帕博利珠单抗在三阴性乳腺癌中的应用
The Application of Pembrolizumab in the Treatment of Triple Negative Breast Cancer
DOI: 10.12677/ACM.2024.142331, PDF, HTML, XML, 下载: 143  浏览: 236  科研立项经费支持
作者: 陈 波:西安医学院研究生工作部,陕西 西安;宋张骏*:陕西省人民医院肿瘤外科,陕西 西安
关键词: 帕博利珠单抗三阴性乳腺癌免疫联合疗法PD-1抑制剂Pembrolizumab Triple-Negative Breast Cancer Immunotherapy Combination Therapy PD-1 Inhibitor
摘要: 乳腺癌是全球最常见的癌症,而三阴性乳腺癌(TNBC)是乳腺癌中恶性程度最高的亚型,标准的治疗手段仅限于手术、化疗和放疗,但是治疗效果并不理想。TNBC具有高度异质性以及较强的免疫原性,因此,免疫疗法以及靶向治疗被证明可以提高TNBC的总体生存率和反应,为TNBC提供了有前景的治疗选择。在这篇综述中,我们总结了在TNBC患者中基于免疫检查点抑制剂(ICIs)的各种治疗策略的研究进展,并探讨TNBC免疫治疗前景。
Abstract: Breast cancer is the most common cancer in the world, and triple-negative breast cancer is the most malignant subtype of breast cancer. The standard treatment methods are limited to surgery, chemotherapy and radiotherapy, but the treatment effect is not ideal. Triple-negative breast cancer has high heterogeneity and strong immunogenicity. Therefore, immunotherapy and targeted ther-apy have been proved to improve the overall survival rate and response of triple-negative breast cancer, providing a promising treatment option for triple-negative breast cancer. In this review, we summarized the research progress of various treatment strategies based on immune checkpoint inhibitors in triple-negative breast cancer patients, and discussed the prospect of triple-negative breast cancer immunotherapy.
文章引用:陈波, 宋张骏. 帕博利珠单抗在三阴性乳腺癌中的应用[J]. 临床医学进展, 2024, 14(2): 2360-2367. https://doi.org/10.12677/ACM.2024.142331

1. 前言

乳腺癌是目前最常见的恶性肿瘤,也是全球癌症相关死亡的第二大原因 [1] 。乳腺癌严重危害女性身体健康,更是我国女性癌症死亡的第四大主要原因,且发病率和死亡率逐年上升 [2] 。三阴性乳腺癌(triple-negative breast cancer, TNBC)是其中一个高异质性亚群,占所有原发性乳腺癌的15%~20%,其特点是侵袭性高、早期复发和转移风险高 [3] 。由于不表达雌、孕激素受体(estrogen receptor, ER、progestogen receptor, PR)和人表皮生长因子受体2 (human epidermal growth factor receptor 2, HER2),无法从内分泌或传统的HER2靶向治疗中获益,全身治疗仍以化疗为主,预后较差 [4] 。与非TNBC患者相比,TNBC患者的3年无进展生存(progression-free survival, PFS)时间和总生存(overall survival, OS)时间显著降低,如果实现病理完全缓解(pathological complete response, pCR),将提高TNBC的生存率 [5] 。

近年来,免疫疗法在癌症治疗中受到了极大的关注。以程序性死亡受体-1/程序性死亡受体–配体1 (programmed cell death-1/programmed cell death-ligand 1, PD-1/PD-L1)抑制剂为代表的免疫检查点抑制剂(immune checkpoint inhibitors, ICIs)正越来越多的用于癌症治疗,且疗效显著 [6] 。作为全球最早上市的PD-1抑制剂——帕博利珠单抗,获批适应症最多、覆盖瘤种最广,得到广泛的认可,是目前全球唯一获批治疗TNBC的免疫抑制剂。本文就目前在TNBC患者中以帕博利珠单抗为主要治疗策略的临床研究进行总结,为临床医生和患者选择免疫治疗提供更可靠的依据。

