PD-L1和PD-1在胸腺癌中的表达情况和临床意义
Expression and Clinical Significance of PD-L1 and PD-1 in Thymic Carcinoma
DOI: 10.12677/ACM.2023.133531, PDF, HTML, XML, 下载: 313  浏览: 576 
作者: 王 雪:延安大学附属医院病理科,陕西 延安;空军军医大学第二附属医院病理科,陕西 西安;杜 雄*:延安大学附属医院病理科,陕西 延安
关键词: 胸腺癌程序性死亡配体1程序性死亡受体1免疫治疗Thymic Carcinoma PD-L1 PD-1 Immunotherapy
摘要: 近几年,应用免疫检查点抑制剂阻断PD-1/PD-L1信号通路的免疫治疗在多种恶性肿瘤中表现出显著的临床获益。胸腺癌作为一种罕见的恶性肿瘤,预后较差,治疗方式有限。目前已有证据表明PD-L1在胸腺癌中高表达且其表达情况与预后相关。本文就PD-L1和PD-1在胸腺癌中的临床研究进展作一综述。
Abstract: In recent years, immunotherapy with the application of immune checkpoint inhibitors to block the PD-1/PD-L1 signaling pathway has shown significant clinical benefit in a variety of malignancies. Thymic carcinoma, a rare malignancy, has a poor prognosis and limited therapeutic options. It has been proved that PD-L1 is highly expressed in thymic carcinoma and its expression level is signifi-cantly correlated with the prognosis of thymic carcinoma. This article reviews the clinical research progress of PD-L1 and PD-1 in thymic carcinoma.
文章引用:王雪, 杜雄. PD-L1和PD-1在胸腺癌中的表达情况和临床意义[J]. 临床医学进展, 2023, 13(3): 3702-3708. https://doi.org/10.12677/ACM.2023.133531

1. 引言

胸腺癌(Thymic carcinoma, TC)是一种罕见的肿瘤,占所有胸腺肿瘤不到1%,每100万人中存在1.5例 [1] [2] 。TC最广泛采用的分期是Masaoka-Koga分期 [1] [3] ,其预后较差,生存率主要因肿瘤分期(stages I~II: 91%; stages III~IV: 31%)和切除完整程度而产生差异 [4] 。目前,其治疗方案以手术为首选 [2] [3] [5] [6] 。对于不可切除、转移和复发的肿瘤,首选白蛋白结合紫杉醇为主的化疗方案进行姑息治疗 [5] [7] [8] ,然而,上述治疗方案的疗效不能显著改善患者总体生存率,且铂类药物化疗失败后,尚无明确有效的二线治疗方案 [9] 。随着免疫治疗的深入研究,酪氨酸激酶作为TC的已知靶向突变位点,已有多项靶向治疗方案进入临床试验,但除多激酶抑制剂舒尼替尼外,其余均不成功 [10] 。近年来针对PD-1/PD-L1通路的抑制剂已在多种恶性肿瘤中表现出显著获益,如头颈部鳞状细胞癌、恶性黑色素瘤、非小细胞肺癌、肾细胞癌和食管癌,在Nivolumab治疗头颈部复发性鳞状细胞癌的研究中,治疗组患者比对照组患者总生存期延长2.4个月,另外,关于PD-L1阳性非小细胞肺癌患者的研究表明Pembrolizumab治疗相比化疗来说,缓解率提高了1.6倍(44.8% vs 27.8%) [11] [12] [13] [14] [15] 。现有研究表明PD-1及PD-L1在TC中呈高表达状态,同时其表达情况与预后相关,所以本文主要讨论PD-1和PD-L1在TC中的表达情况及其抑制剂在TC中的临床研究进展。

2. PD-1/PD-L1信号通路及作用机制

PD-1活化后表达于外周CD4+T细胞、CD8+T细胞、自然杀伤性T细胞,B细胞等,PD-L1表达在肿瘤细胞和抗原呈递细胞(APC)等 [16] 。PD-1受体(CD279)及其配体PD-L1 (B7-H1; CD274)组成的信号通路,其本质就是程序性死亡通路,即活化T细胞和APC上表达的PD-1受体与肿瘤细胞上表达的PD-L1配体结合形成抑制通路后,会使得效应T细胞的活化减少,从而使肿瘤细胞逃脱细胞毒性T细胞(CD8+T细胞)介导的细胞杀伤作用,促进肿瘤发生进展 [17] [18] [19] 。在人类胸腺中,PD-L1广泛表达在胸腺皮质和胸腺细胞,并且PD-1和PD-L1之间的通路效应在T细胞的活化中占据重要地位 [20] 。提示,阻断PD-1/PD-L1通路,释放出CD8+T细胞,活化后的CD8+T细胞进而发挥抗肿瘤免疫作用,从而杀伤肿瘤。因为PD-L1在TC中存在表达,已有多项研究对其进行探讨,其中包括3项前瞻性临床试验,针对该通路的研究已成为TC治疗的研究热点 [21] [22] [23] 。

