor >3 cm but ≤5 cm or having any of the following features: involves the main bronchus regardless of distance to the carina, but without involvement of the carina; invades visceral pleura (PL1 or PL2); associated with atelectasis or obstructive pneumonitis that extends to the hilar region, involving part or all of the lung. T2 tumors with these features are classified as T2a if ≤4 cm or if the size cannot be determined and T2b if >4 cm but ≤5 cm. | |||
–T2a = Tumor >3 cm but ≤4 cm in greatest dimension. | |||
–T2b = Tumor >4 cm but ≤5 cm in greatest dimension. | |||
T3 = Tumor >5 cm but ≤7 cm in greatest dimension or directly invading any of the following: parietal pleura (PL3), chest wall (including superior sulcus tumors), phrenic nerve, parietal pericardium; or separate tumor nodule(s) in the same lobe as the primary. | |||
T4 = Tumor >7 cm or tumor of any size invading one or more of the following: diaphragm, mediastinum, heart, great vessels, trachea, recurrent laryngeal nerve, esophagus, vertebral body, or carina; separate tumor nodule(s) in an ipsilateral lobe different from that of the primary. | |||
NX = Regional lymph nodes cannot be assessed. | |||
N0 = No regional lymph node metastasis. | |||
N1 = Metastasis in ipsilateral peribronchial and/or ipsilateral hilar lymph nodes and intrapulmonary nodes, including involvement by direct extension. | |||
N2 = Metastasis in ipsilateral mediastinal and/or subcarinal lymph node(s). | |||
N3 = Metastasis in contralateral mediastinal, contralateral hilar, ipsilateral or contralateral scalene, or supraclavicular lymph node(s). | |||
M1 = Distant metastasis. | |||
IVA | Any T, Any N, M1a | Any T = See T descriptions above in Any T, Any N, M1. | |
Any N = See N descriptions above in Any T, Any N, M1. | |||
M1 = Distant metastasis. | |||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | |||
Any T, Any N, M1b | Any T = See T descriptions above in Any T, Any N, M1. | ||
Any N = See N descriptions above in Any T, Any N, M1. | |||
M1 = Distant metastasis. | |||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | |||
–M1b = Single extrathoracic metastases in a single organ (including involvement of a single nonregional node). | |||
IVB | Any T, Any N, M1c | Any T = See T descriptions above in Any T, Any N, M1. | |
Any N = See N descriptions above in Any T, Any N, M1. | |||
M1 = Distant metastasis. | |||
–M1a = Separate tumor nodule(s) in a contralateral lobe; tumor with pleural or pericardial nodules or malignant pleural or pericardial effusion. Most pleural (pericardial) effusions with lung cancer are a result of the tumor. In a few patients, however, multiple microscopic examinations of pleural (pericardial) fluid are negative for tumor, and the fluid is nonbloody and not an exudate. If these elements and clinical judgment dictate that the effusion is not related to the tumor, the effusion should be excluded as a staging descriptor. | |||
–M1b = Single extrathoracic metastases in a single organ (including involvement of a single nonregional node). | |||
–M1c = Multiple extrathoracic metastases in a single organ or in multiple organs. | |||
T = primary tumor; N = regional lymph node; M = distant metastasis. | |||
aReprinted with permission from AJCC: Lung. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 431–56. |
在非小细胞肺癌(NSCLC)中,标准治疗结果只适用于大多数局限期癌症。 所有新诊断的NSCLC患者都可作为评估新治疗的研究候选参与者。
手术可能是本病最有效的治疗方法。 NSCLC患者在肿瘤切除后行术后化疗可能增加获益。 少数患者可通过放疗联合化疗治愈,多数患者可通过放疗联合化疗缓解症状。 预防性颅脑照射(PCI)可能降低脑转移发生率,但目前无生存期获益证据,PCI对生活质量的影响亦不明确。
对于晚期癌症患者,化疗或表皮生长因子受体(EGFR)激酶抑制剂可适度改善中位生存期,但总生存期仍较差。
化疗能短期缓解晚期NSCLC患者的肿瘤相关症状。 一些临床研究尝试评估化疗对肿瘤相关症状与生活质量的影响。 这些研究提示,可通过化疗控制肿瘤相关症状,同时不影响总体生活质量;
但化疗对生活质量的影响需更深入的研究。 通常身体状况较好的老年患者的治疗获益与年轻患者相同。
肺癌中基因突变的发现促进了分子靶向治疗的研发,以提高肿瘤转移患者的生存期。
尤其是在NSCLC患者中发现的EGFR、MAPK、PI3K信号通路基因异常可能揭露了药物敏感性或对激酶抑制剂的原发、获得性耐药的机制。EGFR突变充分预测了EGFR抑制剂有效率增加、无进展生存期延长。未选定的NSCLC中3%到7%出现ALK与EML4基因融合产生易位产物,介导了克唑替尼等药对ALK的药理抑制作用。MET致癌基因编码肝细胞生长因子受体。已发现该基因的扩增与继发性EGFR酪氨酸激酶受体耐药相关。
NSCLC各期的标准治疗选择见表7.
分期(TNM分期标准) | 标准治疗选择 | |
---|---|---|
隐匿性NSCLC | 手术 | |
0期NSCLC | ||
IA与IB期NSCLC | ||
IIA与IIB期NSCLC | ||
IIIA期NSCLC | 已切除或可切除肿瘤 | |
不可切除的肿瘤 | ||
上沟瘤 | ||
侵及胸壁的肿瘤 | ||
IIIB与IIIC期NSCLC | ||
新诊断的复发性IV期NSCLC | ||
一线化疗后的维持治疗(针对四个疗程铂类联合化疗后病情稳定或缓解的患者) | ||
ALK抑制剂(针对ALK易位的患者) | ||
ROS1抑制剂(针对ROS1重排的患者) | ||
BRAF V600E和MEK抑制剂(针对BRAF V600E突变的患者) | ||
进展的复发性IV期NSCLC | ||
ALK =间变性淋巴瘤激酶;BRAF = v-raf鼠肉瘤病毒癌基因同源物B1;EGFR =表皮生长因子受体;MEK = MAPK激酶1;NSCLC =非小细胞肺癌;PD-L1 =编程化死亡配体1;TKI =酪氨酸激酶抑制剂;TNM = T,肿瘤大小以及癌细胞向附近组织扩散;N,癌细胞扩散到附近的淋巴结;M,癌细胞转移或扩散到人体其他部位。 |
除表7所列出的标准治疗选择之外,经过临床评估亦可选择下列治疗:
一些小规模病例分析发现化疗、放疗或放化疗后的氟F 18-脱氧葡萄糖正电子发射断层扫描(18F-FDG PET)上病灶摄取值降低与病理学完全缓解和良好预后相关。
一些研究曾使用不同的评估时机、18F-FDG PET参数与阈值来定义18F-FDG PET缓解。最大标准摄取值(SUV)降低至少80%预测病理学完全缓解的灵敏度为90%,特异性为100%,准确率为96%。
肿瘤SUV值小于4的患者行肿瘤切除术后的中位生存期延长(分别56个月与19个月)。
据报道,放疗后有完全代谢反应的患者中位生存期为31个月,而其他患者为11个月。
18F-FDG PET评估诱导治疗反应的灵敏度与特异性可能高于计算机断层扫描(CT)。最佳检查时机仍待研究;但一项研究发现如果放疗后30天再行18F-FDG PET检查,灵敏度与特异性更佳。
常规治疗后PET-CT扫描无明确的作用。
[证据等级:3iiA]
证据(放化疗或不放化疗后的监测影像):
利用我们先进的临床试验检索工具查找NCI支持的癌症临床试验(当前正在招募患者)。可通过试验地点、治疗类型、药物名称和其他标准缩小检索范围。还可获得关于临床试验的基本信息。
In non-small cell lung cancer (NSCLC), results of standard treatment are poor except for the most localized cancers. All newly diagnosed patients with NSCLC are potential candidates for studies evaluating new forms of treatment.
Surgery is potentially the most curative therapeutic option for this disease. Postoperative chemotherapy may provide an additional benefit to patients with resected NSCLC. Radiation therapy combined with chemotherapy can produce a cure in a small number of patients and can provide palliation in most patients. Prophylactic cranial irradiation may reduce the incidence of brain metastases, but there is no evidence of a survival benefit and the effect of prophylactic cranial irradiation on quality of life is not known.
In patients with advanced-stage disease, chemotherapy or epidermal growth factor receptor (EGFR) kinase inhibitors offer modest improvements in median survival, although overall survival is poor.
Chemotherapy has produced short-term improvement in disease-related symptoms in patients with advanced NSCLC. Several clinical trials have attempted to assess the impact of chemotherapy on tumor-related symptoms and quality of life. In total, these studies suggest that tumor-related symptoms may be controlled by chemotherapy without adversely affecting overall quality of life;
however, the impact of chemotherapy on quality of life requires more study. In general, medically fit elderly patients with good performance status obtain the same benefits from treatment as younger patients.
The identification of gene mutations in lung cancer has led to the development of molecularly targeted therapy to improve the survival of subsets of patients with metastatic disease.
In particular, genetic abnormalities in EGFR, MAPK, and PI3K signaling pathways in subsets of NSCLC may define mechanisms of drug sensitivity and primary or acquired resistance to kinase inhibitors. EGFR mutations strongly predict the improved response rate and progression-free survival of inhibitors of EGFR. Fusions of ALK with EML4 and other genes form translocation products that occur in ranges from 3% to 7% in unselected NSCLC and are responsive to pharmacological inhibition of ALK by agents such as crizotinib. The MET oncogene encodes hepatocyte growth factor receptor. Amplification of this gene has been associated with secondary resistance to EGFR tyrosine kinase inhibitors.
The standard treatment options for each stage of NSCLC are presented in Table 7.
Stage (TNM Definitions) | Standard Treatment Options | |
---|---|---|
Occult NSCLC | Surgery | |
Stage 0 NSCLC | ||
Stages IA and IB NSCLC | ||
Stages IIA and IIB NSCLC | ||
Stage IIIA NSCLC | Resected or resectable disease | |
Unresectable disease | ||
Superior sulcus tumors | ||
Tumors that invade the chest wall | ||
Stages IIIB and IIIC NSCLC | ||
Newly Diagnosed Stage IV, Relapsed, and Recurrent NSCLC | ||
Maintenance therapy following first-line chemotherapy (for patients with stable or responding disease after four cycles of platinum-based combination chemotherapy) | ||
ALK inhibitors (for patients with ALK translocations) | ||
ROS1 inhibitors (for patients with ROS1 rearrangements) | ||
BRAF V600E and MEK inhibitors (for patients with BRAF V600E mutations) | ||
Progressive Stage IV, Relapsed, and Recurrent NSCLC | ||
ALK = anaplastic lymphoma kinase; BRAF = v-raf murine sarcoma viral oncogene homolog B1; EGFR = epidermal growth factor receptor; MEK = MAPK kinase 1; NSCLC = non-small cell lung cancer; PD-L1 = programmed death-ligand 1; TKI = tyrosine kinase inhibitors; TNM = T, size of tumor and any spread of cancer into nearby tissue; N, spread of cancer to nearby lymph nodes; M, metastasis or spread of cancer to other parts of body. |
In addition to the standard treatment options presented in Table 7, treatment options under clinical evaluation include the following:
Several small series have reported that reduction in fluorine F 18-fludeoxyglucose positron emission tomography (18F-FDG PET) after chemotherapy, radiation therapy, or chemoradiation therapy correlates with pathological complete response and favorable prognosis.