2. TNBC免疫治疗

TNBC缺乏内分泌治疗和靶向治疗的特异性靶点(ER、PR和HER2),化疗、手术联合放疗仍然是TNBC的标准治疗方法。一项大型中国TNBC数据集分析证明了TNBC所具有的生物学异质性,并根据突变的关键基因将其分为四种亚型:(1) 管腔雄激素受体亚型(23%);(2) 免疫细胞信号增强的免疫调节亚型(24%);(3) 基底样免疫抑制亚型(39%);(4) 间充质样亚型(15%) [7] 。由于TNBC的复杂性,传统治疗方法难以使TNBC患者得到精准治疗。

研究发现,免疫相关基因(或基因集)在TNBC中的表达显著高于非TNBC,表明TNBC具有更强的免疫原性,这使得TNBC成为免疫治疗策略的首选亚型 [8] 。另外,PD-1/PD-L1在TNBC中高表达,并且其表达水平与OS呈正相关,使其有望成为一种治疗TNBC的靶点 [9] ,为TNBC的免疫治疗提供了理论依据。

3. 帕博利珠单抗

帕博利珠单抗(Keytruda,K药)是一种人源化单克隆抗体,通过与PD-1特异性结合,阻碍PD-1和PD-L1/PD-L2之间的相互作用,恢复T细胞对肿瘤细胞的原始作用,进而增强人体免疫系统发现和消灭癌细胞的能力,达到癌症治疗的目的 [10] 。

3.1. 帕博利珠单抗单药

作为单一疗法,帕博利珠单抗首先进行了Ⅰb期KEYNOTE-012研究,其中近47%的转移性三阴性乳腺癌(metastatic TNBC, mTNBC)患者之前至少接受了三种治疗,而四分之一的参与者至少接受了五种治疗。在27例疗效可评估的TNBC患者中观察到客观缓解率(objective response rate, ORR)为18.5%,中位PFS和中位OS分别为1.9个月和11.2个月。最常见的治疗相关不良事件(treatment-related adverse event, TRAE)包括关节痛、疲劳、肌痛和恶心等 [11] 。Ⅱ期KEYNOTE-086研究进一步评估了帕博利珠单抗单药治疗mTNBC患者的疗效和安全性。结果显示,A队列(帕博利珠单抗单药治疗先前接受治疗的mTNBC患者)的ORR仅为5.3%,中位PFS和中位OS分别为2.0个月和9.0个月 [12] 。虽然结果较KEYNOTE-012研究相差较大,但同样显示出持久的抗肿瘤活性。B队列(帕博利珠单抗单药一线治疗PD-L1阳性mTNBC患者) ORR达到21.4%,中位PFS和中位OS分别为2.1个月和18.0个月。最常见的TRAE是疲劳、恶心和腹泻,甲状腺疾病是最常见的免疫介导相关不良事件(immune-related adverse event, irAE) [13] 。两个队列的研究结果反映了在未经治疗的mTNBC患者中早期使用ICIs的重要性,尤其是PD-L1阳性肿瘤患者对帕博利珠单抗产生反应的可能性更高。然而,III期KEYNOTE-119研究发现,与单药化疗相比,在先前接受过治疗的mTNBC患者中,帕博利珠单抗单药治疗并未改善ORR、PFS以及OS。虽然如此,仍然在肿瘤PD-L1表达较高的mTNBC患者中观察到帕博利珠单抗单药治疗的临床获益最大 [14] 。综合考虑上述实验的结果,目前暂不建议帕博利珠单抗单独应用于mTNBC的二线或三线治疗。

3.2. 帕博利珠单抗联合化疗

化疗诱导免疫原性细胞死亡(immunogenic cell death, ICD),ICD可以促进树突状细胞成熟,增加肿瘤浸润淋巴细胞和髓系细胞,从而创造免疫反应性肿瘤微环境 [15] 。免疫联合化疗是mTNBC治疗中的第一个组合,并且肿瘤细胞上的PD-L1在化疗后上调 [16] 。免疫疗法和化疗的结合改善了患者的免疫功能,在治疗多种肿瘤方面取得了积极进展,比如黑色素瘤 [17] 、非小细胞肺癌 [18] 、胰腺癌 [19] 、肝癌 [20] 等。

3.2.1. 晚期TNBC

ENHANCE 1研究显示,帕博利珠单抗联合甲磺酸艾立布林在既往未经过系统治疗的mTNBC患者中效果显著,ORR更高(25.8%比21.8%),亚组分析表明,肿瘤PD-L1表达阳性患者的疗效优于PD-L1阴性患者(ORR为28.4%比17.3%)。并且耐受性良好,显示出令人鼓舞的抗肿瘤活性,支持进一步的临床开发 [21] 。