3. PD-1、PD-L1在胸腺癌的表达情况

PD-1、PD-L1在TC中的表达情况从2015年开始研究,到目前为止有12项结果,如表1所示。

由于疾病的罕见性,这些研究仅纳入有限的病例数,从17例~60例不等,得出的结果也存在争议,PD-L1在TC的阳性率在35%~82.9%之间,虽阳性率未趋于一致,但结果在某种程度上表明TC患者的PD-L1存在高表达情况。而对于PD-1表达情况的研究较少,目前结果显示其表达率在1%~65%之间,差异较大。但PD-1和PD-L1表达率之间存在一定程度的一致性,这一点表明PD-1/PD-L1通路在TC肿瘤进展的免疫抑制方面占据重要地位。Sakane等 [24] 研究用四种不同克隆号的PD-L1抗体检测53例TC患者的表达情况,SP142阳性率是92.5% (49/53)、SP263阳性率是49.1% (26/53)、22C3阳性率是64.2% (34/53)、28-8阳性率是77.4% (41/53),此结果表明在TC中22C3和28-8的阳性率一致性较高,其他两种较低。

综上所述,PD-L1在TC的阳性率在35%~82.9%,TC患者的PD-L1存在高表达情况,这为PD-1/PD-L1抑制剂在TC患者的临床应用提供依据。而各研究结果阳性率差异可能是由于使用的抗体不同、判读方法不同或者纳入的患者分期不同等原因。

Table 1. PD-L1 expression profile in thymic carcinoma

表1. 胸腺癌中PD-L1表达特征

4. PD-L1与胸腺癌临床因素及预后的相关性

既往研究显示了PD-L1表达情况与重要临床参数(如年龄、性别、Masaoka-Koga分期、手术切除完整性等)及预后的相关性,如表2所示。但这12项研究对于PD-L1高表达与预后之间的关系存在争议。

Katsuya等 [6] [21] [24] [29] [30] [31] 的研究结果表明PD-L1表达水平与年龄、性别、WHO分期、Masaoka-Koga分期、组织学、原发肿瘤大小、手术可治愈性及新辅助治疗效果无关。Funaki等 [29] 研究认为PD-L1表达水平与是否给予化疗有明显关系,化疗后PD-L1表达显著增高。另外,吴龙等 [32] 研究显示PD-L1表达与肿瘤大小和背景中是否富于淋巴细胞相关,该研究认为对于背景富于淋巴细胞和肿瘤最大径 > 6.0 cm的TC患者,可推荐行PD-L1检测以指导治疗。

关于PD-L1表达与预后的相关研究,Sakane等 [24] 、Yokoyama等 [6] 研究认为PD-L1高表达与预后良好相关,而Funaki等 [29] 、Duan等 [28] 研究结果表明PD-L1表达与预后负相关。其余研究结果显示PD-L1高表达与生存期无明显相关性 [21] [30] [31] 。另外,Yokoyama [6] 和Funaki等 [29] 研究结果认为PD-L1+和PD-1+肿瘤浸润免疫细胞具有预后价值,高表达的患者预后较差。对于PD-L1与预后之间相关性的矛盾结果,原因可能如下:首先,因为各研究在纳入患者时,对Masaoka-Koga分期缺乏严格的限制,使得各研究中Masaoka-Koga分期占比相差很大,因此导致各研究关于生存期方面的结果存在客观偏倚,可比性低;其次,各研究使用的PD-L1抗体克隆号不同,选用的截断值不同,也会导致结果存在差异,Sakane等 [24] 的研究证实了这一点;最后,目前PD-L1与PD-1相关性的研究结果较少且不一致,无法说明PD-L1与预后的关系是否受到PD-1的影响,PD-1可能是导致矛盾结果的重要影响因素。目前因为疾病数量有限,PD-L1和PD-1高表达对预后的意义没有一致的结果,各研究之间的可比性差,需要大样本量的研究进一步来明确其意义,为临床提供可靠依据。