Studies have used different timing of assessments, 18F-FDG PET parameters, and cutpoints to define 18F-FDG PET response. Reduction in maximum standardized uptake value (SUV) of higher than 80% predicted for complete pathological response with a sensitivity of 90%, specificity of 100%, and accuracy of 96%.
Median survival after resection was longer for patients with tumor SUV values of lower than 4 (56 months vs. 19 months).
Patients with complete metabolic response following radiation therapy were reported to have median survivals of 31 months versus 11 months.
18F-FDG PET may be more sensitive and specific than computed tomography (CT) scan in assessing response to induction therapy. Optimal timing of imaging remains to be defined; however, one study suggested that greater sensitivity and specificity of 18F-FDG PET is achieved if repeat imaging is delayed until 30 days after radiation therapy.
There is no clear role for routine posttreatment PET-CT scans.
[Level of evidence: 3iiA]
Evidence (surveillance imaging after radiation therapy with or without chemotherapy):
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.
隐匿性肺癌的诊断评估通常包括胸部X光片,必要时选择性使用支气管镜与密切随访(例如计算机断层扫描),以明确原发性肿瘤的部位与性质,这种方式发现的肿瘤通常为早期,可通过手术治愈。
发现原发性肿瘤后,治疗前先明确肿瘤分期。 治疗方式等同于分期相同的其他非小细胞肺癌(NSCLC)患者。
隐匿性NSCLC的标准治疗选择包括以下:
利用我们先进的临床试验检索工具查找NCI支持的癌症临床试验(当前正在招募患者)。可通过试验地点、治疗类型、药物名称和其他标准缩小检索范围。还可获得关于临床试验的基本信息。
In occult lung cancer, a diagnostic evaluation often includes chest x-ray and selective bronchoscopy with close follow-up (e.g., computed tomography scan), when needed, to define the site and nature of the primary tumor; tumors discovered in this fashion are generally early stage and curable by surgery.
After discovery of the primary tumor, treatment involves establishing the stage of the tumor. Therapy is identical to that recommended for other non-small cell lung cancer (NSCLC) patients with similar-stage disease.
Standard treatment options for occult NSCLC include the following:
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.
0期非小细胞肺癌(NSCLC)常进展为侵袭性癌症。
可对患者行支气管镜随访,如检测到病变,可选治愈性治疗方法。
0期NSCLC的标准治疗选择包括以下:
因0期NSCLC患者发生第二肺癌的风险较高,故可行肺段切除术或楔形切除术以保留最多的正常肺组织。根据0期肿瘤的定义,这些肿瘤为非侵袭性且无转移性,故应行手术切除肿瘤,但如果发现病变位于中心,可能需要行肺叶切除术。
中央型病变患者可根据情况选择治愈性经支气管镜治疗。保留肺功能的支气管镜治疗包括光动力治疗、电烧、冷冻治疗与Nd-YAG激光治疗。
证据(支气管镜治疗):
这些治疗方式对早期NSCLC患者的有效性仍有待确定性随机对照临床试验的证实。
第二原发性肿瘤的发病率较高。
利用我们先进的临床试验检索工具查找NCI支持的癌症临床试验(当前正在招募患者)。可通过试验地点、治疗类型、药物名称和其他标准缩小检索范围。还可获得关于临床试验的基本信息。
Stage 0 non-small cell lung cancer (NSCLC) frequently progresses to invasive cancer.
Patients may be offered surveillance bronchoscopies and, if lesions are detected, potentially curative therapies.
Standard treatment options for stage 0 NSCLC include the following:
Segmentectomy or wedge resection are used to preserve maximum normal pulmonary tissue because patients with stage 0 NSCLC are at a high risk for second lung cancers. Because these tumors are by definition noninvasive and incapable of metastasizing, they should be curable with surgical resection; however, such lesions, when identified, are often centrally located and may require a lobectomy.
Patients with central lesions may be candidates for curative endobronchial therapy. Endobronchial therapies that preserve lung function include photodynamic therapy, electrocautery, cryotherapy, and Nd-YAG laser therapy.
Evidence (endobronchial therapies):
Efficacy of these treatment modalities in the management of patients with early NSCLC remains to be proven in definitive randomized controlled trials.
There is a high incidence of second primary cancers developing.
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.
IA期非小细胞肺癌(NSCLC)与IB期NSCLC的标准治疗选择如下所示:
在已经完全切除的I期NSCLC中,化疗和放疗后并未改善结局。
手术是I期NSCLC患者的首选治疗。可根据情况选择肺叶切除术或肺段切除术、楔形切除术或袖状切除术。肺功能受损的原发性肿瘤患者适合行肺段切除术或楔形切除术。术前应详细评估患者的整体病情,尤其是肺储备功能,这对于考量手术获益至关重要。术后即刻死亡率与年龄相关,肺叶切除术的死亡率约为3%-5%。
证据(手术):
目前证据表明在I、II或IIIA期NSCLC患者中,与肺癌切除术联合纵隔淋巴结系统采样相比,肺癌切除术联合CMLND无法改善生存期。
[证据等级:1iiA]
证据局限性(手术):
局灶性与区域性NSCLC患者行手术的有效性结论受限于迄今参与人数较少和试验的潜在方法学缺陷。
很多术后患者发生区域或远端转移。
这些患者适合纳入评估术后化疗或放疗的临床试验。 目前尚未证实术后化疗或放疗可改善已完全切除肿瘤的I期NSCLC患者的预后。
有研究评估了术后(辅助)放疗(PORT)的意义,发现并不能改善完全切除肿瘤的I期NSCLC患者的预后。
证据(辅助放疗):
需行进一步分析判断这些预后是否可能随着技术进步、目标病变体积更明确和放疗射野中心脏受累体积更小而变化。
已经在接受肺叶切除术的I期NSCLC患者中评价了术中应用缝合线(辅助)近距离放射治疗的意义,以改善局部控制;尚未发现可改善预后。
证据(辅助近距离放射治疗):
根据一项荟萃分析的结果,除临床试验之外,不建议I期NSCLC患者在完全切除术后行化疗。
[证据等级:1iiA]
证据(对I期NSCLC的辅助化疗):
尽管有充分证据表明术后化疗对II期或IIIA期NSCLC患者有效,但对IB期NSCLC患者的疗效尚不明确。
证据(对IB期NSCLC的辅助化疗):
对于观察到的生存期差异程度,CALGB-9633的效力可能不足以检出轻度但有临床意义的生存期改善。此外,卡铂与顺铂联合治疗可能会影响结果。 目前,尚无可靠证据证实术后化疗可延长IB期NSCLC患者的生存期
[证据等级:1iiA]
可手术切除但有手术禁忌或肺储备功能良好但无法行手术的I期患者可选择治愈性放疗。
既往常用的主要放疗方法为用兆伏级设备对已知肿瘤体积中平面行约60 Gy至70 Gy分割放疗(1.8–2.0 Gy/天)。
预后:
在多项大型回顾性常规放疗研究中,接受确定性放疗且不可手术病灶患者的5年生存率达10%-30%。
多项研究证实了T1、N0肿瘤患者的预后更佳,并发现该亚组5年生存率达30%-60%。
然而,在接受60 Gy-65 Gy剂量常规放疗的患者中,多达50%的患者仅局部治疗失败。
证据(常规放疗):
很多患者因存在增加围手术期风险的合并症而无法行标准切除术。此时这些患者可考虑观察与放疗。
非随机观察研究对比了切除术、放疗与观察的治疗结果,发现观察组患者的生存时间偏短、死亡率偏高。
放疗技术的改进包括解释肿瘤运动的计划技术、更适形的计划技术(如3-D适形放疗和调强放疗)和治疗期间的影像引导。提供EBRT的现代治疗方法包括大分割放疗和立体定向体部放疗。但是,由于对比性研究的可靠数据有限,无法确定产生最佳效果的放疗方法。
与常规分割放疗相比,大分割放疗涉及在较短的时间内每天给予略高剂量的放疗(如2.4-4.0 Gy)。 多项前瞻性I/II期试验证明,3-4周的60 Gy-70 Gy剂量大分割放疗(每天2.4 Gy-4.0 Gy)后中至重度毒性反应发生率较低,2年OS为50%-60%,2年肿瘤局部控制率为80%-90%。
[证据等级:3iiiA]
SBRT包括在1至2周内给予的极大分割疗程(例如,1至5次治疗)中给予高度适形、高剂量放疗。 常用的治疗方案包括18 Gy×3、12 Gy-12.5 Gy×4、10 Gy-12 Gy×5,其生物学有效剂量明显高于历史常规放疗方案。
多项前瞻性I/II期试验和中心多项研究已证明,SBRT的肺毒性反应发生率较低(症状性放射性肺炎的风险< 10%),2年OS为50%-60%,2年肿瘤控制率为90%-95%。
[证据等级:3iiiA]
证据(SBRT):
常规放疗vs.SBRT(NCT01014130)、大分割放疗vs.SBRT(LUSTRE [NCT01968941])的随机试验正在进行中,目的是确定最佳放疗方案,但立体定向体部放疗已广泛用于临床无法手术的I期NSCLC患者。
处于临床评估阶段的治疗选择如下所示:
利用我们先进的临床试验检索工具查找NCI支持的癌症临床试验(当前正在招募患者)。可通过试验地点、治疗类型、药物名称和其他标准缩小检索范围。还可获得关于临床试验的基本信息。
Standard treatment options for stages IA non-small cell lung cancer (NSCLC) and IB NSCLC include the following:
Chemotherapy and radiation therapy have not been shown to improve outcomes in stage I NSCLC that has been completely resected.
Surgery is the treatment of choice for patients with stage I NSCLC. A lobectomy or segmental, wedge, or sleeve resection may be performed as appropriate. Patients with impaired pulmonary function are candidates for segmental or wedge resection of the primary tumor. Careful preoperative assessment of the patient’s overall medical condition, especially the patient’s pulmonary reserve, is critical in considering the benefits of surgery. The immediate postoperative mortality rate is age related, but a 3% to 5% mortality rate with lobectomy can be expected.
Evidence (surgery):
Current evidence suggests that lung cancer resection combined with CMLND is not associated with improvement in survival compared with lung cancer resection combined with systematic sampling of mediastinal lymph nodes in patients with stage I, II, or IIIA NSCLC.
[Level of evidence: 1iiA]
Limitations of evidence (surgery):
Conclusions about the efficacy of surgery for patients with local and locoregional NSCLC are limited by the small number of participants studied to date and the potential methodological weaknesses of the trials.
Many patients treated surgically subsequently develop regional or distant metastases.
Such patients are candidates for entry into clinical trials evaluating postoperative treatment with chemotherapy or radiation therapy following surgery. At present, neither chemotherapy nor radiation therapy has been found to improve the outcome of patients with stage I NSCLC that has been completely resected.
The value of postoperative (adjuvant) radiation therapy (PORT) has been evaluated and has not been found to improve the outcome of patients with completely resected stage I NSCLC.
Evidence (adjuvant radiation therapy):
Further analysis is needed to determine whether these outcomes can potentially be modified with technical improvements, better definitions of target volumes, and limitation of cardiac volume in the radiation portals.
The value of intraoperative (adjuvant) brachytherapy applied to the suture line has been evaluated in patients undergoing sublobar resections for stage I NSCLC to improve local control; it has not been found to improve outcomes.