Ⅲ期KEYNOTE-355研究显示,在肿瘤表达PD-L1且联合阳性评分(combined positive score, CPS) ≥ 10的局部复发性不可切除或TNBC或mTNBC患者中,帕博利珠单抗联合化疗组的PFS和OS显著获益(中位PFS 9.7比5.6个月,中位OS 23.0比16.1个月),且治疗效果与PD-L1的富集呈正相关 [22] [23] 。在长期随访中并没有发现帕博利珠单抗联合化疗累积毒性作用的证据。最常见的不良事件(adverse event, AE)通常与化疗的毒性作用相关(最常见的不良反应是疲劳、恶心、腹泻、便秘、呕吐、脱发、皮疹、咳嗽、食欲下降、头痛等;最常见的实验室异常是贫血、白细胞减少、中性粒细胞减少、淋巴细胞减少、血小板减少、谷丙转氨酶和谷草转氨酶升高等),irAE主要是甲状腺功能减退和甲状腺功能亢进,这些事件一般都是低级别的,可以通过中断用药和适当的支持治疗来控制 [18] 。基于该研究结果,2020年11月美国食品和药品管理局(FDA)批准帕博利珠单抗联合化疗用于肿瘤PD-L1阳性且CPS ≥ 10的局部复发性不可切除或mTNBC患者,同时批准PD-L1 IHC 22C3 pharmDx测定作为该适应症的伴随诊断。

基于KEYNOTE-355数据进行的一项生存质量调整的无疾病或毒性症状时间(Q-TWiST)分析表明,在PD-L1阳性mTNBC的一线治疗中,帕博利珠单抗联合化疗的Q-TWiST具有统计学意义和临床意义的改善;并且,随着随访时间的延长,Q-TWiST效益也会增加 [24] 。这进一步加强了帕博利珠单抗联合化疗在mTNBC中的价值。

3.2.2. 早期TNBC

Ⅰb期KEYNOTE-173研究表明,帕博利珠单抗联合新辅助化疗治疗高危早期TNBC (early TNBC, eTNBC)具有可控的毒性和良好的抗肿瘤活性,探索性分析表明pCR率与肿瘤PD-L1表达和肿瘤浸润淋巴细胞水平呈正相关。常见的AE与化疗中观察到的AE一致 [25] 。II期随机I-SPY2试验中,帕博利珠单抗联合新辅助化疗(紫杉醇、阿霉素、环磷酰胺)在TNBC组的pCR率最高,达到60% (对照组22%) [26] 。

KEYNOTE-522研究发现,在新辅助化疗中加入帕博利珠单抗并术后帕博利珠单抗辅助治疗可使高危eTNBC患者的pCR率从51.2%提高到64.8% [27] ,3年无事件生存(event-free survival, EFS)率显著改善(84.5%比76.8%) [28] 。因此,2021年7月,FDA再次批准帕博利珠单抗联合化疗作为新辅助治疗用于高危eTNBC患者,术后继续使用帕博利珠单抗辅助治疗。这无疑为广大eTNBC患者带来了新的治愈希望,此次新适应证的获批将为处于早期治疗阶段的TNBC患者带来更多获益。

2022年11月,国家药品监督管理局(NMPA)批准术前帕博利珠单抗联合化疗并在术后继续帕博利珠单抗辅助治疗用于肿瘤表达PD-L1 (CPS ≥ 20)的高危eTNBC患者,且在中国临床肿瘤学会(CSCO)乳腺癌诊疗指南2023版中,紫杉烷类和铂类联合帕博利珠单抗获Ⅱ级推荐(证据级别2A),且无论是否达到pCR,均推荐继续使用帕博利珠单抗辅助治疗至满1年为Ⅱ级推荐(证据级别1A)。2023年欧洲肿瘤内科学会(ESMO)上公布的结果再一次确证了该方案的持续获益:与安慰剂组相比60个月EFS提高了9% (81.3%比72.3%) [29] 。该临床研究进一步强调了帕博利珠单抗联合化疗在TNBC治疗中的价值,为TNBC患者提供了一种新的治疗方法,同时,也为医生和患者做出医疗决策提供了有力的证据。