Table 2. Clinicopathological characteristics and prognostic significance associated with PD-L1 expression in thymic carcinoma

表2. 胸腺癌中与PD-L1表达相关的临床病理学特征和预后意义

5. PD-1/PD-L1抑制剂在TC的应用

5.1. 前瞻性临床试验

目前有关PD-1/PD-L1抑制剂治疗TC的研究较少,但这些研究为PD-1/PD-L1抑制剂在TC的临床应用提供依据。主要有以下3个研究(表3):2018年Giaccone等 [22] 的研究入组40例一线化疗后进展的TC患者,进行PD-1抑制剂pembrolizumab免疫治疗,其缓解率达到22.5%,1年无进展生存率达29%,1年总生存率为71%。后续的随访结果显示缓解率无变化(22.5%),五年生存率达18%。在前期研究中发现大多数患者仅发生轻度(1-2级)不良事件,与其他恶性肿瘤采用pembrolizumab治疗报告的不良事件相似。但是,虽然入组的患者既往无自身免疫性疾病,但有6例(15%)患者发生重度自身免疫相关不良反应(immune-related adverse events, irAEs)。同年,Cho等 [9] 的临床试验纳入26例难治性或复发性TC患者进行pembrolizumab免疫治疗,缓解率达19.2% (5/26)。在irAEs的发生方面,有4例(15.4%) TC患者出现3级或4级irAEs,其中3例(11.5%) TC患者因不良反应停止治疗。Giaccone和Cho的研究都表明免疫检查点抑制剂pembrolizumab在TC中具有很好的应答,可以达到一定的临床获益,但其自身免疫毒性的风险也较高,而且TC患者重度irAEs发生率似乎远高于其他类型肿瘤患者。2019年Katsuya等 [23] 研究招募15例不可切除或复发性TC患者,进行PD-1抑制剂nivolumab治疗,11例患者达到疾病稳定。但其研究在第一阶段因无患者应答,研究停止。此研究的患者数量较少,根据实体瘤疗效评价标准(RECIST),在既往接受过治疗的不可切除或复发性TC患者中,nivolumab未能使肿瘤缩小,但研究确实提示了nivolumab具有临床获益。对于nivolumab单抗在TC治疗方面的临床疗效还需进一步的临床试验进行研究,对其临床应用提供更具说服力的数据。

Table 3. Prospective studies of PD-1 inhibitor therapy in thymic adenocarcinoma

表3. 胸腺癌PD-1抑制剂治疗的前瞻性研究

PFS:无病进展生存期;OS:总生存期。

5.2. 案例报道

目前已有的病例报道 [34] [35] [36] [37] 指出针对晚期、多处转移的老年患者,无论PD-L1表达情况如何,都可以采用pembrolizumab或nivolumab治疗,其病情可以达到部分缓解的程度。另外化疗联合免疫治疗的获益不只限于PD-L1 > 50%的转移性TC患者。这些病例报道提示我们,在一些病情严重,情况较为复杂的患者治疗过程中,在充分考虑患者状态和免疫治疗带来的不良反应的基础下,不能将PD-L1的检测值作为用药的唯一指标,要结合病情综合分析。

以上的前瞻性临床试验和病例报道都表明针对PD-1/PD-L1通路的免疫治疗在TC患者中获得了一定的临床获益,为难治性、转移性和晚期患者提供了临床用药依据。并且2022年美国国家综合癌症网络(NCCN)指南也将pembrolizumab纳入TC的二线治疗药物 [37] [38] ,但是因治疗引起的irAEs的发生需要引起足够的重视,在筛选接受免疫治疗的患者时要着重考虑这一点。