Evidence (adjuvant brachytherapy):
Based on a meta-analysis, postoperative chemotherapy is not recommended outside of a clinical trial for patients with completely resected stage I NSCLC.
[Level of evidence: 1iiA]
Evidence (adjuvant chemotherapy for stage I NSCLC):
Although there is sufficient evidence that postoperative chemotherapy is effective in patients with stage II or stage IIIA NSCLC, its usefulness in patients with stage IB NSCLC is less clear.
Evidence (adjuvant chemotherapy for stage IB NSCLC):
Given the magnitude of observed survival differences, CALGB-9633 may have been underpowered to detect small but clinically meaningful improvements in survival. In addition, the use of a carboplatin versus a cisplatin combination might have affected the results. At present, there is no reliable evidence that postoperative chemotherapy improves survival of patients with stage IB NSCLC.
[Level of evidence: 1iiA]
Patients with potentially resectable tumors with medical contraindications to surgery or those with inoperable stage I disease and with sufficient pulmonary reserve may be candidates for radiation therapy with curative intent.
Historically, conventional primary radiation therapy consisted of approximately 60 Gy to 70 Gy delivered with megavoltage equipment to the midplane of the known tumor volume using conventional fractionation (1.8–2.0 Gy per day).
Prognosis:
In the largest retrospective conventional radiation therapy series, patients with inoperable disease treated with definitive radiation therapy achieved 5-year survival rates of 10% to 30%.
Several series demonstrated that patients with T1, N0 tumors had better outcomes, and 5-year survival rates of 30% to 60% were found in this subgroup.
However, local-only failure occurs in as many as 50% of patients treated with conventional radiation therapy to doses in the range of 60 Gy to 65 Gy.
Evidence (conventional radiation therapy):
A substantial number of patients are ineligible for standard surgical resection because of comorbid conditions that are associated with unacceptably high perioperative risk. Observation and radiation therapy may be considered for these patients.
Nonrandomized observational studies comparing treatment outcomes associated with resection, radiation therapy, and observation have demonstrated shorter survival times and higher mortality for patients treated with observation only.
Improvements in radiation techniques include planning techniques to account for tumor motion, more conformal planning techniques (e.g., 3-D conformal radiation therapy and intensity-modulated radiation therapy), and image guidance during treatment. Modern approaches to delivery of EBRT include hypofractionated radiation therapy and stereotactic body radiation therapy (SBRT).However, there are limited reliable data from comparative trials to determine which approaches yield superior outcomes.
Hypofractionated radiation therapy involves the delivery of a slightly higher dose of radiation therapy per day (e.g., 2.4–4.0 Gy) over a shorter period of time compared with conventionally fractionated radiation therapy. Multiple prospective phase I/II trials have demonstrated that hypofractionated radiation therapy to a dose of 60 Gy to 70 Gy delivered over 3 to 4 weeks with 2.4 Gy to 4.0 Gy per day resulted in a low incidence of moderate to severe toxicity, 2-year OS of 50% to 60%, and 2-year tumor local control of 80% to 90%.
[Level of evidence: 3iiiA]
SBRT involves the delivery of highly conformal, high-dose radiation therapy over an extremely hypofractionated course (e.g., one to five treatments) delivered over 1 to 2 weeks. Commonly used regimens include 18 Gy × 3, 12 Gy to 12.5 Gy × 4, and 10 Gy to 12 Gy × 5, and deliver a substantially higher biologically effective dose compared with historic conventional radiation therapy regimens.
Multiple prospective phase I/II trials and institutional series have demonstrated that SBRT results in a low incidence of pulmonary toxicity (<10% risk of symptomatic radiation pneumonitis), 2-year OS of 50% to 60%, and 2-year tumor control of 90% to 95%.
[Level of evidence: 3iiiA]
Evidence (SBRT):
Randomized trials of conventional radiation therapy versus SBRT (NCT01014130), and hypofractionated radiation therapy versus SBRT (LUSTRE [NCT01968941]) are ongoing to determine the optimal radiation therapy regimen, but stereotactic body radiation therapy has been widely adopted for patients with medically inoperable stage I NSCLC.
Treatment options under clinical evaluation include the following:
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.
IIA期非小细胞肺癌(NSCLC)与IIB期NSCLC的标准治疗选择如下所示:
辅助放疗尚未显示出可改善II期NSCLC患者的预后。
手术是II期NSCLC患者的首选治疗。可根据情况选择肺叶切除术、全肺切除术或肺段切除术、楔形切除术或袖状切除术。术前应详细评估患者的整体病情,尤其是肺储备功能,这对于考量手术获益至关重要。不考虑术后即刻死亡率和术后死亡率与年龄相关之外,全肺切除术的死亡率约为5%-8%,肺叶切除术的死亡率约为3%-5%。
证据(手术):
目前证据表明在I、II或IIIA期NSCLC患者中,与肺癌切除术联合纵隔淋巴结系统采样相比,肺癌切除术联合CMLND无法改善生存期。
[证据等级:1iiA]
证据局限性(手术):
局灶性与区域性NSCLC患者行手术的有效性结论受限于迄今参与人数较少和试验的潜在方法学缺陷。
多数证据表明术后顺铂联合化疗显著改善切除肿瘤的II期NSCLC患者的生存期。 术前化疗也可改善生存期。手术与化疗之间的先后顺序及术后放疗对可切除NSCLC患者的获益风险特征仍待评估。
术后许多患者发生区域性或远端转移病灶。
一些随机对照临床试验与荟萃分析评估了术后化疗对I、II、IIIA期NSCLC患者的作用。
证据(辅助放疗):
基于这些数据,完全切除肿瘤的II期肺癌患者可能从术后含顺铂化疗中获益。
[证据等级:1iiA]
一些临床试验研究了术前化疗的作用。 术前化疗的潜在获益包括:
但术前化疗有可能延误治愈性手术。
证据(新辅助化疗):
已评价了术后(辅助)放疗(PORT)的意义。
证据(辅助放疗):
需行进一步分析判断这些预后是否可能随着技术进步、目标病变体积更明确和放疗射野中心脏受累体积更小而变化。
可手术切除但有手术禁忌或肺储备功能良好但无法行手术的II期患者可选择治愈性放疗。
主要放疗方法为用兆伏级设备对已知肿瘤中平面行约60 Gy分割放疗。 原发性肿瘤锥形束野的加量照射常用于强化对肿瘤的局部控制。 为得到最佳治疗效果,需使用模拟器进行详细的治疗计划,准确了解目标体积,尽可能避开关键的正常结构。
预后:
PS较佳的患者中,如能完成治愈性放疗疗程,则预期3年生存率为20%。
证据(放疗):
处于临床评估阶段的治疗选择如下所示:
利用我们先进的临床试验检索工具查找NCI支持的癌症临床试验(当前正在招募患者)。可通过试验地点、治疗类型、药物名称和其他标准缩小检索范围。还可获得关于临床试验的基本信息。
Standard treatment options for stages IIA non-small cell lung cancer (NSCLC) and IIB NSCLC include the following:
Adjuvant radiation therapy has not been shown to improve outcomes in patients with stage II NSCLC.
Surgery is the treatment of choice for patients with stage II NSCLC. A lobectomy, pneumonectomy, or segmental resection, wedge resection, or sleeve resection may be performed as appropriate. Careful preoperative assessment of the patient’s overall medical condition, especially the patient’s pulmonary reserve, is critical in considering the benefits of surgery. Despite the immediate and age-related postoperative mortality rate, a 5% to 8% mortality rate with pneumonectomy or a 3% to 5% mortality rate with lobectomy can be expected.
Evidence (surgery):
Current evidence suggests that lung cancer resection combined with CMLND is not associated with improvement in survival compared with lung cancer resection combined with systematic sampling of mediastinal lymph nodes in patients with stage I, II, or IIIA NSCLC.
[Level of evidence: 1iiA]
Limitations of evidence (surgery):
Conclusions about the efficacy of surgery for patients with local and locoregional NSCLC are limited by the small number of participants studied to date and potential methodological weaknesses of the trials.
The preponderance of evidence indicates that postoperative cisplatin combination chemotherapy provides a significant survival advantage to patients with resected stage II NSCLC. Preoperative chemotherapy may also provide survival benefit. The optimal sequence of surgery and chemotherapy and the benefits and risks of postoperative radiation therapy in patients with resectable NSCLC remain to be determined.
After surgery, many patients develop regional or distant metastases.
Several randomized, controlled trials and meta-analyses have evaluated the use of postoperative chemotherapy in patients with stage I, II, and IIIA NSCLC.
Evidence (adjuvant chemotherapy):
Based on these data, patients with completely resected stage II lung cancer may benefit from postoperative cisplatin-based chemotherapy.
[Level of evidence: 1iiA]
The role of chemotherapy before surgery was tested in clinical trials. The proposed benefits of preoperative chemotherapy include the following:
Preoperative chemotherapy may, however, delay potentially curative surgery.
Evidence (neoadjuvant chemotherapy):
The value of postoperative (adjuvant) radiation therapy (PORT) has been evaluated.
Evidence (adjuvant radiation therapy):
Further analysis is needed to determine whether these outcomes can potentially be modified with technical improvements, better definitions of target volumes, and limitation of cardiac volume in the radiation portals.
Patients with potentially operable tumors with medical contraindications to surgery or those with inoperable stage II disease and with sufficient pulmonary reserve are candidates for radiation therapy with curative intent.
Primary radiation therapy often consists of approximately 60 Gy delivered with megavoltage equipment to the midplane of the volume of the known tumor using conventional fractionation. A boost to the cone down field of the primary tumor is frequently used to enhance local control. Careful treatment planning with precise definition of target volume and avoidance of critical normal structures, to the extent possible, is needed for optimal results; this requires the use of a simulator.
Prognosis:
Among patients with excellent PS, a 3-year survival rate of 20% may be expected if a course of radiation therapy with curative intent can be completed.
Evidence (radiation therapy):
Treatment options under clinical evaluation include the following:
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.