目前还在研究中的新方案,比如:通过pCR率和TRAE来评估帕博利珠单抗加多西他赛化疗联合白细胞介素-12基因治疗难治性TNBC的有效性和安全性(NCT04095689),以及评估帕博利珠单抗加环磷酰胺联合新型长效重组人白细胞介素7 (GX-I7)治疗难治性或复发性TNBC安全性、耐受性、药效学和抗肿瘤活性的研究(NCT03752723)等。

3.3. 帕博利珠单抗联合靶向治疗

3.3.1. 聚腺苷二磷酸核糖聚合酶(PARP)抑制剂

PARP抑制剂通过激活cGAS-STING途径,诱导I型干扰素的释放及其相关免疫应答 [30] 。同时发现PARP抑制剂使GSK3β失活增强了TNBC中PD-L1的表达。PARP抑制剂通过上调PD-L1和阻断PD-L1使PARP抑制剂处理的癌细胞对T细胞杀伤重新致敏,从而减弱抗癌免疫 [31] 。

II期TOPACIO研究显示,帕博利珠单抗联合尼拉帕利治疗晚期或TNBC或mTNBC患者具有可耐受的安全性,疗效显著,总体ORR为21%,疾病控制率(disease control rate, DCR)为49%,尤其对肿瘤乳腺癌易感基因突变患者效果更明显,ORR和DCR分别高达47%和80% [32] 。当然还有正在进行的KEYLYNK-009 (NCT04191135)试验:在局部复发、无法手术的TNBC或mTNBC患者中帕博利珠单抗化疗诱导后评估帕博利珠单抗联合PARP抑制剂治疗的最终PFS和OS,以评估不同联合治疗策略对疾病的缓解作用。

3.3.2. 内分泌治疗

管腔雄激素受体亚型约占TNBC的15%~20%,由过度表达雄激素受体(androgen receptor, AR)定义,AR靶向治疗在AR阳性TNBC中表现出适度的活性,引起了人们的兴趣 [33] 。

在一项帕博利珠单抗联合Enobosarm (MK-2866,一种选择性雄激素受体调节剂)治疗AR阳性mTNBC患者的临床试验中,其临床获益率(clinical benefit rate, CBR)为25%,且耐受性良好 [34] 。进一步说明了优化分子分型体系在提高精准诊断和靶向治疗中的重要性,未来AR靶向治疗联合ICIs治疗AR阳性TNBC的临床试验值得继续研究。

3.3.3. 癌症疫苗

癌症疫苗是通过向患者注射癌症特异性抗原来触发针对肿瘤的免疫反应,以增强免疫系统识别和杀死癌细胞的能力 [35] 。由于现代技术的进步以及受到新型冠状病毒疾病大流行的推动,癌症疫苗这一积极的免疫疗法,再次成为人们关注的焦点。

在TNBC患者中使用帕博利珠单抗联合p53MVA (表达p53的改良安卡拉痘苗),每三周静脉注射一次帕博利珠单抗,并在至少30分钟后皮下注射p53MVA,这有助于在实体瘤患者中建立有效的免疫反应,结果显示治疗具有较高的安全性和耐受性,最常见的AE是注射部位反应、疲劳和流感样症状 [36] 。另外还有许多癌症疫苗相关临床试验正在进行中(见表1)。

Table 1. Clinical study on the combination of pembrolizumab and cancer vaccine in TNBC

表1. TNBC中帕博利珠单抗联合癌症疫苗相关临床研究

注:HLA-A2,人白细胞抗原A2;TNBC,三阴性乳腺癌;PD-L1,程序性死亡受体-配体1;mTNBC,转移性三阴性乳腺癌;BC,乳腺癌;HER2,人表皮生长因子受体2;HER3,人类表皮生长因子受体3。

3.4. 帕博利珠单抗联合联合放疗

放射治疗(radiation therapy, RT)同样可以诱导ICD,增强抗肿瘤免疫反应。局部RT联合PD-1抑制剂可以增强系统性抗肿瘤作用,通过协同方式利用免疫系统的潜力来促进癌症治疗,导致肿瘤的全身控制 [37] [38] 。

