Advances in Clinical Medicine
Vol. 13  No. 10 ( 2023 ), Article ID: 73434 , 10 pages
10.12677/ACM.2023.13102187

肺肉瘤样癌的诊断和治疗研究进展

梁亚男,冯龄鑫,綦琦,杨雪,王静*

青岛大学附属医院肿瘤科,山东 青岛

收稿日期:2023年9月6日;录用日期:2023年10月1日;发布日期:2023年10月9日

摘要

肺肉瘤样癌(pulmonary sarcomatoid carcinoma, PSC)是属于一种含有肉瘤或肉瘤样分化成分的非小细胞肺癌,分化差且具有高度侵袭性。发病率在非小细胞肺癌(non-small cell lung carcinoma, NSCLC)中占2%~3%。肺肉瘤样癌的准确诊断是一大难题,主要依据组织病理学和免疫组织化学。治疗方面尚未有明确的诊疗方案,主要以综合治疗为主。其对传统化学治疗和放射治疗反应性差,早期手术几率低。目前免疫和靶向治疗是提高PSC治疗的突破点。

关键词

肺,肉瘤样癌,诊断,免疫治疗,靶向治疗,预后

Advances in Diagnosis and Treatment of Pulmonary Sarcomatoid Carcinoma

Yanan Liang, Lingxin Feng, Qi Qi, Xue Yang, Jing Wang*

Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Shandong

Received: Sep. 6th, 2023; accepted: Oct. 1st, 2023; published: Oct. 9th, 2023

ABSTRACT

Pulmonary sarcomatoid carcinoma (PSC) is a kind of non-small cell lung cancer with sarcoma or sarcomatoid differentiation components, which is poorly differentiated and highly aggressive. The incidence rate accounts for 2%~3% of the total number of non-small cell lung cancer (NSCLC). The accurate diagnosis of pulmonary sarcomatoid carcinoma is a difficult problem, which mainly depends on histopathology and immunohistochemistry. In terms of treatment, there is no clear diagnosis and treatment plan, mainly comprehensive treatment. Its response to traditional chemotherapy and radiotherapy is poor, and the probability of early surgery is low. At present, immunotherapy and targeted therapy are the hotspots of PSC treatment.

Keywords:Lung, Sarcomatoid Carcinoma, Diagnosis, Immunotherapy, Targeted Therapy, Prognosis

Copyright © 2023 by author(s) and Hans Publishers Inc.

This work is licensed under the Creative Commons Attribution International License (CC BY 4.0).

http://creativecommons.org/licenses/by/4.0/

1. 前言

肺肉瘤样癌(Pulmonary sarcomatoid carcinoma, PSC)是一种侵袭性的非小细胞肺癌,小标本诊断性能差,预后较差,约占所有肺部恶性肿瘤的0.1%~0.4%。患者多在疾病晚期治疗,手术机会少,且对放化疗不敏感,难以延长患者的生存时间。研究发现,程序性死亡配体-1 (PD-L1)在PSC中过表达,表达率高达100%,且与免疫治疗效果呈正相关。此外,PSC中还存在不同的突变基因,如表皮生长因子受体(EGFR)、KRAS、间充质上皮转化因子(MET)等。EGFR的突变率一般为8.8%~25%,KRAS在PSC中突变率可高达38%。EGFR/KRAS是最常见的共突变,约占NSCLC的30%,其中针对PSC中MET突变药物赛沃替尼治疗效果最显著。肺肉瘤样癌诊断和治疗尚未明确,现综述如下。

2. 肺肉瘤样癌的诊断进展

根据2021版胸部肿瘤WHO分类 [1] ,将肉瘤样癌分为多形性癌(Pleomorphic Carcinoma, PC)、肺母细胞瘤(Pulmonary Blastoma Carcinoma, PBC)、癌肉瘤(Carcinosarcoma, CS)、巨细胞癌(Giant Cell Carcinoma, GCC)及梭形细胞癌(Spindle Cell Carcinoma, SCC)。PC是一种由至少10%的梭形细胞和巨细胞组成的癌;SCC是一种由上皮梭形细胞组成的癌;GCC是一种单纯由巨细胞组成的癌;CS是一种NSCLC和真肉瘤的混合体;PBC由原始上皮成分和间叶成分组成。其中,多形性癌在肺肉瘤样癌中最多见 [2] 。

肺肉瘤样癌的精确诊断仍是临床上的难题,需要综合临床特点、影像学检查及病理诊断。该病诊断难点有三个方面。

第一、肺肉瘤样癌的发病率占原发性肺癌的0.1%~0.4%,对该疾病的认识程度不够,易误诊为其他类型肺癌。

第二、活检取得的组织量少,细胞病理学难以通过活检取得的标本量诊断清楚。Sun等人 [3] 通过分析55例肺肉瘤样癌患者的特征发现,8例(共16例,阳性率为50%)通过纤维支气管镜检查确诊。26例(共34例,阳性率76.47%)通过CT引导下肺穿刺活检确诊,其余21例通过手术切除病灶后行病理检查确诊。由此可见,穿刺活检难以精确诊断PSC。2015年世界卫生组织(WHO)肺肿瘤病理分类指出活检或细胞学不能诊断SCC、GCC和PC,也不足以诊断PBC和CS。患者就诊时疾病多处于晚期,此时选择手术的可能性小,无法通过手术获得足够标本,给诊断带来困难。所以发明获取更多诊断所需标本量的方式是提高病理诊断精确度的重要途径。

第三、临床特征及影像学表现很难将PSC与其余NSCLC精确区分,只能辅助诊断。发病人群上,PSC多见于60岁左右有吸烟史的男性 [4] [5] ,NSCLC多见于65~84岁有吸烟史的男性 [6] ,临床表现上,PSC患者就诊时咳嗽、乏力、胸痛、呼吸困难、体重下降多见,NSCLC患者的咳嗽、咳痰、呼吸困难常见。影像学表现上,PSC的病灶多单发于肺上叶,外周型较中心型多见;直径多为40~180 mm,有的可达到210 mm,胸膜浸润、胸腔积液及纵隔淋巴结肿大较常见,胸部平扫CT上密度多均匀,边缘大多光滑,增强CT上密度多不均匀,肿块周围呈不规则环状或斑块状高密度影 [7] - [13] 。此外,PSC在PET-CT (氟-18氟脱氧葡萄糖正电子发射断层扫描/计算机断层扫描)上通常表现为高摄取,并有报道发现SUVmax > 19.85可以作为PSC与其他类型的NSCLC的鉴别点 [14] 。