IIIA期非小细胞肺癌(NSCLC)患者群具有较大的异质性。 患者可能有同侧转移、纵膈淋巴结转移、可能切除的T3期肿瘤侵及胸壁、或纵膈受累、支气管周围或肺门淋巴结转移(N1)。 肿瘤表现从有镜下转移的可切除肿瘤到淋巴结转移到有多站淋巴结受累的不可切除的较大肿瘤病变。
预后:
临床IIIA N2期患者的5年总生存(OS)率为10%-15%,但存在较大纵膈受累(即胸片可见病变)的患者5年生存率为2%-5%。 III期NSCLC患者可选的主要治疗方式包括放疗、化疗、手术及联合治疗,具体选择取决于临床情况。
根据肿瘤部位与是否可切除选择治疗方法。
尽管经过细致的术前分期,仍发现一些患者在开胸手术时有纵膈N2淋巴结转移。
切除术后/可切除肿瘤的标准治疗选择包括:
多数证据表明术后顺铂联合化疗显著改善隐匿性N2期NSCLC切除术后患者的生存期。手术与化疗之间的先后顺序及术后放疗对可切除NSCLC患者的获益风险特征仍待评估。
如果完全切除肿瘤与淋巴结具可行性,此类患者可获益于手术及术后化疗。目前证据表明I、II或IIIA期NSCLC患者中,与行肺癌切除术加纵隔淋巴结系统采样相比,行肺癌切除术联合同侧纵膈系统性淋巴结清扫术(CMLND)无法改善生存期。
[证据等级:1iiA]
在一项III期试验中,对IIIA期NSCLC患者在放化疗基础上行手术治疗并没有改善OS,但确实改善了无进展生存期(PFS)和局部控制。
[证据等级:1iiDiii]
证据(手术):
证据局限性(手术):
局灶性与区域性NSCLC患者行手术的有效性结论受限于迄今参与人数较少和试验的潜在方法学缺陷。
一些临床试验研究了术前化疗对III N2期NSCLC患者的作用。术前(新辅助)化疗的潜在获益包括:
证据(新辅助化疗):
术前给予新辅助化疗联合放疗可强化治疗效果,增加肿瘤负荷降期的可能性。 已在II期试验中研究的常用治疗方案包括顺铂/依托泊苷(EP5050)和每周一次卡铂/紫杉醇。
在新辅助放化疗和手术vs.单独同步放化疗的随机试验中,OS无差异,但手术治疗后改善了PFS和局部控制。
[证据等级:1iiDiii]
证据(新辅助放化疗):
迄今为止,尚未使用现代治疗方案对新辅助化疗与新辅助放化疗进行直接比较;最佳新辅助治疗方法仍不明确。
完全切除肿瘤的IIIA期NSCLC患者可能从术后含顺铂化疗中获益。
[证据等级:1iiA]
证据(辅助放疗):
随机对照临床试验证据表明如手术中意外发现IIIA期NSCLC,完全切除术后行化疗可延长生存期。
一些随机对照临床试验与荟萃分析评估了术后化疗对I、II、IIIA期NSCLC患者的作用。
术前或术后给予的联合化疗和放疗应视为试验性治疗,并需要在未来的临床试验中进行评价。
证据(辅助放化疗):
已评估了PORT的意义。
虽然一些研究认为PORT可改善术后淋巴结阳性患者的局部控制,但PORT是否改善患者生存期目前仍有争议。胸部PORT的最佳剂量尚不明确。多数研究的剂量为30 Gy-60 Gy,通常分割为2Gy-2.5Gy。
加拿大国家癌症研究所与组间研究JBR.10(NCT00002583)发现,符合下列任一条件,可以考虑在选定的患者中行PORT来降低局部复发的风险:
证据(辅助放疗):
来自一项大型荟萃分析、多项随机临床试验的子集分析和一项大型人群研究的证据表明PORT可能降低局部复发率。 这些研究中关于PORT对OS影响的结果并不一致。
IIIA N2期患者可获益于PORT,但PORT对早期NSCLC的作用仍待III期临床试验阐明。 需行进一步分析判断是否可随着技术进步、目标病变体积更明确和放疗射野中心脏受累体积更小而变化。
不可切除NSCLC患者的标准治疗选择包括以下:
化疗后放疗与放疗联合化疗可能有益于局部晚期不可切除III期NSCLC患者。
预后:
传统剂量放疗与分割放疗(每日每次1.8-2.0Gy,6-7周总量60-70Gy)使得患者的长期生存期获益5%-10%,结果可复制,且明显缓解症状。
证据(放疗治疗局部晚期不可切除肿瘤):
虽然不可切除IIIA期肿瘤患者可能获益于放疗,但因存在局部与全身复发,故长期效果总体较差。
放疗可缓解NSCLC患者的局部受累症状,例如:
有时可用经支气管镜激光治疗和/或近距离放射治疗缓解近端梗阻性病变。
证据(放疗作为姑息性治疗):
前瞻性随机临床试验与荟萃分析评估了序贯放化疗与同步放化疗的效果。 总体上,同步治疗的生存获益最大,但毒性反应发生率亦增高。
含铂类同步放化疗可能改善局部晚期NSCLC患者的生存期。但当前数据不足以准确评估治疗潜在获益程度,以及最优化疗方案。
证据(放化疗):
两项随机试验(包括RTOG-9410 [NCT01134861])和一项荟萃分析结果表明同步放化疗的生存获益优于序贯放疗,尽管其毒性反应发生率也增加。
[证据等级:1iiA]
证据(同步与序贯放化疗):
20世纪90年代随着放疗-给药技术的改进,包括肿瘤运动管理和成像引导,I/II期临床验证明了剂量递增至74 Gy放疗同步化疗的可行性。
然而,一项比较常规剂量60 Gy与剂量递增至74 Gy联合每周一次卡铂/紫杉醇治疗的III期临床试验未显示局部控制或PFS得到改善,OS随剂量递增而更差(HR,1.38 [1.09–1.76];P = 0.004)。随着剂量递增,5级事件增加不明显(10% vs. 2%),3级食管炎的发生率增加(21% vs. 7%;P =0.0003)。因此,对于III期NSCLC,剂量递增至60 Gy以上的放疗无明显获益。
[证据等级:1iiA]
证据(全身治疗同步放化疗):
主要目的是评价OS。本研究设计为优效性试验,检测到OS HR为0.74的把握度达80%,一类错误为0.05。本研究随机选取598例患者(A组,301;B组,297),收治患者555例(A组,283;B组,272)。
在同步化疗和放疗前添加诱导化疗尚未显示可延长生存期。
[证据等级:1iiA]
度伐利尤单抗是一种选择性人IgG1单克隆抗体,可阻断程序性死亡配体1(PD-L1)与程序性死亡1(PD-1)和CD80结合,从而使T细胞识别并杀死肿瘤细胞。
证据(度伐利尤单抗):
其他全身巩固治疗(包括多西他赛、
吉非替尼、
和tecemotide(MUC1抗原特异性免疫治疗))
的随机试验尚未显示出OS改善。[证据等级:1iiA]
上沟瘤的标准治疗选择包括以下:
上沟部位NSCLC常称为Pancoast瘤,患者发生率不足5%。
上沟瘤通常起源于肺尖部,其治疗具挑战性,因其临近胸廓入口处器官。该部位的肿瘤可能侵及壁层胸膜、胸壁、臂丛、锁骨下静脉、星状神经节和邻近的椎体。但Pancoast瘤可能治愈,尤其是T3,N0期肿瘤。
不良的预后因素包括有纵隔淋巴结转移(N2期)、脊柱或锁骨下静脉受累(T4期)与切除受限(R1或R2)。
尽管放疗是Pancoast瘤治疗方法的一部分,已发表的病例分析所用的放疗剂量、方法和分期差别较大,故很难判断其有效性。
预后:
对仅有临床分期的患者行放疗的小规模回顾性病例分析结果显示5年生存率为0%-40%,生存率与T分期、总放疗剂量和其他预后因素相关。诱导放疗与整块切除可能治愈该病。
证据(放疗):
证据(手术):
证据(放化疗):
20世纪90年代随着放疗-给药技术的改进,包括肿瘤运动管理和成像引导,I/II期临床试验证明了剂量递增至74 Gy放疗同步化疗的可行性。
然而,一项比较常规剂量60 Gy与剂量递增至74 Gy联合每周一次卡铂/紫杉醇治疗的III期临床试验未显示局部控制或PFS得到改善,OS随剂量递增而更差(HR,1.38 [1.09–1.76];P = 0.004)。随着剂量递增,5级事件增加不明显(10% vs. 2%),3级食管炎的发生率增加(21% vs. 7%;P =0.0003)。因此,对于III期NSCLC,剂量递增至60 Gy以上的放疗无明显获益。
[证据等级:1iiA]
证据(全身治疗同步放化疗):
主要目的是评价OS。本研究设计为优效性试验,检测到OS HR为0.74的把握度达80%,一类错误为0.05。本研究随机选取598例患者(A组,301;B组,297),收治患者555例(A组,283;B组,272)。
在同步放化疗前添加诱导化疗尚未显示可延长生存期。
[证据等级:1iiA]
同步放化疗后巩固全身治疗对不可切除NSCLC的作用尚不明确。 巩固全身治疗(包括多西他赛、 吉非替尼、 和tecemotide(MUC1抗原特异性免疫治疗)) 的随机试验尚未显示出OS改善。[证据等级:1iiA]
侵及胸壁肿瘤的标准治疗选择包括:
一些直接侵及胸壁的大肿块原发性肿瘤患者行手术完全切除肿瘤后可长期生存。
证据(根治性手术):
对于切缘不明的患者,推荐辅助化疗,必要时也可选放疗。不完全切除和纵隔淋巴结受累的患者生存率较低。已有研究评估了综合治疗的效果以提高完全切除机率。
处于临床评估阶段的治疗选择如下所示:
利用我们先进的临床试验检索工具查找NCI支持的癌症临床试验(当前正在招募患者)。可通过试验地点、治疗类型、药物名称和其他标准缩小检索范围。还可获得关于临床试验的基本信息。
Patients with stage IIIA non-small cell lung cancer (NSCLC) are a heterogenous group. Patients may have metastases to ipsilateral mediastinal nodes, potentially resectable T3 tumors invading the chest wall, or mediastinal involvement with metastases to peribronchial or hilar lymph nodes (N1). Presentations of disease range from resectable tumors with microscopic metastases to lymph nodes to unresectable, bulky disease involving multiple nodal stations.
Prognosis:
Patients with clinical stage IIIA N2 disease have a 5-year overall survival (OS) rate of 10% to 15%; however, patients with bulky mediastinal involvement (i.e., visible on chest radiography) have a 5-year survival rate of 2% to 5%. Depending on clinical circumstances, the principal forms of treatment that are considered for patients with stage IIIA NSCLC are radiation therapy, chemotherapy, surgery, and combinations of these modalities.
Treatment options vary according to the location of the tumor and whether it is resectable.
Despite careful preoperative staging, some patients will be found to have metastases to mediastinal N2 lymph nodes at thoracotomy.
Standard treatment options for resected/resectable disease include the following:
The preponderance of evidence indicates that postoperative cisplatin combination chemotherapy provides a significant survival advantage to patients with resected NSCLC with occult N2 disease discovered at surgery. The optimal sequence of surgery and chemotherapy and the benefits and risks of postoperative radiation therapy in patients with resectable NSCLC are yet to be determined.
If complete resection of tumor and lymph nodes is possible, such patients may benefit from surgery followed by postoperative chemotherapy. Current evidence suggests that lung cancer resection combined with complete ipsilateral mediastinal lymph node dissection (CMLND) is not associated with improvement in survival compared with lung cancer resection combined with systematic sampling of mediastinal lymph nodes in patients with stage I, II, or IIIA NSCLC.
[Level of evidence: 1iiA]
The addition of surgery to chemoradiation therapy for patients with stage IIIA NSCLC did not result in improved OS in a phase III trial but did improve progression-free survival (PFS) and local control.
[Level of evidence: 1iiDiii]
Evidence (surgery):
Limitations of evidence (surgery):
Conclusions about the efficacy of surgery for patients with local and locoregional NSCLC are limited by the small number of participants studied to date and by the potential methodological weaknesses of the trials.
The role of chemotherapy before surgery in patients with stage III N2 NSCLC has been extensively tested in clinical trials. The proposed benefits of preoperative (neoadjuvant) chemotherapy include the following:
Evidence (neoadjuvant chemotherapy):
Administering concurrent neoadjuvant chemotherapy and radiation therapy before surgery may intensify treatment and increase the likelihood of downstaging the tumor burden. Commonly utilized regimens that have been tested in the phase II setting include cisplatin/etoposide (EP5050) and weekly carboplatin/paclitaxel.
In a randomized trial of neoadjuvant chemoradiation therapy and surgery versus concurrent chemoradiation therapy alone, there was no difference in OS, but surgery improved PFS and local control.