6. 讨论

已有的研究结果表明,PD-L1在TC中存在高表达的情况,且PD-1/PD-L1免疫检查点抑制剂治疗在TC患者中存在生存获益。但是PD-1/PD-L1高表达与免疫治疗及预后的关系尚不明确。目前PD-1/PD-L1免疫检查点抑制剂治疗在TC中所面临的问题主要有以下2个方面:① 缺乏预测因子,目前仅单独检测PD-L1不能很好地筛选出免疫治疗的最适患者;② irAEs发生率高。针对以上问题,我们需要研究联合预测指标,更好地选择免疫治疗的最佳患者,如PD-L1与CD8的联合检测,PD-L1+,CD8+是最佳应答患者;而PD-L1+,CD8−的患者,因为缺乏T细胞,阻断抑制通路也无济于事,针对此类患者,我们可以通过联合疫苗接种,过继转移等方式引入T细胞,从而使免疫抑制剂治疗发挥疗效。胸腺作为免疫器官,其irAEs的发生率比其他肿瘤高,现有的临床试验有患者因重度irAEs而中止治疗,其中TC患者发生神经肌肉和心脏毒性的机率比其他肿瘤高 [39] ,针对这一点,已有文献指出乙酰胆碱受体结合自身抗体可考虑为irAEs的预测性标志物 [22] [40] 。检查点抑制剂免疫治疗已参与多种恶性肿瘤患者的管理,已是PD-L1高表达非小细胞肺癌患者的一线标准治疗 [11] [12] [13] [14] [41] [42] 。但在TC中还处于研究阶段,因为不同研究使用的PD-L1抗体、临界值及判读方法不同,导致研究之间可比性较差,寻找单一可靠抗体、确定唯一临界值及判读方法至关重要。并且由于TC罕见这一特性,已有研究都是小样本的回顾性研究,研究可靠性差。

本文主要针对目前已发表的有关TC的实验研究和临床试验,对其进行归纳总结,结果表明PD-L1在TC中存在表达,且针对PD-1/PD-L1通路的PD-1抑制剂pembrolizumab对患者的治疗存在临床获益。对于晚期、复发性患者来说,pembrolizumab是存在临床疗效的治疗方案。但由于疾病的罕见性,已有研究的患者数量受限,大样本的前瞻性研究显得尤为重要。另外,随着肿瘤免疫微环境的不断研究,对于TC的发病机制会有更深入的认识,也会有更多的免疫检查点被发现,PD-L1阴性患者会有更多可选择的治疗方案,TC治疗目前所面对的困难也会迎刃而解。