鉴别PSC与其他肺部肿瘤需根据细胞形态、生物标记物、影像学等因素综合分析。根据细胞学形态进行诊断时,PSC需与原发或转移性单相型(梭形细胞构成)和双相型(上皮样细胞和梭形细胞构成)肉瘤(即滑膜肉瘤、恶性孤立性纤维瘤、炎性肌纤维母细胞)、肉瘤样间皮瘤、梭形细胞黑色素瘤、肉瘤样胸腺癌、绒毛膜癌等鉴别 [15] 。在对45例胸膜SM和27例LSC进行免疫组织化学分析中,生物标记物D2-40在胸膜间皮瘤中的阳性表达(86.7%)显著高于LSC (25.9%),可以将肺肉瘤样癌与胸膜肉瘤样间皮瘤区别开 [16] 。根据影像学鉴别诊断时,CT表现为肺部单发肿块,应与肺鳞癌、胸膜孤立性纤维瘤、肺炎性假瘤相鉴别 [17] 。位于双肺上叶的PSC应与增殖性肺结核鉴别 [18] 。

3. 肺肉瘤样癌的治疗进展

该疾病初诊时多为晚期,故治疗多采用非手术为主的综合治疗。对于早期肿瘤,首选根治性手术切除,但其恶性程度使得根治术后仍有较高的复发率及远处转移率。目前研究发现PSC对化疗、放疗均不敏感。近年来兴起的靶向治疗和免疫治疗给肺肉瘤样癌患者带来希望。

3.1. 肺肉瘤样癌的手术治疗

早期PSC首选手术治疗,手术原则同非小细胞肺癌,但PSC患者的术后生存期较NSCLC更短 [19] ,依据TNM分期及美国东部肿瘤协作组(Eastern Cooperative Oncology Group, ECOG)评分,手术方式可选择肺叶切除术、肺段切除术,全肺切除术,全肺切除的患者5年生存率最高 [20] 。

3.2. 肺肉瘤样癌的化学治疗

PSC的化疗类型分为四种,分别为新辅助化疗、围术期化疗、术后辅助化疗、姑息性化疗。对于早期PSC,杰米 [21] 等人评估了接受R0肺癌切除术的4675名PSC患者围术期化疗的效果,发现化疗患者的DFS中位数为34个月,而未化疗患者的DFS中位数为12个月,可见围术期化疗可以带来明显的生存获益;而阿卜杜拉 [22] 等人通过查询2004年~2015年国家癌症据库,统计接受手术根治切除患者的5年生存率,发现接受根治性手术切除的II和III期患者生存率与早期PSC的生存率相似。新辅助化疗和辅助化疗都不能提高早期疾病患者的生存率 [23] 。孙良东等人 [24] 通过收集SEER数据库和上海市肺科医院中做过手术的PSC患者发现,对于无远处转移、体重指数正常或较高(≥18.5)、血红蛋白正常、肿瘤体积较小(≤4 cm)的患者,接受完全切除后的总生存期明显较好(P < 0.05) [22] [24] 。

晚期PSC一线化疗易耐药,且预后差。蒂博·维埃拉等人 [25] 收集了97例接受一线化疗的PSC病人,其中位PFS为4.3月,中位OS为8.9月;而有的研究 [26] 显示应用含铂方案对术后复发及IV期肺肉瘤样癌患者与NSCLC的疗效相似。

3.3. 肺肉瘤样癌的抗血管治疗

实体肿瘤的生长、增殖和转移离不开新生血管的生成。血管浸润是一个肺肉瘤样癌预后不良的因素 [27] ,抗血管生成药物可以抑制肿瘤新生血管生成,阻止肿瘤的血管浸润,进而抑制肿瘤生长 [28] [29] 。在NSCLC中,抗血管生成药物有着明确的疗效。在PSC中,HOT1201/NEJ024研究 [30] 通过对比应用贝伐珠单抗联合或单用紫杉醇/卡铂的PSC病人,发现应用贝伐珠单抗的病人中位PFS和MST均长于化疗组,认为贝伐珠单抗或许可以作为PSC一线治疗药物。此外,李等人 [31] 收集了2016年至2019年共21例使用阿帕替尼(250~425 mg/d)治疗III~IV期及术后复发的肺肉瘤样癌患者,14.3% (3例)达到了部分缓解,33.3% (7例)达到了疾病稳定状态,52.4% (11例)疾病出现进展。盐酸安罗替尼(AL3818)是一种新型口服多靶点酪氨酸激酶抑制剂,于2018年5月9日被中国食品药品监督管理局(CFDA)批准为难治性晚期NSCLC的三线治疗药物 [32] ,徐等人 [33] 报道1例口服安罗替尼(12 mg/d,第1~14天,21 d/周期)治疗的晚期PSC患者,3周期治疗后疗效评估为PR。Li等人 [34] 报道1例术后复发的肺肉瘤样癌患者,复发后接受了6个周期安罗替尼联合达卡巴嗪和顺铂化疗,疾病就达到完全缓解(CR),继续给予安罗替尼维持治疗,无进展生存期达到两年多,可见安罗替尼在PSC中具有很好的疗效。然而,抗血管生成药物并不总是有效,张等人 [31] 报道了1例使用阿帕替尼治疗肺巨细胞癌的病例,在4周期化疗出现疾病进展后,改用口服阿帕替尼(500 mg每晚)治疗,1个月后疾病依旧进展。所以,抗血管生成药物在PSC中的应用需继续观察及验证。

3.4. 肺肉瘤样癌的靶向治疗

PSC肿瘤组织中存在多个癌基因和抑癌基因的异常,例如TP53、EGFR、KRAS、ALK和MET以及PSC独有的POT1突变 [35] ,还有SMARCA4、MLL4、NF1、NOTCH4、TERT等基因突变 [36] 。突变抑癌基因以TP53基因最常见,突变癌基因以EGFR、KRAS和MET突变最常见 [37] [38] 。这些基因改变可以单独存在也可同时发生,并伴有较高的肿瘤突变负荷(tumor mutational burden, TMB) [38] 。

TP53基因最常见的突变为错义突变,PSC基因突变频率的74% [39] ,突变通常聚集在外显子5-8中。p53蛋白突变导致无限性细胞增殖 [40] 。PSC对TP53抑制剂敏感 [41] ,而美国FDA尚未批准任何针对TP53的靶向药物,TP53基因突变与肺肉瘤样癌的不良预后有关 [42] [43] 。