[Level of evidence: 1iiDiii]
Evidence (neoadjuvant chemoradiation therapy):
A direct comparison of neoadjuvant chemotherapy versus neoadjuvant chemoradiation therapy using modern treatment regimens has not been performed to date; the optimal neoadjuvant approach remains unclear.
Patients with completely resected stage IIIA NSCLC may benefit from postoperative cisplatin-based chemotherapy.
[Level of evidence: 1iiA]
Evidence (adjuvant chemotherapy):
Evidence from randomized controlled clinical trials indicates that when stage IIIA NSCLC is encountered unexpectedly at surgery, chemotherapy given after complete resection improves survival.
Several randomized, controlled trials and meta-analyses have evaluated the use of postoperative chemotherapy in patients with stages I, II, and IIIA NSCLC.
Combination chemotherapy and radiation therapy administered before or following surgery should be viewed as investigational and requiring evaluation in future clinical trials.
Evidence (adjuvant chemoradiation therapy):
The value of PORT has been assessed.
Although some studies suggest that PORT can improve local control for node-positive patients whose tumors were resected, it remains controversial whether it can improve survival. The optimal dose of thoracic PORT is not known at this time. The majority of studies cited used doses ranging from 30 Gy to 60 Gy, typically provided in 2 Gy to 2.5 Gy fractions.
As referred to in the National Cancer Institute of Canada (NCIC) Clinical Trials Group JBR.10 study (NCT00002583), PORT may be considered in selected patients to reduce the risk of local recurrence, if any of the following are present:
Evidence (adjuvant radiation therapy):
Evidence from one large meta-analysis, subset analyses of randomized trials, and one large population study suggest that PORT may reduce local recurrence. Results from these studies on the effect of PORT on OS are conflicting.
There is benefit of PORT in stage IIIA N2 disease, and the role of PORT in early stages of NSCLC should be clarified in ongoing phase III trials. Further analysis is needed to determine whether these outcomes can be modified with technical improvements, better definitions of target volumes, and limitation of cardiac volume in the radiation portals.
Standard treatment options for patients with unresectable NSCLC include the following:
Radiation therapy alone, administered sequentially with chemotherapy and concurrently with chemotherapy, may provide benefit to patients with locally advanced unresectable stage III NSCLC.
Prognosis:
Radiation therapy with traditional dose and fractionation schedules (1.8–2.0 Gy per fraction per day to 60–70 Gy in 6–7 weeks) results in reproducible long-term survival benefit in 5% to 10% of patients and significant palliation of symptoms.
Evidence (radiation therapy for locally advanced unresectable tumor):
Although patients with unresectable stage IIIA disease may benefit from radiation therapy, long-term outcomes have generally been poor because of local and systemic relapse.
Radiation therapy may be effective in palliating symptomatic local involvement with NSCLC, such as the following:
In some cases, endobronchial laser therapy and/or brachytherapy has been used to alleviate proximal obstructing lesions.
Evidence (radiation therapy for palliative treatment):
The addition of sequential and concurrent chemotherapy to radiation therapy has been evaluated in prospective randomized trials and meta-analyses. Overall, concurrent treatment may provide the greatest benefit in survival with an increase in toxic effects.
Concomitant platinum-based radiation chemotherapy may improve survival of patients with locally advanced NSCLC. However, the available data are insufficient to accurately define the size of such a potential treatment benefit and the optimal schedule of chemotherapy.
Evidence (chemoradiation therapy):
The results from two randomized trials (including RTOG-9410 [NCT01134861]) and a meta-analysis indicate that concurrent chemotherapy and radiation therapy may provide greater survival benefit, albeit with more toxic effects, than sequential chemotherapy and radiation therapy.
[Level of evidence: 1iiA]
Evidence (concurrent vs. sequential chemoradiation therapy):
With improvement in radiation therapy–delivery technology in the 1990s, including tumor-motion management and image guidance, phase I/II trials demonstrated the feasibility of dose-escalation radiation therapy to 74 Gy with concurrent chemotherapy.
However, a phase III trial of a conventional dose of 60 Gy versus dose escalation to 74 Gy with concurrent weekly carboplatin/paclitaxel did not demonstrate improved local control or PFS, and OS was worse with dose escalation (HR, 1.38 [1.09–1.76]; P = .004). There was a nonsignificant increase in grade 5 events with dose escalation (10% vs. 2%) and higher incidence of grade 3 esophagitis (21% vs. 7%; P =.0003). Thus, there is no clear benefit in radiation dose escalation beyond 60 Gy for stage III NSCLC.
[Level of evidence: 1iiA]
Evidence (systemic therapy for concurrent chemoradiation):
The primary objective was OS. The study was designed as a superiority trial with 80% power to detect an OS HR of 0.74 with a type 1 error of .05. This study randomly assigned 598 patients (arm A, 301; arm B, 297) and treated 555 patients (arm A, 283; arm B, 272).
The addition of induction chemotherapy before concurrent chemotherapy and radiation therapy has not been shown to improve survival.
[Level of evidence: 1iiA]
Durvalumab is a selective human IgG1 monoclonal antibody that blocks programmed death ligand 1 (PD-L1) binding to programmed death 1 (PD-1) and CD80, allowing T cells to recognize and kill tumor cells.
Evidence (durvalumab):
Randomized trials of other consolidation systemic therapies, including docetaxel,
gefitinib,
and tecemotide (MUC1 antigen-specific immunotherapy)
have not shown an improvement in OS.[Level of evidence: 1iiA]
Standard treatment options for superior sulcus tumors include the following:
NSCLC of the superior sulcus, frequently termed Pancoast tumors, occurs in less than 5% of patients.
Superior sulcus tumors usually arise from the apex of the lung and are challenging to treat because of their proximity to structures at the thoracic inlet. At this location, tumors may invade the parietal pleura, chest wall, brachial plexus, subclavian vessels, stellate ganglion, and adjacent vertebral bodies. However, Pancoast tumors are amenable to curative treatment, especially in patients with T3, N0 disease.
Adverse prognostic factors include the presence of mediastinal nodal metastases (N2 disease), spine or subclavian-vessel involvement (T4 disease), and limited resection (R1 or R2).
While radiation therapy is an integral part of the treatment of Pancoast tumors, variations in dose, treatment technique, and staging that were used in various published series make it difficult to determine its effectiveness.
Prognosis:
Small, retrospective series of radiation therapy in patients who were only clinically staged have reported 5-year survival rates of 0% to 40%, depending on T stage, total radiation dose, and other prognostic factors. Induction radiation therapy and en-bloc resection was shown to be potentially curative.
Evidence (radiation therapy):
Evidence (surgery):
Evidence (chemoradiation therapy):
With improvement in radiation therapy–delivery technology in the 1990s, including tumor-motion management and image guidance, phase I/II trials demonstrated the feasibility of dose-escalation radiation therapy to 74 Gy with concurrent chemotherapy.
However, a phase III trial of a conventional dose of 60 Gy versus dose escalation to 74 Gy with concurrent weekly carboplatin/paclitaxel did not demonstrate improved local control or PFS, and OS was worse with dose escalation (HR, 1.38 [1.09–1.76]; P = .004). There was a nonsignificant increase in grade 5 events with dose escalation (10% vs. 2%) and higher incidence of grade 3 esophagitis (21% vs. 7%; P = .0003). Thus, there is no clear benefit in radiation dose escalation beyond 60 Gy for stage III NSCLC.
[Level of evidence: 1iiA]
Evidence (systemic therapy for concurrent chemoradiation):
The primary objective was OS. The study was designed as a superiority trial with 80% power to detect an OS HR of 0.74 with a type 1 error of .05. This study randomly assigned 598 patients (arm A, 301; arm B, 297) and treated 555 patients (arm A, 283; arm B, 272).
The addition of induction chemotherapy before concurrent chemotherapy and radiation therapy has not been shown to improve survival.
[Level of evidence: 1iiA]
The role of consolidation systemic therapy after concurrent chemotherapy and radiation therapy for unresectable NSCLC remains unclear. Randomized trials of consolidation systemic therapy including docetaxel, gefitinib, and tecemotide (MUC1 antigen-specific immunotherapy) have not shown an improvement in OS.[Level of evidence: 1iiA]
Standard treatment options for tumors that invade the chest wall include the following:
Selected patients with bulky primary tumors that directly invade the chest wall can obtain long-term survival with surgical management provided that their tumor is completely resected.
Evidence (radical surgery):
Adjuvant chemotherapy is recommended and radiation therapy is reserved for cases with unclear resection margins. Survival rates were lower in patients who underwent incomplete resection and had mediastinal lymph node involvement. Combined-modality approaches have been evaluated to improve ability to achieve complete resection.
Treatment options under clinical evaluation include the following:
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
根据监测、流行病学和结局(SEER)数据库,IIIB期非小细胞肺癌(NSCLC)的发病率约为17.6%。
临床分期为IIIB期NSCLC的多数患者5年生存率为3%-7%。
一些小规模病例分析发现原发肺叶仅有卫星结节的T4,N0-1期患者的5年生存率为20%。
[证据等级:3iiiA]
IIIB与IIIC期NSCLC的标准治疗选择包括以下:
通常IIIB期和IIIC期NSCLC患者单独手术获益不佳,最好选用初期化疗、化疗联合放疗或单独放疗,具体选择需考虑下列因素:
体能状况较佳的多数患者适合行联合化放疗,但下列患者除外:
针对不可切除III期NSCLC患者的多项随机研究显示,与单独放疗相比,术前或同步含顺铂化疗加胸部放疗可延长生存期。虽然不可切除IIIB期或III期患者可能受益于放疗,但长期预后总体较差,常出现局部和全身复发。一些前瞻性随机临床试验评估了序贯和同步放化疗的效果。
证据(序贯或同步放化疗):
20世纪90年代随着放疗-给药技术的改进,包括肿瘤运动管理和成像引导,I/II期临床试验证明了剂量递增至74 Gy放疗同步化疗的可行性。
然而,一项比较常规剂量60 Gy与剂量递增至74 Gy联合每周一次卡铂/紫杉醇治疗的III期临床试验未显示局部控制或无进展生存期(PFS)得到改善,OS随剂量递增而更差(HR,1.38 [1.09–1.76];P = 0.004)。随着剂量递增,5级事件增加不明显(10% vs. 2%),3级食管炎的发生率增加(21% vs. 7%;P =0.0003)。
[证据等级:1iiA]
在同步放化疗前添加诱导化疗尚未显示可延长生存期。
[证据等级:1iiA]
度伐利尤单抗是一种选择性人IgG1单克隆抗体,可阻断程序性死亡配体1(PD-L1)与程序性死亡1(PD-1)和CD80结合,从而使T细胞识别并杀死肿瘤细胞。
证据(度伐利尤单抗):
其他全身巩固治疗(包括多西他赛、 吉非替尼、 和tecemotide(MUC1抗原特异性免疫治疗)) 的随机试验尚未显示出OS改善。[证据等级:1iiA]
同步化疗和放疗后巩固全身治疗对不可切除NSCLC的作用尚不明确。 巩固全身治疗(包括常规化疗(多西他赛)、 酪氨酸激酶抑制剂(吉非替尼)和免疫治疗(tecemotide:MUC1抗原特异性免疫治疗) 的III期试验尚未显示出OS改善。[证据等级:1iiA]
化疗后放疗与同步放化疗可能有益于局部晚期不可切除III期NSCLC患者。虽然同步放化疗的生存获益最大,但毒性反应发生率亦增高。
预后:
传统剂量放疗与分割放疗(每日每次1.8-2.0Gy,6-7周总量60-70Gy)使得患者的长期生存期获益5%-10%,结果可复制,且明显缓解症状。
证据(放疗治疗局部晚期不可切除肿瘤):
放疗可缓解NSCLC患者的局部受累症状,例如:
有时可用经支气管镜激光治疗和/或近距离放射治疗缓解近端梗阻性病变。
证据(放疗作为姑息性治疗):
PS较差的IIIB或IIIC期患者可选用胸部放疗,以缓解肺部症状(例如咳嗽、呼吸急促、咯血或疼痛)。
[证据等级:3iiiC](如需了解更多信息,请参见PDQ心肺综合征和癌痛总结。)
因IIIB期或IIIC期NSCLC患者的总体结果较差,故这些患者可参与临床试验,可能有助于改善疾病控制。
处于临床评估阶段的治疗选择如下所示:
利用我们先进的临床试验检索工具查找NCI支持的癌症临床试验(当前正在招募患者)。可通过试验地点、治疗类型、药物名称和其他标准缩小检索范围。还可获得关于临床试验的基本信息。
On the basis of the Surveillance, Epidemiology, and End Results (SEER) program registry, the estimated incidence of stage IIIB non-small cell lung cancer (NSCLC) is 17.6%.