一项Ⅱ期研究显示,mTNBC患者在5~7天内每天接受30 Gy/5f的RT(主要目的是减轻患者的疼痛或控制肿瘤的转移)。在第一次RT后1至3天静脉注射200 mg剂量的帕博利珠单抗,此后每3周注射一次,直到出现进行性疾病或不可接受的毒性,结果显示整体ORR为17.6%,安全性良好 [39] 。该结果在未进行PD-L1选择的TNBC患者中令人鼓舞,但是后期需要更大规模的检查点阻断加RT的临床试验,并具有预测反应的生物标志物。目前正在进行的临床试验:研究在TNBC患者接受新辅助治疗后使用帕博利珠单抗联合放疗的有效性和安全性的NCT02954874等。

3.5. 帕博利珠单抗联合溶瘤病毒疗法

溶瘤病毒(Oncolytic virus, OVs)是一种天然(例如呼肠孤病毒)或人工修饰的病毒[例如单纯疱疹病毒-1 (HSV-1)、腺病毒等],可选择性地感染和破坏肿瘤细胞 [40] 。OVs治疗依赖于非致病性病毒株的给药,该病毒株选择性地感染和杀死恶性细胞,同时有利于激发治疗相关的肿瘤靶向免疫应答 [41] 。PD-1/PD-L1抑制剂联合溶瘤病毒治疗有望在mTNBC中取得积极效果。

一项单中心Ⅱ期STOMP临床试验,旨在评估腺病毒介导的单纯疱疹病毒胸苷激酶(ADV/HSV-tk)加阿昔洛韦联合立体定向放疗诱导治疗,然后使用帕博利珠单抗治疗mTNBC患者的疗效。结果显示CBR为21.4%,并且获益患者具有持久的免疫反应 [42] 。但是此方案还需更大规模的随机研究验证。

4. 总结与展望

近年来,ICIs抑制剂在抗肿瘤治疗中有着令人惊讶的表现。尽管ICIs抑制剂治疗在多种恶性肿瘤中显示出积极的临床益处,但在mTNBC患者中单独使用的结果并不理想。虽然以帕博利珠单抗为主的免疫疗法为TNBC患者带来了巨大的希望,但我们必须意识到,许多研究仍在进行中,并且还有一些问题仍未得到解答。我们更多更深入的临床试验,寻找更合适、更可靠的肿瘤生物标志物和联合治疗策略,以便为更多TNBC患者带来益处。

另外,免疫治疗的不良反应也是治疗过程中需要重视的问题,在选择治疗前,要权衡利弊,谨慎选择。许多帕博利珠单抗的临床试验还在进行中,未来或将改变TNBC患者的治疗格局,我们迫切等待它们的综合结果为TNBC免疫治疗得出最终结论。

基金项目

陕西省自然科学基础研究计划——重点项目(2022JZ-57);陕西省感染与免疫疾病重点实验室开放课题——重点项目(2022-ZD-2);陕西省人民医院科技人才支持计划项目——领军人才(2021LJ-01);秦创原中医药创新研发转化项目(2022-QCYZH-004)。