EGFR基因的突变频率一般在8.8%~23.8% [39] [44] [45] [46] ,突变类型主要为19号外显子缺失、21号外显子L858R突变、20号外显子插入等 [39] 。表皮生长因子受体–酪氨酸激酶抑制剂(epidermal growth factor receptor-tyrosine kinase inhibitor, EGFR-TKI)类药物是靶向EGFR突变位点的治疗药物。关于EGFR-TKI类药物在PSC中只有个案报道,邹方文等人 [47] 报道了1例EGFR野生型的PSC患者,他首先接受了放疗(60 Gy)和4个21天周期的化疗(1600 mg吉西他滨,第1天和第8天;30 mg,顺铂,第1~3天)后,在主动脉旁淋巴结,双侧髂窝和右臀部区域发现了转移病灶,穿刺右臀转移灶发现有EGFR外显子21 L858R基因突变,于是服用厄洛替尼治疗,达到了6个月的无进展生存期。

KRAS是肺肉瘤样癌的常见突变基因,与吸烟密切相关,是肺肉瘤样癌患者预后不良的标志 [48] ,KRAS G12C为最常见的突变类型 [49] 。针对KRAS基因突变的药物仅有一种,即已经上市的靶向药物AMG510,被美国FDA批准用于既往接受至少一次系统治疗的携带KRAS G12C突变的局部晚期或转移性NSCLC成年患者 [50] 。但该靶向药未见在PSC中使用,KRAS也存在很多共突变 [51] ,例如,在KRAS突变晚期NSCLC患者中,KRAS和KEAP1/NFE2L2的共突变可预测较短的生存期、对初始铂类化疗的反应持续时间以及免疫治疗开始后的生存期 [52] 。

MET信号的激活通常与上皮细胞到间充质细胞的转化(EMT)有关,并在功能上支持EMT [53] ,突变包括MET 14外显子跳跃突变、MET扩增和MET过表达三种类型,MET 14外显子跳跃突变最常见,在PSC中的突变率为31.8%,其靶向药物是赛沃替尼 [54] [55] ,可达到36个月的无进展生存期 [56] ,此外,具有MET激活改变(包括MET位点扩增)的NSCLC还对靶向MET活性的卡博替尼、卡马替尼表现出显著反应 [57] ,王等人 [44] 报道了1例EGFR和MET同时存在的PSC病例,使用吉非替尼和克唑替尼分别靶向突变位点,病灶达到部分缓解,停药9.7月后,病灶仍处于稳定状态。多靶点突变获得的治疗可能会获得更长生存时间。

目前被FDA批准的ALK抑制剂有克唑替尼、塞瑞替尼等,ALK基因重排可选择ALK抑制剂治疗。陈等人 [58] 通过141例PSC病例统计中国人群中PSC中ALK重排的发生率为3.5%,年轻且从不吸烟者的PSC患者更常携带ALK重排。费德里卡·德安东尼奥等人 [59] 报道了1例既有ALK重排又有PD-L1高表达患者,帕博利珠单抗耐药后依次使用克唑替尼、塞瑞替尼、布加替尼治疗,ALK抑制剂的使用使总生存期共延长了10个月。可见ALK靶向药物可以给PSC治疗带来显著疗效。朱塞佩·佩洛西等人 [60] 通过荧光原位杂交(FISH)评估了98个PSCs的MET和ALK突变状态,并通过免疫组织化学分析评估相关蛋白表达,同时利用磷酸化抗体,发现在PSC患者亚群中,ALK和MET似乎是下游信号的协同、非随机共激活因子,即二者为联合驱动基因,共同驱动癌症的发生发展。

3.5. 肺肉瘤样癌的免疫治疗

近年来,免疫治疗逐渐成为是PSC患者治疗的主要方式。PD-L1/PD-1 抗体属于免疫检查点抑制剂,免疫治疗的作用机制是PD-1作为一种负性共刺激分子,恢复正常T细胞对肿瘤细胞的识别和杀伤能力,从而杀灭肿瘤细胞,延长患者的无进展生存期和总生存期 [61] [62] [63] 。PSC中PD-L1表达率高于NSCLC [64] ,阳性率可高达90% [65] ,免疫药物分两类,一类是PD-1抑制剂,例如纳武单抗、帕博利珠单抗等,另一类是PD-L1抑制剂,例如阿维单抗、度伐利尤单抗、阿替利珠单抗 [14] 。

罗塞尔等人 [61] 报道了2例利用单药纳武单抗治疗的肺转移性肉瘤样癌,均达到肿瘤体积的显著缩小。PSC除了单用免疫药物治疗,也可联合化疗 [66] ,孔等人 [65] 报道1例一线卡瑞利珠单抗联合阿霉素和顺铂,随后在联合治疗期间由于4级白细胞减少和血小板减少而停用化疗,只进行卡瑞利珠单抗单药治疗,疾病持续缓解且维持了20多个月。东布利德斯等人 [67] 通过分析39名应用免疫治疗的PSC患者,94.9%的病人首先接受了以铂为基础的化疗,5.1%、51.3%和43.6%的免疫治疗分别用在一线、二线和三线治疗中,这些免疫治疗药物87.2%用的是纳武单抗,7.7%的帕博利珠单抗、5.1%的阿替利珠单抗。治疗时间中位数为4.5个月。总体反应率(ORR)为38.5%,疾病得到控制发生率(DCR) 61.6%。中位PFS为4.59个月,1年PFS率为10.3% (4/39),中位生存期为20个月。还对18例肿瘤的PD-L1状态进行了评估,在PDL1+的PSC患者中,ORR为53.3% (8/15),DCR为66.7% (10/15)。

PSC免疫治疗也可联合放疗,焦玉岩等人 [68] 报道1例PD-L1过表达的PSC患者,5周期特瑞普利单抗治疗后肿块缩小达到PR,后来特瑞普利单抗耐药疾病进展,将放疗加入,疾病再次得到缓解。阿沃巴霍等人 [69] 报道了1例肺梭形细胞癌患者,首先对原发肿瘤进行了立体定向体部放射治疗(SBRT),结束放疗2个月后,远处转移病灶迅速进展,但胸部CT检查显示右上叶原发性疾病的局部控制良好。患者继续使用帕博利珠单抗进行全身治疗,原发及转移部位病灶持续缩小,效果良好。

