国际妇产科学杂志, 2021, 48(6): 605-609 doi: 10.12280/gjfckx.20210173

妇科肿瘤研究:综述

PD-1/PD-L1免疫抑制剂治疗卵巢癌的临床应用进展

毛若南, 姜伟,

200090 上海,复旦大学附属妇产科医院妇科

Progress in the Clinical Application of PD-1/PD-L1 Inhibitor in the Treatment of Ovarian Neoplasms

MAO Ruo-nan, JIANG Wei,

Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China

通讯作者: 姜伟,E-mail:jw52317@126.com

审校者

责任编辑: 秦娟

收稿日期: 2021-02-21  

基金资助: 国家自然科学基金(81672567)

Corresponding authors: JIANG Wei, E-mail:jw52317@126.com

Received: 2021-02-21  

摘要

在女性生殖系统肿瘤中,卵巢癌的致死率是最高的。目前治疗卵巢癌的方法主要有手术联合放、化疗以及靶向治疗,但是对于患者的生存率没有明显改善。免疫治疗是近年提出的治疗卵巢恶性肿瘤的新策略,旨在增强患者免疫系统识别和攻击肿瘤细胞的能力。目前已有多种免疫疗法被证实有一定的疗效,其中程序性死亡蛋白-1(programmed death-1,PD-1)/程序性死亡蛋白配体-1(programmed death ligand-1,PD-L1)检查点抑制剂已进入临床用来改善晚期卵巢癌、复发性卵巢癌、铂敏感或者铂耐药卵巢癌患者的预后。最常见的PD-1/PD-L1免疫抑制剂包括纳武单抗、派姆单抗、阿特珠单抗、阿维单抗和度伐利尤单抗等,就这些免疫抑制剂在卵巢癌中的临床治疗进展,尤其是最新的联合治疗进展进行了综述。

关键词: 卵巢肿瘤; 免疫疗法; 程序性细胞死亡受体1; 抗原,CD274; 药物疗法,联合

Abstract

Ovarian neoplasm has the highest mortality among female reproductive system tumors. The methods of treating ovarian cancer involve surgery combined with radiotherapy, chemotherapy and targeted therapy, yet there is no significant improvement in the survival rate of patients. Immunotherapy is a new strategy for malignant ovarian tumors and aims to boost the capacity of the immune system to recognize and attack tumor cells. Some of them have been proven to show effects, among which programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) checkpoint inhibitors had been applied to improve the prognosis of advanced, recurrent platinum-sensitive or platinum-resistant ovarian cancer patients. PD-1/PD-L1 inhibitors include Nivolumab, Pembrolizumab, Atezolizumab, Avelumab, Durvalumab, etc. In this paper, we reviewed the progress of PD-1/PD-L1 inhibitors for ovarian cancer especially the combination therapy.

Keywords: Ovarian neoplasms; Immunotherapy; Programmed cell death 1 receptor; Antigens,CD274; Drug therapy,combination

PDF (700KB) 元数据 多维度评价 相关文章 导出 EndNote| Ris| Bibtex  收藏本文

本文引用格式

毛若南, 姜伟. PD-1/PD-L1免疫抑制剂治疗卵巢癌的临床应用进展[J]. 国际妇产科学杂志, 2021, 48(6): 605-609 doi:10.12280/gjfckx.20210173

MAO Ruo-nan, JIANG Wei. Progress in the Clinical Application of PD-1/PD-L1 Inhibitor in the Treatment of Ovarian Neoplasms[J]. Journal of International Obstetrics and Gynecology, 2021, 48(6): 605-609 doi:10.12280/gjfckx.20210173