The anticipated 5-year survival for the vast majority of patients who present with clinical stage IIIB NSCLC is 3% to 7%.
In small case series, selected patients with T4, N0-1 disease, solely as the result of satellite tumor nodule(s) within the primary lobe, have been reported to have 5-year survival rates of 20%.
[Level of evidence: 3iiiA]
Standard treatment options for stages IIIB NSCLC and IIIC NSCLC include the following:
In general, patients with stages IIIB and IIIC NSCLC do not benefit from surgery alone and are best managed by initial chemotherapy, chemotherapy plus radiation therapy, or radiation therapy alone, depending on the following:
Most patients with excellent PS are candidates for combined-modality chemotherapy and radiation therapy with the following exceptions:
Many randomized studies of patients with unresectable stage III NSCLC show that treatment with preoperative or concurrent cisplatin-based chemotherapy and radiation therapy to the chest is associated with improved survival compared with treatment that uses radiation therapy alone. Although patients with unresectable stages IIIB or IIIC disease may benefit from radiation therapy, long-term outcomes have generally been poor, often the result of local and systemic relapse. The addition of sequential and concurrent chemotherapy to radiation therapy has been evaluated in prospective randomized trials.
Evidence (sequential or concurrent chemotherapy and radiation therapy):
With improvement in radiation therapy–delivery technology in the 1990s, including tumor-motion management and image guidance, phase I/II trials demonstrated the feasibility of dose-escalation radiation therapy to 74 Gy with concurrent chemotherapy.
However, a phase III trial of a conventional dose of 60 Gy versus dose escalation to 74 Gy with concurrent weekly carboplatin/paclitaxel did not demonstrate improved local control or progression-free survival (PFS), and OS was worse with dose escalation (HR, 1.38 [1.09–1.76]; P = .004). There was a nonsignificant increase in grade 5 events with dose escalation (10% vs. 2%) and higher incidence of grade 3 esophagitis (21% vs. 7%; P = .0003).
[Level of evidence: 1iiA]
The addition of induction chemotherapy before concurrent chemotherapy and radiation therapy has not been shown to improve survival.
[Level of evidence: 1iiA]
Durvalumab is a selective human IgG1 monoclonal antibody that blocks programmed death ligand 1 (PD-L1) binding to programmed death 1 (PD-1) and CD80, allowing T cells to recognize and kill tumor cells.
Evidence (durvalumab):
Randomized trials of other consolidation systemic therapies, including docetaxel, gefitinib, and tecemotide (MUC1 antigen-specific immunotherapy) have not shown an improvement in OS.[Level of evidence: 1iiA]
The role of consolidation systemic therapy after concurrent chemotherapy and radiation therapy for unresectable NSCLC remains unclear. Phase III trials of consolidation systemic therapy including conventional chemotherapy (docetaxel), tyrosine kinase inhibitors (gefitinib), and immunotherapy (tecemotide: MUC1 antigen-specific immunotherapy) have not shown an improvement in OS.[Level of evidence: 1iiA]
Radiation therapy alone, administered sequentially or concurrently with chemotherapy, may provide benefit to patients with locally advanced unresectable stage III NSCLC. However, combination chemoradiation therapy delivered concurrently provides the greatest benefit in survival with an increase in toxic effects.
Prognosis:
Radiation therapy with traditional dose and fractionation schedules (1.8–2.0 Gy per fraction per day to 60–70 Gy in 6–7 weeks) results in reproducible long-term survival benefit in 5% to 10% of patients and significant palliation of symptoms.
Evidence (radiation therapy for locally advanced unresectable tumor):
Radiation therapy may be effective in palliating symptomatic local involvement with NSCLC, such as the following:
In some cases, endobronchial laser therapy and/or brachytherapy has been used to alleviate proximal obstructing lesions.
Evidence (radiation therapy for palliative treatment):
Patients with stages IIIB or IIIC disease with poor PS are candidates for chest radiation therapy to palliate pulmonary symptoms (e.g., cough, shortness of breath, hemoptysis, or pain).
[Level of evidence: 3iiiC] (Refer to the PDQ summaries on Cardiopulmonary Syndromes and Cancer Pain for more information.)
Because of the poor overall results, patients with stages IIIB or IIIC NSCLC are candidates for clinical trials, which may lead to improvement in the control of disease.
Treatment options under clinical evaluation include the following:
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.
40%的新诊断非小细胞肺癌(NSCLC)患者为IV期肿瘤。治疗目的是延长生存期与控制疾病相关症状。 治疗选择包括细胞毒性药物化疗、靶向药物和免疫治疗。 影响治疗选择的因素包括合并症、体能状况评分(PS)、肿瘤的组织学和分子遗传学特征。因此,在开始治疗之前,评估肿瘤基因组变化和程序性死亡配体1(PD-L1)表达至关重要。放疗和手术常用于缓解某些患者的症状。
在体能状况评分良好的IV期肿瘤患者中开展的随机对照临床试验显示含顺铂化疗可改善患者生存期并可缓解疾病相关症状。
[证据等级:1iiA]紫杉醇与卡铂联合贝伐单抗可能有助于使组织学非鳞状细胞、身体状况评分良好、无咯血或其他出血性疾病病史且近期亦无心血管事件的患者获益。以EGFR酪氨酸激酶抑制剂(TKI)取代一线或二线化疗可能有助于使携带EGFR外显子19或21突变的肿瘤患者,尤其是东亚患者、从未吸烟者与腺癌患者获益。以ALK或ROS1抑制剂取代一线或二线化疗可能有助于使携带间变性淋巴瘤激酶(ALK)易位或ROS1重排的肿瘤患者获益。帕博利珠单抗治疗可延长表达PD-L1(免疫组织化学> 50%)的肿瘤患者的生存期。卡铂加培美曲塞化疗中添加帕博利珠单抗治疗非鳞状晚期肺癌后患者生存期延长,与PD-L1表达水平无关。
[证据等级:1iiA] 纳武利尤单抗、多西他赛、培美曲塞或帕博利珠单抗二线全身治疗PD-L1阳性肿瘤后PS良好患者(在一线治疗中未接受相同或相似药物治疗)的生存期延长。
[证据等级:1iiA]
尚不确定全身治疗在东部肿瘤协作组PS低于2的患者中的作用。
腺癌患者可能从培美曲塞治疗中获益和贝伐珠单抗以及帕博利珠单抗联合化疗中获益。
有证据表明,即PS良好且合并症有限的老年患者可从联合化疗中获益。单纯年龄无法说明晚期NSCLC患者的治疗相关决策。与单独的支持治疗相比,接受化疗的PS良好老年患者的生存期更长,生活质量更佳。当将老年患者(70-79岁)的数据外推到80岁或80岁以上患者时,应谨慎行事,因为只有极少数80岁或80岁以上患者入组了临床试验,并且该组的获益仍有待证明。
证据(年龄 vs.合并症):
PS是NSCLC患者生存期最重要的预后因素之一。
通过回顾性分析和前瞻性临床试验评估了该组患者治疗的获益。
结果支持进一步评价化疗在转移性和局部晚期NSCLC中的作用;然而,由于确定了当前含铂化疗联合治疗的疗效,故不能将任何特定治疗方案视为标准疗法。在临床试验以外,仅PS良好且可评估肿瘤病变的患者可以接受化疗,在充分了解其预期风险和有限获益后才能进行此治疗。
证据(PS):
这项研究在巴西的八个研究中心和美国的一个中心进行,报告的OS和PFS率高于大多数(尽管不是全部)其他已发表研究的历史记录。 这可能表明患者选择有所差异。
新诊断为复发性IV期疾病患者的标准治疗选择包括:
在随机对照试验和荟萃分析中已充分评估了用于治疗晚期NSCLC患者的化疗药物的类型和数量。
多项随机试验评估了多种药物联合顺铂或卡铂在先前未接受治疗的晚期NSCLC患者中的有效性。 根据试验的荟萃分析,可以得出以下结论:
证据(联合化疗):
在活性药物联合治疗中,除了卡铂、培美曲塞和帕博利珠单抗之外。无法就药物剂量和给药方案对组织学非鳞状肿瘤患者提出明确的建议。
证据(药物和给药方案):
证据(贝伐珠单抗):
证据(西妥昔单抗):
证据(Necitumumab):
一种经过广泛研究的NSCLC治疗策略是对化疗初次反应后的维持治疗。已研究的维持治疗选择包括:
很多随机临床试验评估了继续沿用一线联合细胞毒性化疗药超过3-4疗程的效果。
证据(一线化疗后的维持治疗):
这些数据表明,非鳞状NSCLC患者的PFS和OS可以通过继续四个疗程以上的有效化疗或立即开始替代化疗而得到改善。然而,PFS改善因不良事件的增加而受到抑制,这些不良事件包括额外的细胞毒性化疗以及未持续改善的生活质量。对于疾病稳定或对一线治疗有反应的患者,证据不支持在病情进展前继续联合细胞毒性化疗或在病情进展前开始不同的化疗。总之,这些试验表明,对于疾病治疗无反应的患者,应在疾病进展时或在四个疗程治疗后停止一线细胞毒性联合化疗;最多可以给予6个疗程治疗。
对于在四至六个疗程的铂类药物联合化疗后有反应或疾病稳定的非鳞状NSCLC患者,应考虑使用培美曲塞维持化疗。
证据(一线含铂药物联合化疗继之以培美曲塞治疗):
证据(含铂双药化疗后厄洛替尼维持治疗):
某些患者可能获益于EGFR TKI单药治疗。在未行过化疗的携带EGFR突变的NSCLC患者中开展的随机对照临床试验显示EGFR抑制剂与联合化疗相比,提高PFS,但不提高OS,且毒性反应较轻。
证据(奥希替尼):
奥希替尼已被FDA批准用于EGFR突变NSCLC(外显子19缺失或L858R)的一线治疗。
证据(吉非替尼):
证据(厄洛替尼):
证据(阿法替尼):
证据(阿来替尼):
证据(克唑替尼):
证据(塞瑞替尼):
证据(布加替尼):
证据(Lorlatinib)。
大约1%的NSCLC患者出现ROS1重排。
克唑替尼已获批用于治疗肿瘤呈ROS1阳性的转移性NSCLC患者,不考虑既往全身治疗的次数。
证据(克唑替尼):
BRAF V600E突变,见于1%到2%的肺腺癌。
证据(达拉非尼和曲美替尼):
达拉非尼和曲美替尼联合治疗已获批,可用于治疗通过FDA批准的试验检测出其肿瘤携带BRAF V600E突变的NSCLC患者。
NTRK中的体细胞基因融合发生在许多实体瘤中,包括少于0.5%的NSCLC肿瘤。
不吸烟的肺腺癌患者出现这些融合的概率更高。
证据(Larotrectinib):
Larotrectinib已获得FDA批准,用于治疗携带NTRK基因融合且无已知获得性耐药突变的局部晚期或转移性肿瘤患者,以及无令人满意的替代治疗或在治疗后癌症进展的患者。
帕博利珠单抗是一种人源化单克隆抗体,可抑制肿瘤细胞上表达的PD-1共抑制性免疫检查点与浸润免疫细胞及其配体PD-L1和程序性细胞死亡配体2(PD-L2)之间的相互作用。
证据(帕博利珠单抗联合化疗):
证据(帕博利珠单抗单药治疗):
帕博利珠单抗联合培美曲塞和卡铂已获得FDA批准,作为转移性非鳞状NSCLC患者的一线治疗,不考虑PD-L1表达水平。帕博利珠单抗也获批作为一线单药治疗,用于治疗肿瘤表达PD-L1(经FDA批准的试验确定染色≥50%)的NSCLC患者。在接受帕博利珠单抗之前,EGFR或ALK基因组肿瘤异常的患者应先行FDA批准的治疗确定疾病进展(参见FDA的帕博利珠单抗标签)。
放疗可有效缓解NSCLC局部受累症状患者,例如:
有时可用经支气管镜激光治疗和/或近距离放射治疗缓解近端梗阻性病变。
虽然EBRT常用于缓解症状,但对于何时应使用分割方案尚未达成共识。尽管不同多分割治疗方案缓解症状的程度相似,
NCT00003685临床试验显示,与大分割或标准治疗方案相比,单独分割放疗缓解症状的效果不佳。
[证据等级:1iiC]有证据表明,PS较佳的患者行高剂量放疗后生存期适度延长。
[证据等级:1iiA]对于密切观察中的无症状患者,治疗通常应适当推迟到肿瘤进展产生症状或体征时再进行。
证据(放疗):
最初切除的支气管癌发生孤立性肺转移可能不常见。 肺是原发性肺癌患者中第二原发性恶性肿瘤的发病部位。 新病变是新的原发性癌还是转移灶可能很难确定。 研究表明,在大多数患者中,新病变是第二原发性肿瘤,在切除后,一些患者可以实现长期生存。因此,如果第一原发性肿瘤得到控制,应切除第二原发性肿瘤(如果可能)。
切除原发性NSCLC病灶后出现孤立性脑转移且无颅外肿瘤迹象的患者可通过手术切除脑转移瘤和术后全脑放疗来延长无病生存期。
在这种情况下无法切除的脑转移瘤可采用立体定向放射外科手术进行治疗。
约50%的患者接受切除术和术后放疗后肿瘤在大脑中复发。某些患者将适合进一步治疗。
对于选择的PS良好且在脑外无转移的患者,治疗选择包括再次手术或立体定向放射手术。
对于大多数患者,可以考虑其他放疗;但是,这种治疗的姑息治疗作用有限。
[证据等级:3iiiDiii]
处于临床评估阶段中新诊断的复发性IV期NSCLC治疗选择(一线治疗)包括
利用我们先进的临床试验检索工具查找NCI支持的癌症临床试验(当前正在招募患者)。可通过试验地点、治疗类型、药物名称和其他标准缩小检索范围。还可获得关于临床试验的基本信息。
Forty percent of patients with newly diagnosed non-small cell lung cancer (NSCLC) have stage IV disease. Treatment goals are to prolong survival and control disease-related symptoms. Treatment options include cytotoxic chemotherapy, targeted agents, and immunotherapy. Factors influencing treatment selection include comorbidity, performance status (PS), histology, and molecular and immunologic features of the cancer. Therefore, assessment of tumor-genomic changes and programmed death-ligand 1 (PD-L1) expression is critical before initiating therapy. Radiation therapy and surgery are generally used in selective cases for symptom palliation.