综上,单用免疫治疗可以使病灶得到明显缓解,PSC一线常规放化疗进展后,免疫治疗的加入也可以给治疗带来转机,放化疗可以增强免疫治疗的疗效,该增强效果可能与以下几个机制有关,例如化疗使免疫原性细胞死亡、使癌细胞抗原性增强、使免疫抑制细胞消耗,以及放疗可以诱发免疫原性细胞死亡、降低细胞表面的CD47表达从而增强癌细胞的摄取和抗原呈递、修饰蛋白质和DNA等大分子而增加抗原性等。如何选用最佳的免疫药物,这需要更大的人群研究。免疫治疗的最优选择以及PSC复杂的异质性使之需要更准确的预测治疗效果的生物标志物。

有研究表明PD-L1和TMB以及JMJD3蛋白水平是预测PSC免疫治疗疗效的生物标志物,TMB和PD-L1表达高者对免疫应答更好 [70] [71] ,但也有病例报道PD-L1低表达联合化疗疗效也可达到PR状态,联合治疗或许是通过增加免疫原性使病灶减小 [72] 。另有研究表明PD-L1表达与总的TMB和KRAS突变状态呈正相关 [73] 。发现在肺肉瘤样癌中,PD-L1与CD47共表达与更差的预后相关 [74] 。

3.6. 肺肉瘤样癌的综合治疗

单用一种治疗并不能将疾病OS延长到最长时间,对任何肿瘤来说都不是最佳的方案,肿瘤的治疗在于首先确定肿瘤的早中晚期,根据肿瘤的具体情况,将手术、放化疗与免疫、靶向治疗等不同的治疗方式相结合,最大限度的提高治疗肿瘤的疗效。萨拉蒂等人 [75] 报道1例手术联合化学治疗及免疫治疗的晚期PSC患者,他首先进行了姑息性手术,将原发病灶切除,术后接受了一线和二线化疗,但是化疗对残存病灶并无明显效果,所以将化疗改为纳武单抗,持续治疗了22个月,最后发现脑部病变消失,其他转移部位病灶也逐渐缩小。罗塞尔等人 [61] 报道了2例肺肉瘤癌患者。一位是cT4患者,他首先使用4周期顺铂联合吉西他滨新辅助化疗,肿瘤获得了部分缓解,然后进行了肺叶切除术,4周后疾病进展,改为纳武单抗免疫治疗,6个月免疫治疗后,病灶达到了完全缓解。另一位患者首先接受了根治性肺切除术,然后进行2周期长春瑞滨联合顺铂化疗,后疾病进展,使用8个月纳武单抗治疗,期间病灶持续缓解。西野和美等人 [76] 报道了1例免疫治疗联合放化疗的PSC病例,患者的基因检测结果显示无可靶向治疗的突变,所以他首先接受了2周期长春瑞滨和顺铂化疗以及60 Gy的放疗,放化疗使病灶达到PR反应,第三周期改用度伐利尤单抗治疗,7周期免疫治疗后疾病出现进展,将度伐利尤单抗换为帕博利珠单抗继续治疗,7个月后,肿块获得完全缓解。可见,肿瘤综合治疗表现出良好的控制疾病的能力,甚至可以使疾病达到完全缓解。目前单一治疗中各种药物及方式对该病的疗效尚不完全明确,我们应该根据最新进展,将不同的治疗方式结合起来,制定出更完善的综合治疗方案治疗该病。

肺肉瘤样癌侵袭性强,无论是诊断还是治疗都面临着困难,在诊断方面,提高对该疾病的重视,解决因穿刺组织少而降低诊断准确率的方法,降低漏诊率。在治疗方面,对PSC来说,以免疫治疗为主的综合治疗更为重要,解决治疗方案如何搭配的问题,是PSC得到有效治疗的必经之路。

基金项目

山东省医学会临床科研资金YXH2022ZX02020一线瑞菲乐联合安可达治疗EGFR非经典突变晚期非小细胞肺癌患者单中心单臂临床研究。

文章引用

梁亚男,冯龄鑫,綦 琦,杨 雪,王 静. 肺肉瘤样癌的诊断和治疗研究进展
Advances in Diagnosis and Treatment of Pulmonary Sarcomatoid Carcinoma[J]. 临床医学进展, 2023, 13(10): 15635-15644. https://doi.org/10.12677/ACM.2023.13102187

参考文献

  1. 1. 方三高, 陈真伟, 魏建国. 2021年第5版WHO胸部肿瘤分类[J]. 诊断病理学杂志, 2021, 28(7): 591-593+607.

  2. 2. Baldovini, C., Rossi, G. and Ciarrocchi, A. (2019) Approaches to Tumor Classification in Pulmo-nary Sarcomatoid Carcinoma. Lung Cancer (Auckl), 10, 131-149. https://doi.org/10.2147/LCTT.S186779

  3. 3. Sun, J.C., Jiang, Z.Y., Shan, T.Y., Yang, R.N., Kong, D.J., Rui, J.S., Li, X.Y., Kong, G.Q. and Chang, B.P. (2022) Charac-teristics and Prognostic Analysis of 55 Patients with Pulmonary Sarcomatoid Carcinoma. Frontiers in Oncology, 12, Ar-ticle ID: 833486. https://doi.org/10.3389/fonc.2022.833486

  4. 4. Hou, J.N., Xing, L.H. and Yuan, Y.F. (2018) A Clinical Analysis of 114 Cases of Sarcomatoid Carcinoma of the Lung. Clinical Experimental Medical, 18, 555-562. https://doi.org/10.1007/s10238-018-0517-2

  5. 5. Liu, X., Wang, F., Chen, X., Hou, X., Li, Q., Xie, Z., Liu, Y., et al. (2021) P35.01 Genomic Origin and Immune-Related Status of Pulmonary Sarcomatoid Carcinoma. Journal of Tho-racic Oncology, 16, S419. https://doi.org/10.1016/j.jtho.2021.01.702

  6. 6. Duma, N., Santana-Davila, R. and Molina, J.R. (2019) Non-Small Cell Lung Cancer: Epidemiology, Screening, Diagnosis, and Treatment. Mayo Clinic Proceedings, 94, 1623-1640. https://doi.org/10.1016/j.mayocp.2019.01.013

  7. 7. Yoshino, N., Kubokura, H., Yamauchi, S., Ohaki, Y., Koizumi, K. and Shimizu, K. (2009) A True Pulmonary Carcinosarcoma That Required Diagnostic Differentiation from a Pleo-morphic Adenoma: A Case Report. Annals of Thoracic and Cardiovascular Surgery, 15, 42-45.