卵巢癌是全球范围内死亡率最高的妇科恶性肿瘤,由于早期症状不明显,没有特异性的筛查方法,大多数患者确诊时已是晚期[1]。目前治疗卵巢癌的方法主要有手术联合放、化疗以及靶向疗法,但由于其高的耐药率、复发率和死亡率严重威胁着患者生命,所以寻求更好的治疗方法提高卵巢癌患者的生存率显得尤为重要[2]。正常情况下,人体的免疫系统可以识别并清除肿瘤细胞,但是由于肿瘤细胞可以采取不同的“逃避”方法,从而导致机体无法杀伤肿瘤细胞,使其在抗肿瘤的免疫应答中得以幸存,这一过程称为肿瘤免疫逃逸[3]。肿瘤免疫逃逸有多种途径,包括抗原呈递机制的减弱、肿瘤相关抗原的表达下调、肿瘤细胞内在干扰素(IFN)信号通路的失衡、免疫抑制细胞的募集以建立“免疫特权”微环境、免疫抑制分子的上调和肿瘤细胞的代谢变化等[4]。其中,程序性死亡蛋白配体-1(programmed death ligand-1,PD-L1)作为肿瘤细胞表面的抑制性膜分子参与肿瘤细胞的免疫逃逸[5]。针对程序性死亡蛋白-1(programmed death-1,PD-1)/PD-L1的检查点抑制剂疗法在对黑色素瘤、卵巢癌在内的多种肿瘤中已取得显著疗效。现对PD-1/PD-L1抑制剂治疗卵巢癌,尤其是联合用药的临床研究进展予以综述,对其临床治疗提供新思路。

1 PD-1/PD-L1抑制剂肿瘤免疫治疗原理

PD-1属于CD28家族,主要表达于活化的T细胞、B细胞、自然杀伤(NK)细胞等免疫细胞表面。PD-1的配体是PD-L1(也称为B7-H1、CD274),常表达于各种肿瘤细胞表面。肿瘤细胞可以激活PD-1/PD-L1的抑制性信号通路,限制T细胞活性,形成适合肿瘤细胞生长的免疫微环境,从而发生肿瘤免疫逃逸[6]。因此,针对PD-1/PD-L1设计特定的蛋白质抗体(抑制剂)能阻断该通路,使T细胞恢复免疫活性,增强免疫应答,提高免疫系统杀伤肿瘤细胞的能力,这成为了近年肿瘤治疗领域的重大突破。另有研究发现,大多数卵巢肿瘤具有高表达水平的PD-L1[7],为其PD-1/PD-L1抑制剂免疫治疗提供了可能性。目前常见的PD-1/PD-L1抑制剂有Nivolumab(纳武利尤单抗,纳武单抗)、Pembrolizumab(派姆单抗)、Atezolizumab(阿特珠单抗,阿替利珠单抗)、Avelumab(阿维单抗)、Durvalumab(度伐利尤单抗,德瓦鲁单抗)以及国产的特瑞普利单抗、信迪利单抗、卡瑞利珠单抗和替雷利珠单抗等。

2 PD-1/PD-L1抑制剂在卵巢癌中的临床应用

2.1 Nivolumab治疗卵巢癌的临床进展

2018年Matsuo等[8]对乳腺癌易感基因(breast cancer suscepti-bility gene,BRCA)突变阳性的复发性上皮性卵巢癌患者(n=5)和输卵管癌患者(n=1)进行了回顾性分析,这些患者接受了Nivolumab单药治疗(3.0 mg/kg,静脉注射,每4周的第1天和第15天各1次)。中位随访时间为13.4个月,有3例完全缓解(complete response,CR),1例部分缓解(partial response,PR)和2例疾病进展(progressive disease,PD),客观缓解率(objective response rate,ORR)为67%。该研究表明,Nivolumab单一疗法耐受性良好,可以在一定程度上阻断复发性上皮性卵巢癌的发展。之后,Normann等[9]进行的一项临床研究评估了PD-1抑制剂Nivolumab对铂耐药卵巢癌患者的毒性和临床疗效,该研究纳入了18例铂耐药的卵巢癌患者,对其应用Nivolumab单药治疗(3 mg/kg,静脉注射,每2周1次),疾病控制率(disease control rate,DCR)为44%,其中8例疾病稳定(stable disease,SD),6例PD,4例死亡;18例患者的中位总生存期(overall survival,OS)为30周,中位无进展生存期(progression-free survival,PFS)为15周,中位随访时间为30周。另有研究评估了Nivolumab在20例铂耐药卵巢癌患者中的安全性和抗肿瘤活性,分为低剂量组(1 mg/kg,静脉滴注,每2周1次)和高剂量组(3 mg/kg,静脉滴注,每2周1次),20例患者的总缓解率(overall response,OR)为15%,其中包括2例持续CR患者(3 mg/kg),DCR为45%。该研究终止时中位PFS为3.5个月(1.7~3.9个月),中位OS为20.0个月(7.0个月~∞)[10]。该研究中,心律失常的发生率高于以前对其他实体瘤展开的更大规模的研究,但心律失常均为1级或2级,并且是可控的,同时并未发现在其他研究中出现的严重免疫相关不良事件的肺炎或结肠炎。该研究证明了Nivolumab治疗铂耐药性卵巢癌患者的安全性和临床疗效。