Randomized controlled trials of patients with stage IV disease and good PS have shown that cisplatin-based chemotherapy improves survival and palliates disease-related symptoms.
[Level of evidence: 1iiA] Patients with nonsquamous cell histology, good PS, no history of hemoptysis or other bleeding, or recent history of cardiovascular events may benefit from the addition of bevacizumab to paclitaxel and carboplatin. Patients with tumors harboring sensitizing mutations in exons 19 or 21 of EGFR, particularly those from East Asia, never smokers, and those with adenocarcinoma may benefit from EGFR tyrosine kinase inhibitors (TKI) as an alternative to first- or second-line chemotherapy. Patients with tumors harboring anaplastic lymphoma kinase (ALK) translocations or ROS1 rearrangements may benefit from ALK or ROS1 inhibitors as an alternative to first- or second-line chemotherapy. Patients with tumors expressing PD-L1 (>50% by immunohistochemistry) have improved survival with pembrolizumab. The addition of pembrolizumab to carboplatin plus pemetrexed chemotherapy for nonsquamous advanced lung cancer improves survival irrespective of PD-L1 expression.
[Level of evidence: 1iiA] Second-line systemic therapy with nivolumab, docetaxel, pemetrexed, or pembrolizumab for PD-L1-positive tumors also improves survival in patients with good PS (who have not received the same or a similar agent in the first-line setting).
[Level of evidence: 1iiA]
The role of systemic therapy in patients with an Eastern Cooperative Oncology Group PS below 2 is less certain.
Patients with adenocarcinoma may benefit from pemetrexed and bevacizumab as well as from combination chemotherapy with pembrolizumab.
Evidence supports the concept that elderly patients with good PS and limited comorbidity may benefit from combination chemotherapy. Age alone should not dictate treatment-related decisions in patients with advanced NSCLC. Elderly patients with a good PS enjoy longer survival and a better quality of life when treated with chemotherapy compared with supportive care alone. Caution should be exercised when extrapolating data for elderly patients (aged 70–79 years) to patients aged 80 years or older because only a very small number of patients aged 80 years or older have been enrolled on clinical trials, and the benefit in this group is uncertain.
Evidence (age vs. comorbidity):
PS is among the most important prognostic factors for survival of patients with NSCLC.
The benefit of therapy for this group of patients has been evaluated through retrospective analyses and prospective clinical trials.
The results support further evaluation of chemotherapeutic approaches for both metastatic and locally advanced NSCLC; however, the efficacy of current platinum-based chemotherapy combinations is such that no specific regimen can be regarded as standard therapy. Outside of a clinical trial setting, chemotherapy should be given only to patients with good PS and evaluable tumor lesions, who desire this treatment after being fully informed of its anticipated risks and limited benefits.
Evidence (PS):
This study, which was performed in eight centers in Brazil and one center in the United States, reported rates of OS and PFS that were higher than has historically been noted in most, although not all, other published studies. This may indicate differences in patient selection.
Standard treatment options for patients with newly diagnosed stage IV, relapsed, and recurrent disease include the following:
The type and number of chemotherapy drugs to be used for the treatment of patients with advanced NSCLC has been extensively evaluated in randomized controlled trials and meta-analyses.
Several randomized trials have evaluated various drugs combined with either cisplatin or carboplatin in previously untreated patients with advanced NSCLC. On the basis of meta-analyses of the trials, the following conclusions can be drawn:
Evidence (combination chemotherapy):
Among the active combinations, definitive recommendations regarding drug dose and schedule cannot be made, with the exception of carboplatin, pemetrexed, and pembrolizumab for patients with nonsquamous tumor histology.
Evidence (drug and dose schedule):
Evidence (bevacizumab):
Evidence (cetuximab):
Evidence (necitumumab):
One extensively investigated treatment strategy in NSCLC is maintenance therapy after initial response to chemotherapy. Options for maintenance therapy that have been investigated include the following:
Multiple randomized trials have evaluated the efficacy of continuing first-line combination cytotoxic chemotherapy beyond three to four cycles.
Evidence (maintenance therapy following first-line chemotherapy):
These data suggest that PFS and OS for patients with nonsquamous NSCLC may be improved either by continuing an effective chemotherapy beyond four cycles or by immediate initiation of alternative chemotherapy. The improvement in PFS, however, is tempered by an increase in adverse events including additional cytotoxic chemotherapy and no consistent improvement in quality of life. For patients who have stable disease or who respond to first-line therapy, evidence does not support the continuation of combination cytotoxic chemotherapy until disease progression or the initiation of a different chemotherapy before disease progression. Collectively, these trials suggest that first-line cytotoxic combination chemotherapy should be stopped at disease progression or after four cycles in patients whose disease is not responding to treatment; it can be administered for no more than six cycles.
For patients with nonsquamous NSCLC who have a response or stable disease after four to six cycles of platinum combination chemotherapy, maintenance chemotherapy with pemetrexed should be considered.
Evidence (first-line platinum-based combination chemotherapy followed by pemetrexed):
Evidence (maintenance erlotinib following platinum-based doublet chemotherapy):
Selective patients may benefit from single-agent EGFR TKIs. Randomized controlled trials of patients with chemotherapy-naïve NSCLC and EGFR mutations have shown that EGFR inhibitors improved PFS but not OS and have favorable toxicity profiles compared with combination chemotherapy.
Evidence (osimertinib):
Osimertinib was approved by the FDA for first-line treatment of EGFR-mutant NSCLC (exon 19 deletion or L858R).
Evidence (gefitinib):
Evidence (erlotinib):
Evidence (afatinib):
Evidence (alectinib):
Evidence (crizotinib):
Evidence (ceritinib):
Evidence (brigatinib):
Evidence (lorlatinib):
ROS1 rearrangements occur in approximately 1% of patients with NSCLC.
Crizotinib was approved for patients with metastatic NSCLC whose tumors are ROS1-positive, regardless of the number of previous systemic therapies.
Evidence (crizotinib):
BRAF V600E mutations occur in 1% to 2% of lung adenocarcinomas.
Evidence (dabrafenib and trametinib):
The combination of dabrafenib and trametinib received approval in the treatment of patients with NSCLC whose tumors harbor BRAF V600E mutations as detected by an FDA-approved test.
Somatic gene fusions in NTRK occur across a range of solid tumors including in fewer than 0.5% of NSCLC tumors.
These fusions appear to occur more frequently in nonsmokers with lung adenocarcinoma.
Evidence (larotrectinib):
The FDA has approved larotrectinib for the treatment of patients who have locally advanced or metastatic tumors that harbor an NTRK gene fusion without a known acquired resistance mutation, and who have no satisfactory alternative treatments or whose cancer has progressed following treatment.
Pembrolizumab is a humanized monoclonal antibody that inhibits the interaction between the PD-1 coinhibitory immune checkpoint expressed on tumor cells and infiltrating immune cells and its ligands, PD-L1 and programmed cell death-ligand 2 (PD-L2).
Evidence (pembrolizumab plus chemotherapy):
Evidence (pembrolizumab alone):
Pembrolizumab in combination with pemetrexed and carboplatin received FDA approval as first-line treatment of patients with metastatic nonsquamous NSCLC, regardless of PD-L1 expression. Pembrolizumab also received approval as a first-line monotherapy for patients with NSCLC whose tumors express PD-L1 (≥50% staining as determined by a test approved by the FDA). Patients with EGFR or ALK genomic tumor aberrations should have disease progression on FDA-approved therapies before receiving pembrolizumab (refer to the FDA label for pembrolizumab).