  8. 8. Ung, M., Rouquette, I., Filleron, T., Taillandy, K., Brouchet, L., Bennouna, J., et al. (2016) Characteristics and Clinical Outcomes of Sarcomatoid Carcinoma of the Lung. Clinical Lung Cancer, 17, 391-397. https://doi.org/10.1016/j.cllc.2016.03.001

  9. 9. Xu, X.-L., Song, W., Sui, X., Song, L., Wang, X., Feng, R.-E. and Li, Y. (2016) Computed Tomographic and Pathological Features of Primary Pulmonary Sarcomatoid Carcinoma. Acta Academiae Medicinae Sinicae, 38, 93-98.

  10. 10. 张新东, 张谷青, 陈月芹. 肺肉瘤样癌的CT诊断及误诊分析[J]. 医学影像学杂志, 2014, 24(4): 531-533+552.

  11. 11. Rapicetta, C., Lococo, F., Stefani, A., Rossi, G., Ricchetti, T., Filice, A., et al. (2016) Primary Sarcomatoid Carcinoma of the Lung: Radiometabolic ((18)F-FDG PET/CT) Findings and Cor-relation with Clinico-Pathological and Survival Results. Lung, 194, 653-657. https://doi.org/10.1007/s00408-016-9904-1

  12. 12. 王雷, 李向平, 孙健, 李超, 张海滨. 肺癌肉瘤11例临床分析[J]. 临床荟萃, 2010, 25(23): 2080-2082.

  13. 13. Vohra, A. and Narula, H. (2014) Imaging Findings of Pulmonary Car-cinosarcoma: A Case Report. Lung India, 31, 164-167. https://doi.org/10.4103/0970-2113.129861

  14. 14. Xu, L., Tao, N.-N., Liang, B., Li, D.-W., Li, H.-C. and Su, L.-L. (2022) Use of PD-1 Inhibitor Tislelizumab in the Treatment of Ad-vanced Pulmonary Sarcomatoid Carcinoma: A Case Report. Thoracic Cancer, 13, 502-505. https://doi.org/10.1111/1759-7714.14290

  15. 15. Pelosi, G., Sonzogni, A., De Pas, T., Galetta, D., Veronesi, G., Spaggiari, L., et al. (2010) Review Article: Pulmonary Sarcomatoid Carcinomas: A Practical Overview. International Journal of Surgical Pathology, 18, 103-120. https://doi.org/10.1177/1066896908330049

  16. 16. Takeshima, Y., Amatya, V.J., Kushitani, K., Kaneko, M. and Inai, K. (2009) Value of Immunohistochemistry in the Differential Diagnosis of Pleural Sarcomatoid Mesothelioma from Lung Sarcomatoid Carcinoma. Histopathology, 54, 667-676. https://doi.org/10.1111/j.1365-2559.2009.03298.x

  17. 17. 吕冬亮, 张玲红, 戴丹丹, 王春, 杨汉卿. 肺肉瘤样癌11例CT表现及鉴别诊断[J]. 中国乡村医药, 2018, 25(21): 46-47.

  18. 18. 张卉, 王道清, 程留慧. 原发性肺肉瘤样癌的CT表现及鉴别诊断[J]. 中国中西医结合影像学杂志, 2019, 17(6): 625-627.

  19. 19. Rahouma, M., Kamel, M., Narula, N., Nasar, A., Harrison, S., Lee, B., et al. (2018) Pulmo-nary Sarcomatoid Carcinoma: An Analysis of a Rare Cancer from the Surveillance, Epidemiology, and End Results Da-tabase. European Association for Cardio-Thoracic Surgery, 53, 828-834. https://doi.org/10.1093/ejcts/ezx417

  20. 20. 杨文静, 张百华, 王永岗, 张宏图. 肺癌肉瘤13例临床分析[J]. 实用癌症杂志, 2012, 27(5): 506-509.

  21. 21. Chaft, J.E., Sima, C.S., Ginsberg, M.S., Huang, J., Kris, M.G., Travis, W.D. and Azzoli, C.G. (2012) Clinical Outcomes with Perioperative Chemotherapy in Sarcomatoid Carcinomas of the Lung. Journal of Thoracic Oncology, 7, 1400-1405. https://doi.org/10.1097/JTO.0b013e3182614856

  22. 22. Abdallah, H.M., Martinez-Meehan, D., Lutfi, W., Dhupar, R., Grenda, T., Schuchert, M.J., et al. (2021) Adjuvant Chemotherapy for Pulmonary Sarcomatoid Carcinoma: A Retrospec-tive Analysis of the National Cancer Database. The Journal of Thoracic and Cardiovascular Surgery, 163, 1669-1681.E3. https://doi.org/10.1016/j.jtcvs.2021.01.081

  23. 23. Lin, Y.B., Yang, H., Cai, Q.Q., Wang, D.F., Rao, H.L., Lin, S.X., et al. (2016) Characteristics and Prognostic Analysis of 69 Patients with Pulmonary Sarcomatoid Carci-noma. American Journal of Clinical Oncology, 39, 215-222. https://doi.org/10.1097/COC.0000000000000101

  24. 24. Sun, L.D., Dai, J., Chen, Y., Duan, L., He, W.X., Chen, Q.K., et al. (2020) Pulmonary Sarcomatoid Carcinoma: Experience from SEER Database and Shanghai Pulmonary Hos-pital. The Annals of Thoracic Surgery, 110, 406-413. https://doi.org/10.1016/j.athoracsur.2020.02.071

  25. 25. Vieira, T., Girard, N., Ung, M., Monnet, I., Cazes, A., Bonnette, P., et al. (2013) Efficacy of First-Line Chemotherapy in Patients with Advanced Lung Sarcomatoid Carcinoma. Journal of Thoracic Oncology, 8, 1574-1577. https://doi.org/10.1097/01.JTO.0000437008.00554.90

  26. 26. 熊伟杰, 张新星, 黄媚娟, 陈柳, 宋玲玲, 薛建新, 王永生, 卢轴. 32例晚期及术后复发肺肉瘤样癌患者的治疗及生存分析[J]. 四川大学学报(医学版), 2014, 45(2): 320-323.