NOTES

*通讯作者。

参考文献

[1] Yang, X., et al. (2017) The Optimal First-Line Treatment for Advanced Thymic Carcinomas. Journal of Thoracic On-cology, 12, S2061.
https://doi.org/10.1016/j.jtho.2017.09.1083
[2] Eng, T.Y., et al. (2004) Thymic Carcinoma: State of the Art Review. International Journal of Radiation Oncology Biology Physics, 59, 654-664.
https://doi.org/10.1016/j.ijrobp.2003.11.021
[3] Kurup, A. and Loehrer, P.J. (2004) Thymoma and Thymic Car-cinoma: Therapeutic Approaches. Clinical Lung Cancer, 6, 28-32.
https://doi.org/10.3816/CLC.2004.n.018
[4] Litvak, A.M., et al. (2014) Clinical Characteristics and Outcomes for Patients with Thymic Carcinoma: Evaluation of Masaoka Staging. Journal of Thoracic Oncology, 9, 1810-1815.
https://doi.org/10.1097/JTO.0000000000000363
[5] Thomas, A., et al. (2015) Sunitinib in Patients with Chemo-therapy-Refractory Thymoma and Thymic Carcinoma: An Open-Label Phase 2 Trial. The Lancet Oncology, 16, 177-186.
https://doi.org/10.1016/S1470-2045(14)71181-7
[6] Yokoyama, S., et al. (2016) Prognostic Value of Pro-grammed Death Ligand 1 and Programmed Death 1 Expression in Thymic Carcinoma. Clinical Cancer Research, 22, 4727-4734.
https://doi.org/10.1158/1078-0432.CCR-16-0434
[7] Girard, N. (2014) Chemotherapy and Targeted Agents for Thymic Malignancies. Expert Review of Anticancer Therapy, 12, 685-695.
https://doi.org/10.1586/era.12.29
[8] Giaccone, G., et al. (2018) Pembrolizumab in Patients with Thymic Carci-noma: A Single-Arm, Single-Centre, Phase 2 Study. The Lancet Oncology, 19, 347-355.
https://doi.org/10.1016/S1470-2045(18)30062-7
[9] Cho, J., et al. (2018) Pembrolizumab for Patients with Re-fractory or Relapsed Thymic Epithelial Tumor: An Open-Label Phase II Trial. Journal of Clinical Oncology, 37, 2162-2170.
[10] 王凯瑞, 等. 胸腺上皮肿瘤中程序性死亡蛋白1及其配体的临床研究进展[J]. 现代肿瘤医学, 2020, 28(9): 1580-1582.
[11] Ferris, R.L., et al. (2016) Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. New England Journal of Medicine, 375, 1856-1867.
https://doi.org/10.1056/NEJMoa1602252
[12] Postow, M.A., et al. (2015) Nivolumab and Ipilimumab versus Ipilimumab in Untreated Melanoma. New England Journal of Medicine, 372, 2006-2017.
https://doi.org/10.1056/NEJMoa1414428
[13] Reck, M., et al. (2016) Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. New England Journal of Medicine, 375, 1823-1833.
https://doi.org/10.1056/NEJMoa1606774
[14] Motzer, R.J., et al. (2015) Nivolumab versus Everolimus in Ad-vanced Renal-Cell Carcinoma. New England Journal of Medicine, 373, 1803-1813.
https://doi.org/10.1056/NEJMoa1510665
[15] Takashi Kojima, M., et al. (2020) Randomized Phase III KEYNOTE-181 Study of Pembrolizumab versus Chemotherapy in Advanced Esophageal Cancer. Journal of Clinical Oncology, 38, 4138-4148.
https://doi.org/10.1200/JCO.20.01888
[16] Keir, M.E., et al. (2008) PD-1 and Its Ligands in Tolerance and Im-munity. Annual Review of Immunology, 26, 677-704.
https://doi.org/10.1146/annurev.immunol.26.021607.090331
[17] Sharpe, A.H., et al. (2007) The Function of Pro-grammed Cell Death 1 and Its Ligands in Regulating Autoimmunity and Infection. Nature Immunology, 8, 239-245.
https://doi.org/10.1038/ni1443
[18] Okazaki, T. and Honjo, T. (2007) PD-1 and PD-1 Ligands: From Discovery to Clinical Application. International Immunology, 19, 813-824.
https://doi.org/10.1093/intimm/dxm057
[19] Nurieva, R.I., Liu, X. and Dong, C. (2009) Yin-Yang of Costimulation: Crucial Controls of Immune Tolerance and Function. Immunological Reviews, 229, 88-100.
https://doi.org/10.1111/j.1600-065X.2009.00769.x
[20] Francisco, L.M., Sage, P.T. and Sharpe, A.H. (2010) The PD-1 Pathway in Tolerance and Autoimmunity. Immunological Reviews, 236, 219-242.
https://doi.org/10.1111/j.1600-065X.2010.00923.x
[21] Katsuya, Y., et al. (2015) Immunohistochemical Status of PD-L1 in Thymoma and Thymic Carcinoma. Lung Cancer, 88, 154-159.
https://doi.org/10.1016/j.lungcan.2015.03.003
[22] Giaccone, G. and Kim, C. (2021) Durable Response in Patients with Thymic Carcinoma Treated with Pembrolizumab after Prolonged Follow-Up. Journal of Thoracic Oncology, 16, 483-485.
https://doi.org/10.1016/j.jtho.2020.11.003