Radiation therapy may be effective in palliating symptomatic patients with local involvement of NSCLC with any of the following:
In some cases, endobronchial laser therapy and/or brachytherapy have been used to alleviate proximal obstructing lesions.
Although EBRT is frequently prescribed for symptom palliation, there is no consensus on which fractionation scheme should be used. Although different multifraction regimens appear to provide similar symptom relief,
single-fraction radiation may be insufficient for symptom relief compared with hypofractionated or standard regimens, as evidenced in the NCT00003685 trial.
[Level of evidence: 1iiC] Evidence of a modest increase in survival in patients with a better PS given high-dose radiation therapy is available.
[Level of evidence: 1iiA] In closely observed asymptomatic patients, treatment may often be appropriately deferred until symptoms or signs of a progressive tumor develop.
Evidence (radiation therapy):
A solitary pulmonary metastasis from an initially resected bronchogenic carcinoma is unusual. The lung is frequently the site of second primary malignancies in patients with primary lung cancers. Whether the new lesion is a new primary cancer or a metastasis may be difficult to determine. Studies have indicated that in most patients the new lesion is a second primary tumor, and after its resection, some patients may achieve long-term survival. Thus, if the first primary tumor has been controlled, the second primary tumor should be resected, if possible.
Patients who present with a solitary cerebral metastasis after resection of a primary NSCLC lesion and who have no evidence of extracranial tumor can achieve prolonged disease-free survival with surgical excision of the brain metastasis and postoperative whole-brain radiation therapy.
Unresectable brain metastases in this setting may be treated with stereotactic radiosurgery.
Approximately 50% of patients treated with resection and postoperative radiation therapy will develop recurrence in the brain; some of these patients will be suitable for additional treatment.
In those selected patients with good PS and without progressive metastases outside of the brain, treatment options include reoperation or stereotactic radiation surgery.
For most patients, additional radiation therapy can be considered; however, the palliative benefit of this treatment is limited.
[Level of evidence: 3iiiDiii]
Treatment options under clinical evaluation for newly diagnosed stage IV, recurrent, and relapsed NSCLC (first-line therapy) include the following:
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.
进展的复发性IV期非小细胞肺癌(NSCLC)患者的标准治疗选择(二线治疗及以上)包括:
对于转移性病灶患者,化疗使用产生了客观缓解并且生存率略有提高。
[证据等级:[1iiA]在检查对症反应的研究中显示,主观症状改善的发生率高于客观缓解。
体能状态(PS)良好且有症状性复发的知情患者可采用含铂类药物化疗方案缓解症状。对于含铂类药物化疗后复发的患者,可以考虑二线治疗。
证据(多西他赛):
证据(多西他赛联合雷莫芦单抗):
证据(培美曲塞):
证据(厄洛替尼):
证据(吉非替尼):
厄洛替尼和吉非替尼治疗后从未吸烟的东亚女性患者以及腺癌和支气管肺泡癌患者的ORR较高。
缓解可能与EGFR酪氨酸激酶结构域中的致敏突变有关,
但与KRAS突变无关。
[证据等级:3iiiDiii]对于通过免疫组化检测EGFR蛋白表达水平或通过荧光原位杂交研究获得的EGFR基因拷贝数增加的患者,其生存期获益可能更大,
但是通过免疫组化进行EGFR检测的临床实用性受到质疑。
证据(阿法替尼):
证据(奥希替尼):
证据(克唑替尼):
证据(塞瑞替尼):
证据(阿来替尼):
证据(布加替尼):
大约1%的NSCLC患者出现ROS1重排。
克唑替尼已获批用于治疗肿瘤呈ROS1阳性的转移性NSCLC患者,不考虑既往全身治疗的次数。
证据(克唑替尼):
BRAF V600E突变,见于1%到2%的肺腺癌。
证据(达拉非尼和曲美替尼):
达拉非尼和曲美替尼联合治疗已获批用于治疗肿瘤携带BRAF V600E突变的NSCLC患者(通过FDA批准的试验检测)。
纳武利尤单抗是一种完全人单克隆抗体,可抑制在肿瘤细胞和浸润性免疫细胞上表达的程序性死亡1(PD-1)共抑制免疫检查点。
帕博利珠单抗是一种人源化单克隆抗体,可抑制肿瘤细胞上表达的PD-1共抑制性免疫检查点与浸润免疫细胞及其配体PD-L1和PD-L2之间的相互作用。
阿替利珠单抗是一种阻断PD-L1的抗体。
证据(纳武利尤单抗):
目前对于在一线含铂类药物化疗期间或之后出现疾病进展的转移性NSCLC患者来说,纳武利尤单抗是标准二线治疗。与多西他赛相比,纳武利尤单抗治疗后生存期延长,毒性反应发生率降低。但是,迄今为止,纳武利尤单抗的临床试验尚未招募有自身免疫疾病、间质性肺病病史或ECOG PS >1的患者。自身免疫疾病活跃的患者不能用纳武利尤单抗治疗。需要密切监测所有患者治疗中出现的自身免疫毒性反应。用于治疗自身免疫毒性反应的特定算法纳入纳武利尤单抗的FDA标签中。
证据(帕博利珠单抗):
帕博利珠单抗已获得加速批准,作为NSCLC患者的二线治疗,其中该患者的肿瘤在一线化疗或一线化疗后出现PD-L1表达(经FDA批准的试验确定染色率>50%)。在接受帕博利珠单抗之前,EGFR或ALK基因组肿瘤异常的患者应先行FDA批准的治疗确定疾病进展(参见FDA的帕博利珠单抗标签)。
证据(阿替利珠单抗):
处于临床评估阶段的进展的复发性IV期NSCLC治疗选择(二线治疗)包括:
利用我们先进的临床试验检索工具查找NCI支持的癌症临床试验(当前正在招募患者)。可通过试验地点、治疗类型、药物名称和其他标准缩小检索范围。还可获得关于临床试验的基本信息。
Standard treatment options for patients with progressive stage IV, relapsed, and recurrent non-small cell lung cancer (NSCLC) (second-line therapy and beyond) include the following:
The use of chemotherapy has produced objective responses and small improvement in survival for patients with metastatic disease.
[Level of evidence: 1iiA] In studies that have examined symptomatic response, improvement in subjective symptoms has been reported to occur more frequently than objective response.
Informed patients with good performance status (PS) and symptomatic recurrence can be offered treatment with a platinum-based chemotherapy regimen for palliation of symptoms. For patients who have relapsed after platinum-based chemotherapy, second-line therapy can be considered.
Evidence (docetaxel):
Evidence (docetaxel plus ramucirumab):
Evidence (pemetrexed):
Evidence (erlotinib):
Evidence (gefitinib):
ORR to erlotinib and gefitinib are higher in patients who have never smoked, in females, in East Asians, and in patients with adenocarcinoma and bronchioloalveolar carcinoma.
Responses may be associated with sensitizing mutations in the tyrosine kinase domain of the EGFR-
and, with the absence of, KRAS mutations.
[Level of evidence: 3iiiDiii] Survival benefit may be greater in patients with EGFR protein expression by immunohistochemistry or increased EGFR gene copy number by fluorescence in situ hybridization studies,
but the clinical utility of EGFR testing by immunohistochemistry has been questioned.
Evidence (afatinib):
Evidence (osimertinib):
Evidence (crizotinib):
Evidence (ceritinib):
Evidence (alectinib):
Evidence (brigatinib):
ROS1 rearrangements occur in approximately 1% of patients with NSCLC.
Crizotinib was approved for patients with metastatic NSCLC whose tumors are ROS1-positive, regardless of the number of previous systemic therapies.
Evidence (crizotinib):
BRAF V600E mutations occur in 1% to 2% of lung adenocarcinomas.
Evidence (dabrafenib and trametinib):
The combination of dabrafenib and trametinib received approval for patients with NSCLC whose tumors harbor BRAF V600E mutations as detected by an FDA-approved test.
Nivolumab is a fully human monoclonal antibody that inhibits the programmed death-1 (PD-1) co-inhibitory immune checkpoint expressed on tumor cells and infiltrating immune cells.
Pembrolizumab is a humanized monoclonal antibody that inhibits the interaction between the PD-1 co-inhibitory immune checkpoint expressed on tumor cells and infiltrating immune cells and its ligands, PD-L1 and PD-L2.
Atezolizumab is a PD-L1–blocking antibody.
Evidence (nivolumab):
Nivolumab is now considered a standard second-line therapy for patients with metastatic NSCLC with progression on or after first-line platinum-based chemotherapy and is associated with improved survival and lower rates of toxicity than docetaxel. However, clinical trials of nivolumab to date have not enrolled patients with a history of autoimmune disease, interstitial lung disease, or an ECOG PS higher than 1. Patients with active autoimmune conditions cannot be treated with nivolumab. Closely monitoring all patients for autoimmune toxicities from treatment is required. Specific algorithms for the management of autoimmune toxicity are included in the FDA label for nivolumab.
Evidence (pembrolizumab):
Pembrolizumab received accelerated approval as a second-line therapy for patients with NSCLC whose tumors express PD-L1 (>50% staining as determined by an FDA-approved test) with progression on or after first-line chemotherapy. Patients with EGFR or ALK genomic tumor aberrations should have disease progression on FDA-approved therapies before receiving pembrolizumab (refer to the FDA label for pembrolizumab).
Evidence (atezolizumab):
Treatment options under clinical evaluation for progressive stage IV, relapsed, and recurrent NSCLC (second-line therapy) include the following:
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
定期审查PDQ癌症信息总结内容,并在获得最新信息后进行更新。 本章节描述了截至上述日期对本总结所做的最新变更。
更新了2020年估计的新发病例和死亡病例的统计数据(引用美国癌症协会的数据作为参考文献1)。
本总结由PDQ成人治疗编辑委员会撰写并维护,其编辑内容独立于NCI。 总结反映了独立审查文献,不代表NCI或NIH的政策声明。 如需了解更多关于总结政策和PDQ编辑委员会在维护PDQ总结内容中作用的信息,请参见有关“本PDQ总结”和“PDQ®-NCI综合癌症数据库”页。
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.
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the treatment of non-small cell lung cancer. It is intended as a resource to inform and assist clinicians who care for cancer patients. It does not provide formal guidelines or recommendations for making health care decisions.
This summary is reviewed regularly and updated as necessary by the PDQ Adult Treatment Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
The lead reviewers for Non-Small Cell Lung Cancer Treatment are:
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website's Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Adult Treatment Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Adult Treatment Editorial Board. PDQ Non-Small Cell Lung Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
Based on the strength of the available evidence, treatment options may be described as either “standard” or “under clinical evaluation.” These classifications should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the treatment of non-small cell lung cancer. It is intended as a resource to inform and assist clinicians who care for cancer patients. It does not provide formal guidelines or recommendations for making health care decisions.
This summary is reviewed regularly and updated as necessary by the PDQ Adult Treatment Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
The lead reviewers for Non-Small Cell Lung Cancer Treatment are:
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website's Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Adult Treatment Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Adult Treatment Editorial Board. PDQ Non-Small Cell Lung Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
Based on the strength of the available evidence, treatment options may be described as either “standard” or “under clinical evaluation.” These classifications should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.