  27. 27. Vieira, T., Antoine, M., Ruppert, A.-M., Fallet, V., Duruisseaux, M., Leprieur, E.G., et al. (2014) Blood Vessel Invasion Is a Major Feature and a Factor of Poor Prognosis in Sarcomatoid Carcinoma of the Lung. Lung Cancer, 85, 276-281. https://doi.org/10.1016/j.lungcan.2014.06.004

  28. 28. Socinski, M.A., Nishio, M., Jotte, R.M., Cap-puzzo, F., Orlandi, F., Stroyakovskiy, D., et al. (2021) IMpower150 Final Overall Survival Analyses for Atezolizumab plus Bevacizumab and Chemotherapy in First-Line Metastatic Nonsquamous NSCLC. Journal of Thoracic Oncology, 16, 1909-1924. https://doi.org/10.1016/j.jtho.2021.07.009

  29. 29. Pérez-Gutiérrez, L. and Ferrara, N. (2023) Biology and Therapeutic Targeting of Vascular Endothelial Growth Factor A. Nature Reviews Molecular Cell Biology. https://doi.org/10.1038/s41580-023-00631-w

  30. 30. Oizumi, S., Takamura, K., Harada, T., Tachihara, M., Morikawa, N., Honda, R., et al. (2022) Phase II Study of Carboplatin-Paclitaxel Alone or with Bevacizumab in Advanced Sarcoma-toid Carcinoma of the Lung: HOT1201/NEJ024. International Journal of Clinical Oncology, 27, 676-683. https://doi.org/10.1007/s10147-021-02113-5

  31. 31. 李巧珍, 张才金, 王爱民. 阿帕替尼治疗肺巨细胞癌1例并文献复习[J]. 现代肿瘤医学, 2018, 26(4): 530-533.

  32. 32. Chen, X.Z. (2019) Anlotinib for Refractory Advanced Non-Small Cell Lung Cancer in China. JAMA Oncology, 5, 116-117. https://doi.org/10.1001/jamaoncol.2018.5526

  33. 33. 徐涵, 曹雅杰, 周宁宁. 安罗替尼治疗老年晚期肺肉瘤样癌1例[J]. 中华实用诊断与治疗杂志, 2020, 34(10): 1065-1066.

  34. 34. Li, J., Liang, H.J., He, J., Sui, X. and Qin, Y.R. (2021) Anlotinib Combined with Chemotherapy for Recurrence of Pulmonary Sarcomatoid Cancer Previously Surgically Treated: A Case Report and Literature Review. Frontiers in Oncology, 11, Article ID: 639168. https://doi.org/10.3389/fonc.2021.639168

  35. 35. Shen, E., Xiu, J., Bentley, R., López, G.Y. and Walsh, K.M. (2020) Frequent Mutations of POT1 Distinguish Pulmonary Sarcomatoid Carcinoma from Other Lung Cancer Histologies. Clinical Lung Cancer, 21, e523-e527. https://doi.org/10.1016/j.cllc.2020.04.002

  36. 36. Zhou, F., Huang, Y., Cai, W.J., Li, J., Su, C.X., Ren, S.X., et al. (2021) The Genomic and Immunologic Profiles of Pure Pulmonary Sarcomatoid Carcinoma in Chinese Patients. Lung Cancer, 153, 66-72. https://doi.org/10.1016/j.lungcan.2021.01.006

  37. 37. Yang, Z.L., Xu, J.C., Li, L., Li, R.D., Wang, Y.L., Tian, Y.H., et al. (2020) Integrated Molecular Characterization Reveals Potential Therapeutic Strategies for Pulmonary Sarcomatoid Carcinoma. Nature Communications, 11, Article No. 4878. https://doi.org/10.1038/s41467-020-18702-3

  38. 38. Liang, X.H., Li, Q., Xu, B., Hu, S., Wang, Q.Y., Li, Y., et al. (2019) Mutation Landscape and Tumor Mutation Burden Analysis of Chinese Patients with Pulmonary Sarcomatoid Carcinomas. International Journal of Clinical Oncology, 24, 1061-1068. https://doi.org/10.1007/s10147-019-01454-6

  39. 39. Schrock, A.B., Li, S.D., Frampton, G.M., Suh, J., Braun, E., Mehra, R., et al. (2017) Pulmonary Sarcomatoid Carcinomas Commonly Harbor Either Potentially Targetable Genomic Alterations or High Tumor Mutational Burden as Observed by Comprehensive Genomic Profiling. Journal of Thoracic Oncology, 12, 932-942. https://doi.org/10.1016/j.jtho.2017.03.005

  40. 40. Marei, H.E., Althani, A., Afifi, N., Hasan, A., Caceci, T., Pozzoli, G., et al. (2021) P53 Signaling in Cancer Progression and Therapy. Cancer Cell International, 21, Article No. 703. https://doi.org/10.1186/s12935-021-02396-8

  41. 41. Li, X., Zhang, Z.H., Liu, J.H., Wang, D., Wei, S. and Chen, J. (2018) Molecular Features of Giant-Cell Carcinoma of the Lung: A Case Report and Literature Review. OncoTargets and Therapy, 11, 751-756. https://doi.org/10.2147/OTT.S150124

  42. 42. Canale, M. andrikou, K., Priano, I., Cravero, P., Pasini, L., Urbini, M., et al. (2022) The Role of TP53 Mutations in EGFR-Mutated Non-Small-Cell Lung Cancer: Clinical Significance and Im-plications for Therapy. Cancers (Basel), 14, Article No. 1143. https://doi.org/10.3390/cancers14051143

  43. 43. Sabapathy, K. and Lane, D.P. (2018) Therapeutic Targeting of p53: All Mutants Are Equal, but Some Mutants Are More Equal than Others. Nature Reviews Clinical Oncology, 15, 13-30. https://doi.org/10.1038/nrclinonc.2017.151

  44. 44. Wang, X.M., Cao, J., Du, W.J., Zhang, W.H. and Cao, S. (2021) Response to Gefitinib/Crizotinib Combination in a Pulmonary Sarcomatoid Carcinoma Patient Harboring Concurrent EGFR Mutation and MET Amplification. Clinical Case Reports, 9, e04487. https://doi.org/10.1002/ccr3.4487