由于单药治疗的局限性,研究者开始着力于PD-1/PD-L1抑制剂联合疗法。Zamarin等[11]进行了一项Ⅱ期临床试验,比较Nivolumab单药与Nivolumab及Ipilimumab(CTLA-4抑制剂)联合用药对于复发性或者耐药性卵巢癌患者的临床疗效差异。100例患者随机分配接受Nivolumab(n=49)或Nivolumab加Ipilimumab(n=51),在Nivolumab单药治疗组中,6例疾病缓解(12.2%),在联合用药组中16例缓解(31.4%)。单药和联合组的中位PFS分别为2个月和3.9个月。与Nivolumab单药治疗相比,联合用药组的进展或死亡风险显著降低,分层风险比为0.53(0.34~0.82),死亡危险比分别为0.79(0.44~1.42)。该研究说明Nivolumab和Ipilimumab联合用药与单独使用Nivolumab相比有更高的应答率,PFS未达到。Liu等[12]对38例复发性上皮性卵巢癌患者进行了一项单臂Ⅱ期临床试验,该试验评价了联合使用Nivolumab和Bevacizumab(血管内皮生长因子抑制剂)在复发性上皮性卵巢癌中的疗效,其中包括18例铂耐药患者和20例铂敏感患者,总ORR为28.9%,铂敏感患者ORR为40.0%(19.1%~64.0%),铂耐药患者ORR为16.7%(3.6%~41.4%)。在疾病发生进展的患者中,中位PFS为8.1个月(6.3~14.7个月)。该研究提示Nivolumab与Bevacizumab联合用药在复发性上皮性卵巢癌患者中是可行的,尤其是对于铂敏感的复发性上皮性卵巢癌患者,而对于铂耐药患者可能需要选择其他的治疗方式。

2.2 Pembrolizumab治疗卵巢癌的临床进展

一项Ⅱ期研究评价了Pembrolizumab治疗晚期复发性卵巢癌患者的临床疗效,招募了2组患者。A组招募了285例患者,先前接受过1~3种治疗,无铂间隔或无治疗间隔为3~12个月;B组招募了91例患者,先前接受过4~6种治疗,无铂间隔或无治疗间隔≥3个月。2组均每3周静脉注射Pembrolizumab 200 mg,A组的ORR为7.4%,B组的ORR为9.9%,2组中位缓解持续时间(duration of response,DOR)分别为8.2个月和未达到,A组和B组的DCR分别为37.2%和37.4%,2组的中位PFS均为2.1个月,该研究证实Pembrolizumab单药在复发性卵巢癌患者中表现出一定的疗效,并且较高的PD-L1表达与较高的治疗反应有关,而既往治疗、铂敏感程度与疗效无关[13]。另一项临床研究纳入的26例PD-L1阳性晚期转移性卵巢癌患者接受了Pembrolizumab治疗(每2周10 mg/kg);中位随访时间为15.4个月,ORR为11.5%(1例CR,2例PR);7例SD患者(26.9%)。中位PFS和OS分别为1.9(1.8~3.5)个月和13.8(6.7~18.8)个月,该临床试验说明Pembrolizumab在晚期PD-L1阳性卵巢癌患者中具有持久的抗肿瘤活性,并具有可控的安全性和毒性[14]