NOTES

*通讯作者。

参考文献

[1] Sung, H., Ferlay, J., Siegel, R.L., et al. (2021) Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer Journal for Clinicians, 71, 209-249.
https://doi.org/10.3322/caac.21660
[2] Xia, C., Dong, X., Li, H., et al. (2022) Cancer Statistics in China and United States, 2022: Profiles, Trends, and Determinants. Chinese Medical Journal (England), 135, 584-590.
https://doi.org/10.1097/CM9.0000000000002108
[3] Li, Y., Zhang, H., Merkher, Y., et al. (2022) Recent Ad-vances in Therapeutic Strategies for Triple-Negative Breast Cancer. Journal of Hematology & Oncology, 15, Article No. 121.
https://doi.org/10.1186/s13045-022-01341-0
[4] Borri, F. and Granaglia, A. (2021) Pathology of Triple Negative Breast Cancer. Seminars in Cancer Biology, 72, 136-145.
https://doi.org/10.1016/j.semcancer.2020.06.005
[5] Liedtke, C., Mazouni, C., Hess, K.R., et al. (2023) Response to Neoadjuvant Therapy and Long-Term Survival in Patients with Triple-Negative Breast Cancer. Journal of Clinical Oncology, 41, 1809-1815.
https://doi.org/10.1200/JCO.22.02572
[6] Kumar, A.R., Devan, A.R., Nair, B., et al. (2021) Harnessing the Im-mune System against Cancer: Current Immunotherapy Approaches and Therapeutic Targets. Molecular Biology Reports, 48, 8075-8095.
https://doi.org/10.1007/s11033-021-06752-9
[7] Jiang, Y.Z., Ma, D., Suo, C., et al. (2019) Genomic and Tran-scriptomic Landscape of Triple-Negative Breast Cancers: Subtypes and Treatment Strategies. Cancer Cell, 35, 428-440.e5.
[8] Liu, Z., Li, M., Jiang, Z., et al. (2018) A Comprehensive Immunologic Portrait of Triple-Negative Breast Cancer. Translational Oncology, 11, 311-329.
https://doi.org/10.1016/j.tranon.2018.01.011
[9] Barrett, M.T., Lenkiewicz, E., Malasi, S., et al. (2018) The Association of Genomic Lesions and PD-1/PD-L1 Expression in Re-sected Triple-Negative Breast Cancers. Breast Cancer Research, 20, Article No. 71.
https://doi.org/10.1186/s13058-018-1004-0
[10] Homšek, A., Radosavljević, D., Miletić, N., et al. (2022) Review of the Clinical Pharmacokinetics, Efficacy and Safety of Pembrolizumab. Current Drug Metabolism, 23, 460-472.
https://doi.org/10.2174/1389200223666220609125013
[11] Nanda, R., Chow, L.Q.M., Dees, E.C., et al. (2016) Pembrolizumab in Patients with Advanced Triple-Negative Breast Cancer: Phase Ib KEYNOTE-012 Study. Journal of Clinical Oncology, 34, 2460-2467.
https://doi.org/10.1200/JCO.2015.64.8931
[12] Adams, S., Schmid, P., Rugo, H.S., et al. (2019) Pembrolizumab Monotherapy for Previously Treated Metastatic Triple-Negative Breast Cancer: Cohort A of the Phase II KEYNOTE-086 Study. Annals of Oncology, 30, 397-404.
https://doi.org/10.1093/annonc/mdy517
[13] Adams, S., Loi, S., Toppmeyer, D., et al. (2019) Pembrolizumab Monotherapy for Previously Untreated, PD-L1-Positive, Metastatic Triple-Negative Breast Cancer: Cohort B of the Phase II KEYNOTE-086 Study. Annals of Oncology, 30, 405-411.
https://doi.org/10.1093/annonc/mdy518
[14] Winer, E.P., Lipatov, O., Im, S.A., et al. (2021) Pembrolizumab ver-sus Investigator-Choice Chemotherapy for Metastatic Triple-Negative Breast Cancer (KEYNOTE-119): A Randomised, Open-Label, Phase 3 Trial. The Lancet Oncology, 22, 499-511.
https://doi.org/10.1016/S1470-2045(20)30754-3
[15] Li, Z., Lai, X., Fu, S., et al. (2022) Immunogenic Cell Death Activates the Tumor Immune Microenvironment to Boost the Immunotherapy Efficiency. Advanced Science (Weinh), 9, Article ID: 2201734.
https://doi.org/10.1002/advs.202201734