  45. 45. Italiano, A., Cortot, A.B., Ilie, M., Martel-Planche, G., Fabas, T., Pop, D., et al. (2009) EGFR and KRAS Status of Primary Sarcomatoid Carcinomas of the Lung: Implications for Anti-EGFR Treatment of a Rare Lung Malignancy. International Journal of Cancer, 125, 2479-2482. https://doi.org/10.1002/ijc.24610

  46. 46. Li, X., Wu, D., Liu, H.Y. and Chen, J. (2020) Pulmonary Sarcomatoid Car-cinoma: Progress, Treatment and Expectations. Therapeutic Advances in Medical Oncology, 12, 1-19. https://doi.org/10.1177/1758835920950207

  47. 47. Zou, F.W., Xie, G.Y., Ma, J.-A., Zhou, D.-A., Jiang, Y. and Zheng, J.-Y. (2015) Epidermal Growth Factor Receptor Mutation Heterogeneity Analysis of Pulmonary Sarcomatoid Carcinoma Successfully Treated with Erlotinib: A Case Report. Oncology Letters, 9, 2239-2243. https://doi.org/10.3892/ol.2015.3057

  48. 48. Lococo, F., Gandolfi, G., Rossi, G., Pinto, C., Rapicetta, C., Cavazza, A., et al. (2016) Deep Sequencing Analysis Reveals That KRAS Mutation Is a Marker of Poor Prognosis in Patients with Pulmonary Sarcomatoid Carcinoma. Journal of Thoracic Oncology, 11, 1282-1292. https://doi.org/10.1016/j.jtho.2016.04.020

  49. 49. Fu, Y.L., Wang, A.Q., Zhou, J.Q., Feng, W., Shi, M.H., Xu, X., et al. (2021) Advanced NSCLC Patients with EGFR T790M Harboring TP53 R273C or KRAS G12V Cannot Benefit From Osimertinib Based on a Clinical Multicentre Study by Tissue and Liquid Biopsy. Frontiers in Oncology, 11, Article ID: 621992. https://doi.org/10.3389/fonc.2021.621992

  50. 50. Blair, H.A. (2021) Sotorasib: First Approval. Drugs, 81, 1573-1579. https://doi.org/10.1007/s40265-021-01574-2

  51. 51. Reck, M., Carbone, D.P., Garassino, M. and Barlesi, F. (2021) Targeting KRAS in Non-Small-Cell Lung Cancer: Recent Progress and New Approaches. Annals of Oncology, 32, 1101-1110. https://doi.org/10.1016/j.annonc.2021.06.001

  52. 52. Arbour, K.C., Jordan, E., Kim, H.R., Dienstag, J., Yu, H.A., Sanchez-Vega, F., et al. (2018) Effects of Co-Occurring Genomic Alterations on Outcomes in Patients with KRAS-Mutant Non-Small Cell Lung Cancer. Clinical Cancer Research, 24, 334-340. https://doi.org/10.1158/1078-0432.CCR-17-1841

  53. 53. Vuong, H.G., Ho, A.T.N., Altibi, A.M.A., Nakazawa, T., Katoh, R. and Kondo, T. (2018) Clinicopathological Implications of MET Exon 14 Mutations in Non-Small Cell Lung Cancer—A Systematic Review and Meta-Analysis. Lung Cancer, 123, 76-82. https://doi.org/10.1016/j.lungcan.2018.07.006

  54. 54. Liu, X.W., Jia, Y.X., Stoopler, M.B., Shen, Y.F., Cheng, H.Y., Chen, J.L., et al. (2016) Next-Generation Sequencing of Pulmonary Sarcomatoid Carcinoma Reveals High Frequency of Actionable MET Gene Mutations. Journal of Clinical Oncology, 34, 794-802. https://doi.org/10.1200/JCO.2015.62.0674

  55. 55. Lu, S., Fang, J., Li, X.Y., Cao, L.J., Zhou, J.Y., Guo, Q.S., et al. (2021) Once-Daily Savolitinib in Chinese Patients with Pulmonary Sarcomatoid Carcinomas and Other Non-Small-Cell Lung Cancers Harbouring MET Exon 14 Skipping Alterations: A Multicentre, Single-Arm, Open-Label, Phase 2 Study. The Lancet Respiratory Medicine, 9, 1154-1164. https://doi.org/10.1016/S2213-2600(21)00084-9

  56. 56. Han, S., Fang, J., Lu, S., Wang, L.F., Li, J., Cheng, M., et al. (2019) Response and Acquired Resistance to Savolitinib in a Patient with Pulmonary Sarcomatoid Carcinoma Harboring MET Exon 14 Skipping Mutation: A Case Report. OncoTargets and Therapy, 12, 7323-7328. https://doi.org/10.2147/OTT.S210365

  57. 57. Awad, M.M., Oxnard, G.R., Jackman, D.M., Savukoski, D.O., Hall, D., Shivdasani, P., et al. (2016) MET Exon 14 Mutations in Non-Small-Cell Lung Cancer Are Associated with Advanced Age and Stage-Dependent MET Genomic Amplification and c-Met Overexpression. Journal of Clinical Oncology, 34, 721-730. https://doi.org/10.1200/JCO.2015.63.4600

  58. 58. Chen, X.R., Zhang, Y., Lu, J.B., Xu, C.W., Liang, J.Z., Wang, F., et al. (2017) Pulmonary Sarcomatoid Carcinoma with ALK Rear-Rangement: Frequency, Clinical-Pathologic Characteris-tics, and Response to ALK Inhibitor. Translational Oncology, 10, 115-120. https://doi.org/10.1016/j.tranon.2016.11.009

  59. 59. D’Antonio, F., De Sanctis, R., Bolengo, I., Destro, A., Rahal, D., De Vincenzo, F. and Santoro, A. (2019) Pulmonary Sarcomatoid Carcinoma Presenting both ALK Rearrangement and PD-L1 High Positivity: A Case Report on the Therapeutic Regimen. Medicine (Baltimore), 98, e16754. https://doi.org/10.1097/MD.0000000000016754

  60. 60. Pelosi, G., Gasparini, P., Conte, D., Fabbri, A., Perrone, F., Tamborini, E., et al. (2016) Synergistic Activation upon MET and ALK Coamplification Sustains Targeted Therapy in Sarcomatoid Carcinoma, a Deadly Subtype of Lung Cancer. Journal of Thoracic Oncology, 11, 718-728. https://doi.org/10.1016/j.jtho.2016.01.009