[23] Katsuya, Y., et al. (2019) Single-Arm, Multicentre, Phase II Trial of Nivolumab for Unresectable or Recurrent Thymic Carcinoma: PRIMER Study. European Journal of Cancer, 113, 78-86.
https://doi.org/10.1016/j.ejca.2019.03.012
[24] Sakane, T., et al. (2018) A Comparative Study of PD-L1 Immunohistochemical Assays with Four Reliable Antibodies in Thymic Carcinoma. Oncotarget, 9, 6993-7009.
https://doi.org/10.18632/oncotarget.24075
[25] Katsuya, Y., et al. (2016) Expression of Programmed Death 1 (PD-1) and Its Ligand (PD-L1) in Thymic Epithelial Tumors: Impact on Treatment Efficacy and Alteration in Expression after Chemotherapy. Lung Cancer, 99, 4-10.
https://doi.org/10.1016/j.lungcan.2016.05.007
[26] Weissferdt, A., et al. (2017) Expression of PD-1 and PD-L1 in Thymic Epithelial Neoplasms. Modern Pathology, 30, 826-833.
https://doi.org/10.1038/modpathol.2017.6
[27] Chen, Y., et al. (2018) Correlation between the Expression of PD-L1 and Clinicopathological Features in Patients with Thymic Epithelial Tumors. BioMed Research International, 2018, Article ID: 5830547.
https://doi.org/10.1155/2018/5830547
[28] Duan, J., et al. (2018) Impact of PD-L1, Transforming Growth Fac-tor-Beta Expression and Tumor-Infiltrating CD8(+) T Cells on Clinical Outcome of Patients with Advanced Thymic Epi-thelial Tumors. Thoracic Cancer, 9, 1341-1353.
https://doi.org/10.1111/1759-7714.12826
[29] Funaki, S., et al. (2019) The Prognostic Impact of Programmed Cell Death 1 and Its Ligand and the Correlation with Epithelial-Mesenchymal Transition in Thymic Carcinoma. Cancer Medi-cine, 8, 216-226.
https://doi.org/10.1002/cam4.1943
[30] Song, J.S., et al. (2019) Clinicopathologic Significance and Immunoge-nomic Analysis of Programmed Death-Ligand 1 (PD-L1) and Programmed Death 1 (PD-1) Expression in Thymic Epi-thelial Tumors. Frontiers in Oncology, 9, 1055.
https://doi.org/10.3389/fonc.2019.01055
[31] Rouquette, I., et al. (2019) Immune Biomarkers in Thymic Epithelial Tumors: Expression Patterns, Prognostic Value and Comparison of Diagnostic Tests for PD-L1. Biomarker Research, 7, Article No. 28.
https://doi.org/10.1186/s40364-019-0177-8
[32] 杨映红, 吴龙, 冯昌银, 黄建平. 胸腺鳞状细胞癌41例临床病理分析及PD-L1表达的意义[J]. 临床与实验病理学杂志, 2021, 37(10): 1227-1230.
[33] Kashima, J., et al. (2022) CD70 in Thymic Squamous Cell Carcinoma: Potential Diagnostic Markers and Immunotherapeutic Targets. Frontiers in Oncology, 11, Article ID: 808396.
https://doi.org/10.3389/fonc.2021.808396
[34] Isshiki, T., et al. (2018) Suc-cessful Use of Pembrolizumab to Treat Refractory Thymic Carcinoma with High PD-L1 Expression. Case Reports in Oncology, 11, 688-692.
https://doi.org/10.1159/000493187
[35] Yang, P., et al. (2018) Response to Nivolumab as Salvage Therapy in a Patient with Thymic Carcinoma. Journal of Thoracic Oncology, 13, e36-e39.
https://doi.org/10.1016/j.jtho.2017.10.022
[36] Rajan, A., et al. (2019) Efficacy and Tolerability of An-ti-Programmed Death-Ligand 1 (PD-L1) Antibody (Avelumab) Treatment in Advanced Thymoma. Journal for Immu-noTherapy of Cancer, 7, 269.
https://doi.org/10.1186/s40425-019-0723-9
[37] Cafaro, A., et al. (2020) Pembrolizumab in a Patient with Heavily Pre-Treated Squamous Cell Thymic Carcinoma and Cardiac Impairment: A Case Report and Literature Review. Frontiers in Oncology, 10, 1478.
https://doi.org/10.3389/fonc.2020.01478
[38] Thomas, Q.D., et al. (2022) Pembrolizumab plus Chemotherapy in Metastatic Thymic Carcinoma: A Case Report. Frontiers in Oncology, 11, Article ID: 814544.
https://doi.org/10.3389/fonc.2021.814544
[39] Rajan, A., et al. (2021) The Role of Immunotherapy for Manage-ment of Advanced Thymic Epithelial Tumors: A Narrative Review. Mediastinum, 5, 23.
https://doi.org/10.21037/med-20-62
[40] Rajan, A. (2021) Immunotherapy for Thymic Cancers: A Convoluted Path toward a Cherished Goal. Journal of Thoracic Oncology, 16, 352-354.
https://doi.org/10.1016/j.jtho.2020.12.007
[41] Brahmer, J., et al. (2015) Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. New England Journal of Medicine, 373, 123-135.
https://doi.org/10.1056/NEJMoa1504627
[42] Borghaei, H., et al. (2015) Nivolumab versus Docetaxel in Ad-vanced Nonsquamous Non-Small-Cell Lung Cancer. New England Journal of Medicine, 373, 1627-1639.
https://doi.org/10.1056/NEJMoa1507643