[16] Vranic, S., Cyprian, F.S., Gatalica, Z., et al. (2021) PD-L1 Status in Breast Cancer: Current View and Perspectives. Seminars in Cancer Biology, 72, 146-154.
https://doi.org/10.1016/j.semcancer.2019.12.003
[17] De Almeida Gonçalves, V.D.M., De Almeida Camargo Filho, M.F., Zaleski, T., et al. (2021) Chemotherapy in Focus: A Meta-Analysis Confronts Immunotherapy in the Treatment of Advanced Melanoma. Critical Reviews in Oncology/Hematology, 161, Article ID: 103304.
https://doi.org/10.1016/j.critrevonc.2021.103304
[18] Du, C., Chen, Y., Zhou, Y., et al. (2023) Neo-Adjuvant Chemotherapy plus Immunotherapy in Resectable N1/N2 NSCLC. BMC Cancer, 23, Article No. 1260.
https://doi.org/10.1186/s12885-023-11745-x
[19] Lu, J., Liu, X., Liao, Y.P., et al. (2017) Nano-Enabled Pancreas Cancer Immunotherapy Using Immunogenic Cell Death and Reversing Immunosuppression. Nature Communications, 8, Article No. 1811.
https://doi.org/10.1038/s41467-017-01651-9
[20] Zhang, W., Zhang, K., Liu, C., et al. (2023) Hepatic Arterial In-fusion Chemotherapy Combined with Anti-PD-1/PD-L1 Immunotherapy and Molecularly Targeted Agents for Advanced Hepatocellular Carcinoma: A Real World Study. Frontiers in Immunology, 14, Article ID: 1127349.
https://doi.org/10.3389/fimmu.2023.1127349
[21] Tolaney, S.M., Kalinsky, K., Kaklamani, V.G., et al. (2021) Eribulin plus Pembrolizumab in Patients with Metastatic Triple-Negative Breast Cancer (ENHANCE 1): A Phase Ib/II Study. Clinical Cancer Research, 27, 3061-3068.
https://doi.org/10.1158/1078-0432.CCR-20-4726
[22] Cortes, J., Cescon, D.W., Rugo, H.S., et al. (2020) Pem-brolizumab plus Chemotherapy versus Placebo plus Chemotherapy for Previously Untreated Locally Recurrent Inopera-ble or Metastatic Triple-Negative Breast Cancer (KEYNOTE-355): A Randomised, Placebo-Controlled, Double-Blind, Phase 3 Clinical Trial. The Lancet, 396, 1817-1828.
https://doi.org/10.1016/S0140-6736(20)32531-9
[23] Cortes, J., Rugo, H.S., Cescon, D.W., et al. (2022) Pem-brolizumab plus Chemotherapy in Advanced Triple-Negative Breast Cancer. The New England Journal of Medicine, 387, 217-226.
https://doi.org/10.1056/NEJMoa2202809
[24] Huang, M., O’Shaughnessy, J., Haiderali, A., et al. (2022) Q-TWiST Analysis of Pembrolizumab Combined with Chemotherapy as First-Line Treatment of Metastatic Tri-ple-Negative Breast Cancer That Expresses PD-L1. European Journal of Cancer, 177, 45-52.
https://doi.org/10.1016/j.ejca.2022.09.029
[25] Schmid, P., Salgado, R., Park, Y.H., et al. (2020) Pembrolizumab plus Chemotherapy as Neoadjuvant Treatment of High-Risk, Early-Stage Triple-Negative Breast Cancer: Results from the Phase 1b Open-Label, Multicohort KEYNOTE-173 Study. Annals of Oncology, 31, 569-581.
https://doi.org/10.1016/j.annonc.2020.01.072
[26] Nanda, R., Liu, M.C., Yau, C., et al. (2020) Effect of Pembroli-zumab plus Neoadjuvant Chemotherapy on Pathologic Complete Response in Women with Early-Stage Breast Cancer: An Analysis of the Ongoing Phase 2 Adaptively Randomized I-SPY2 Trial. JAMA Oncology, 6, 676-684.
https://doi.org/10.1001/jamaoncol.2019.6650
[27] Schmid, P., Cortes, J., Pusztai, L., et al. (2020) Pembrolizumab for Early Triple-Negative Breast Cancer. The New England Journal of Medicine, 382, 810-821.
https://doi.org/10.1056/NEJMoa1910549
[28] Schmid, P., Cortes, J., Dent, R., et al. (2022) Event-Free Survival with Pembrolizumab in Early Triple-Negative Breast Cancer. The New England Journal of Medicine, 386, 556-567.
https://doi.org/10.1056/NEJMoa2112651