  61. 61. Roesel, C., Kambartel, K., Kopeika, U., Berzins, A., Voshaar, T. and Krbek, T. (2019) Lazarus-Type Tumour Response to Therapy with Nivolumab for Sarcomatoid Carcinomas of the Lung. Current Oncology, 26, e270-e273. https://doi.org/10.3747/co.26.4377

  62. 62. Ma, Y., Li, W.S., Li, Z.Z., Chen, J., Wang, H.T., Jiang, T. and Zhu, J.F. (2022) Immunophenotyping of Pulmonary Sarcomatoid Carcinoma. Frontiers in Immunology, 13, Article ID: 976739. https://doi.org/10.3389/fimmu.2022.976739

  63. 63. Qian, X.Y., Wang, Y., Liu, F.R., Yuan, Y., Fang, C., Zhang, X.W., et al. (2022) The Efficacy and Safety Analysis of First-Line Immune Checkpoint Inhibitors in Pulmonary Sarco-matoid Carcinoma. Frontiers in Immunology, 13, Article ID: 956982. https://doi.org/10.3389/fimmu.2022.956982

  64. 64. Tancoš, V., Farkašová, A., Kviatkovská, Z., Grendár, M., Líško-vá, A., Huťka, Z. and Plank, L. (2020) Non-Small Cell Lung Carcinomas with a Minor Sarcomatoid Component and Pleomorphic Carcinomas Are Associated with High Expression of Programmed Death Ligand 1. Pathology, Research and Practice, 216, Article ID: 153238. https://doi.org/10.1016/j.prp.2020.153238

  65. 65. Kong, F.W., Wang, W.M., Gong, L.B., Wu, W.B. and Liu, Y.Y. (2020) Anti-PD-1 Antibody Camrelizumab plus Doxorubicin Showed Durable Response in Pulmonary Sarcomatoid Carcinoma: Case Report and Literature Review. Journal of Clinical Pharmacy and Therapeutics, 45, 1489-1496. https://doi.org/10.1111/jcpt.13234

  66. 66. Zhou, F., Guo, H.Y., Zhou, X.L., Xie, H.K., Tian, T., Zhao, W.C., et al. (2022) Immune Checkpoint Inhibitors plus Chemotherapy in Patients with Locally Advanced or Metastatic Pulmonary Sarcomatoid Carcinoma: A Multicentric Real-World Study. Therapeutic Advances in Medical Oncology, 14, 1-12. https://doi.org/10.1177/17588359221136759

  67. 67. Diego, S., Roberto, F., Claudia, P., Giuseppe, L.R., Martina, I., Giulia, G., et al. (2019) Immune-Checkpoints Inhibitors in Metastatic Non Small Cell Lung Cancer with Rare Histology. Journal of Clinical Oncology, 37, 9106. https://doi.org/10.1200/JCO.2019.37.15_suppl.9106

  68. 68. Jiao, Y.Y., Liu, M., Luo, N.N., Guo, H. and Li, J.Z. (2021) Successful Treatment of Advanced Pulmonary Sarcomatoid Carcinoma with the PD-1 Inhibitor Toripalimab: A Case Report. Oral Oncology, 112, Article ID: 104992. https://doi.org/10.1016/j.oraloncology.2020.104992

  69. 69. Awobajo, M.D., Vaporciyan, A.A., Lu, C. and Gandhi, S.J. (2020) Stereotactic Body Radiation Therapy (SBRT) in the Management of Pulmonary Spindle Cell Carcinoma. BMJ Case Reports, 13, e234779. https://doi.org/10.1136/bcr-2020-234779

  70. 70. Sukrithan, V., Sandler, J., Gucalp, R., Gralla, R. and Halmos, B. (2019) Immune Checkpoint Blockade Is Associated with Durable Responses in Pulmonary Sarcomatoid Carcinoma. Clinical Lung Cancer, 20, e242-e246. https://doi.org/10.1016/j.cllc.2018.12.013

  71. 71. Li, S.M., Jiang, L., He, Q.M., Wei, W.D., Wang, Y., Zhang, X.K., et al. (2019) The Prognostic Significance of JMJD3 in Primary Sarcomatoid Carcinoma of the Lung, A Rare Subtype of Lung Cancer. OncoTargets and Therapy, 12, 9385-9393. https://doi.org/10.2147/OTT.S225596

  72. 72. Taniguchi, H., Takemoto, S., Ozasa, M., Honda, N., Suyama, T., Umeyama, Y., et al. (2021) Remarkable Response to Pembrolizumab with Platinum-Doublet in PD-L1-Low Pulmonary Sarcomatoid Carcinoma: A Case Report. Thoracic Cancer, 12, 1126-1130. https://doi.org/10.1111/1759-7714.13890

  73. 73. Lococo, F., Torricelli, F., Rossi, G., Alifano, M., Damotte, D., Rapicetta, C., et al. (2017) Inter-Relationship between PD-L1 Expression and Clinic-Pathological Features and Driver Gene Mutations in Pulmonary Sarcomatoid Carcinomas. Lung Cancer, 113, 93-101. https://doi.org/10.1016/j.lungcan.2017.09.009

  74. 74. Yang, Z.L., Xu, J.C., Li, R.D., Gao, Y.B. and He, J. (2019) PD-L1 and CD47 Co-Expression in Pulmonary Sarcomatoid Carcinoma: A Predictor of Poor Prognosis and Potential Targets of Future Combined Immunotherapy. Journal of Cancer Research and Clinical Oncology, 145, 3055-3065. https://doi.org/10.1007/s00432-019-03023-w

  75. 75. Salati, M., Baldessari, C., Calabrese, F., Rossi, G., Pettorelli, E., Grizzi, G., et al. (2018) Nivolumab-Induced Impressive Response of Refractory Pulmonary Sarcomatoid Carcinoma with Brain Metastasis. Case Reports in Oncology, 11, 615-621. https://doi.org/10.1159/000492666

  76. 76. Nishino, K., Kunimasa, K., Kimura, M., Inoue, T., Tamiya, M., Kuhara, H. and Kumagai, T. (2020) Favorable Response to Pembroli-zumab after Durvalumab Failure in a Stage III Sarcomatoid Carcinoma of the Lung: A Case Report. BMC Pharmacology & Toxicology, 21, Article No. 26. https://doi.org/10.1186/s40360-020-00404-7

  77. NOTES

    *通讯作者Email: wangstella2021@qdu.edu.cn

期刊菜单