[29] Schmid, P., Cortés, J., Dent, R.A., et al. (2023) LBA18 Pembroli-zumab or Placebo plus Chemotherapy Followed by Pembrolizumab or Placebo for Early-Stage TNBC: Updated EFS Results from the Phase III KEYNOTE-522 Study. Annals of Oncology, 34, S1257.
https://doi.org/10.1016/j.annonc.2023.10.008
[30] Shen, J., Zhao, W., Ju, Z., et al. (2019) PARPi Triggers the STING-Dependent Immune Response and Enhances the Therapeutic Efficacy of Immune Checkpoint Blockade Inde-pendent of BRCAness. Cancer Research, 79, 311-319.
https://doi.org/10.1158/0008-5472.CAN-18-1003
[31] Jiao, S., Xia, W., Yamaguchi, H., et al. (2017) PARP In-hibitor Upregulates PD-L1 Expression and Enhances Cancer-Associated Immunosuppression. Clinical Cancer Research, 23, 3711-3720.
https://doi.org/10.1158/1078-0432.CCR-16-3215
[32] Vinayak, S., Tolaney, S.M., Schwartzberg, L., et al. (2019) Open-Label Clinical Trial of Niraparib Combined with Pembrolizumab for Treatment of Advanced or Metastatic Tri-ple-Negative Breast Cancer. JAMA Oncology, 5, 1132-1140.
https://doi.org/10.1001/jamaoncol.2019.1029
[33] Khadela, A., Chavda, V.P., Soni, S., et al. (2022) An-ti-Androgenic Therapies Targeting the Luminal Androgen Receptor of a Typical Triple-Negative Breast Cancer. Cancers (Basel), 15, Article No. 233.
https://doi.org/10.3390/cancers15010233
[34] Yuan, Y., Lee, J.S., Yost, S.E., et al. (2021) A Phase II Clinical Tri-al of Pembrolizumab and Enobosarm in Patients with Androgen Receptor-Positive Metastatic Triple-Negative Breast Cancer. Oncologist, 26, e99-e217.
https://doi.org/10.1002/onco.13583
[35] Solinas, C., Aiello, M., Migliori, E., et al. (2020) Breast Cancer Vaccines: Heeding the Lessons of the Past to Guide a Path forward. Cancer Treatment Reviews, 84, Article ID: 101947.
https://doi.org/10.1016/j.ctrv.2019.101947
[36] Chung, V., Kos, F.J., Hardwick, N., et al. (2019) Evaluation of Safety and Efficacy of P53MVA Vaccine Combined with Pembrolizumab in Patients with Advanced Solid Cancers. Clinical and Translational Oncology, 21, 363-372.
https://doi.org/10.1007/s12094-018-1932-2
[37] Song, H.N., Jin, H., Kim, J.H., et al. (2021) Abscopal Effect of Radiotherapy Enhanced with Immune Checkpoint Inhibitors of Triple Negative Breast Cancer in 4T1 Mammary Carci-noma Model. International Journal of Molecular Sciences, 22, Article No. 10476.
https://doi.org/10.3390/ijms221910476
[38] Pointer, K.B., Pitroda, S.P. and Weichselbaum, R.R. (2022) Radio-therapy and Immunotherapy: Open Questions and Future Strategies. Trends in Cancer, 8, 9-20.
https://doi.org/10.1016/j.trecan.2021.10.003
[39] Ho, A.Y., Barker, C.A., Arnold, B.B., et al. (2020) A Phase 2 Clinical Trial Assessing the Efficacy and Safety of Pembrolizumab and Radiotherapy in Patients with Metastatic Tri-ple-Negative Breast Cancer. Cancer, 126, 850-860.
https://doi.org/10.1002/cncr.32599
[40] Gujar, S., Pol, J.G., Kim, Y., et al. (2018) Antitumor Benefits of Antiviral Immunity: An Underappreciated Aspect of Oncolytic Virotherapies. Trends in Immunology, 39, 209-221.
https://doi.org/10.1016/j.it.2017.11.006
[41] Wang, G., Kang, X., Chen, K.S., et al. (2020) An Engineered Onco-lytic Virus Expressing PD-L1 Inhibitors Activates Tumor Neoantigen-Specific T Cell Responses. Nature Communica-tions, 11, Article No. 1395.
https://doi.org/10.1038/s41467-020-15229-5
[42] Sun, K., Xu, Y., Zhang, L., et al. (2022) A Phase 2 Trial of En-hancing Immune Checkpoint Blockade by Stereotactic Radiation and in Situ Virus Gene Therapy in Metastatic Tri-ple-Negative Breast Cancer. Clinical Cancer Research, 28, 4392-4401.
https://doi.org/10.1158/1078-0432.CCR-22-0622