一项开放性单臂非随机Ⅱ期临床研究纳入了40例卵巢癌患者,接受Pembrolizumab、Bevacizumab和环磷酰胺治疗,3例CR(7.5%),16例PR(40.0%),19例SD(47.5%),ORR为47.5%,有38例(95.0%)临床获益(CR+PR+SD)患者,10例(25.0%)持续缓解(DOR>12个月)患者,中位PFS为10.0(6.5~17.4)个月[15]。该研究表明,Pembrolizumab与Bevacizumab联合口服环磷酰胺的耐受性良好,这种联合用药可能为复发性卵巢癌提供新的治疗策略与思路。同时,另一项开放性单臂的Ⅰ期和Ⅱ期临床研究纳入了62例卵巢癌患者,评估Niraparib[多腺苷二磷酸核糖聚合酶(PARP)抑制剂]和Pembrolizumab的联合治疗效果,治疗方案为:每天口服Niraparib 200 mg,每21天静脉注射200 mg Pembrolizumab。ORR为18%(11%~29%),DCR为65%(54%~75%),包括3例CR(5%),8例PR(13%),28例SD(47%)和20例PD(33%)。提示对于治疗方案有限的卵巢癌患者,无论其铂耐药或者敏感,生物标志物状态或先前是否使用Bevacizumab治疗,联合治疗的抗肿瘤疗效是肯定的[16]。Lee等[17]进行的一项单臂多中心的Ⅱ期临床试验纳入26例患者,治疗方案为: Pembrolizumab每3周 200 mg,多柔比星脂质体每4周40 mg/m2,均为静脉给药。23例患者可评估为最佳总体缓解,12例(52.2%)患者获得临床获益,有5例PR(21.7%)和1例CR(4.3%),ORR为26.1%,6例SD持续至少24周。该研究提示组合疗法耐受良好,没有额外的毒性,表明Pembrolizumab和多柔比星脂质体的联合治疗是可行的,该联合疗法提供了铂类耐药性卵巢癌的初步证据,且与既往研究比较,联合治疗的ORR和中位PFS高于单独使用多柔比星脂质体或PD-1/PD-L1抑制剂。

2.3 Atezolizumab治疗卵巢癌的临床进展

一项Ⅰ期多中心的临床试验研究了单药Atezolizumab在复发性上皮性卵巢癌或子宫恶性肿瘤患者中的作用,该研究招募了12例卵巢癌患者,Atezolizumab单一疗法在上皮性卵巢癌患者中耐受性良好,可能具有临床活性,值得进一步研究[18]。最近Atezolizumab的联合疗法也在开展中。在一项开放性多中心Ⅰb期研究中,20例铂耐药卵巢癌患者接受了治疗,给药方案为Atezolizumab 1 200 mg和Bevacizumab 15 mg/kg,ORR为15%;8例SD(40%),DCR为55%,中位DOR为未达到(11.3~∞),中位PFS为4.9个月(1.2~20.2个月),中位OS为10.2个月(1.2~26.6个月)。该研究说明Atezolizumab联合Bevacizumab在一些铂耐药卵巢癌患者中产生持久的缓解和(或)疾病稳定[19]。天津医科大学的一项回顾性研究分析了天津市肿瘤医院妇科肿瘤科的124例上皮性卵巢癌患者,这些患者由于结束铂类治疗后不到6个月而被诊断出具有顺铂耐药性,细胞功能测定表明,Atezolizumab联合Bevacizumab体外协同抑制顺铂耐药卵巢癌细胞系A2780cis的增殖、迁移和侵袭,这可能与Bevacizumab通过靶向信号转导及转录激活因子3(signal transducer and activator of transcription 3,STAT3)而抑制上皮-间质转化(epithelial-mesenchymal transition,EMT)和PD-L1的表达有关。此外,Bevacizumab和Atezolizumab在体内协同诱导抗肿瘤作用[20]。这些发现为顺铂耐药的复发性卵巢癌患者提供了新型治疗策略。

2.4 Durvalumab治疗卵巢癌的临床进展

Lampert等[21]进行了PARP抑制剂Olaparib和PD-L1抑制剂Durvalumab联合治疗复发性卵巢癌的一项单中心Ⅱ期试验,该试验招募的35例复发性卵巢癌患者接受了≥1个周期的治疗(每天接受2次Olaparib 300 mg,每4周接受1次Durvalumab 1 500 mg静脉注射,1个治疗周期为28 d),ORR为14%(5/35;4.8%~30.3%),DCR为71%(25/35;53.7%~85.4%),在30例铂耐药患者中有3例(10%)DOR为17.2个月,7例(23.3%)获得了至少6个月的疾病稳定期。这表明在先前已经接受过多种治疗的铂耐药患者中,相比于PARP抑制剂或免疫检查点阻断单一疗法显示出的有限活性,联合治疗可显示出持久的临床益处(≥6个月)。另外,该研究发现,这种治疗方式引起γ干扰素(IFN-γ)和CXCL9/CXCL10(CXC chemokine ligand)表达增加、全身IFN-γ/肿瘤坏死因子(TNF)的产生和肿瘤浸润淋巴细胞增多,这表明存在免疫刺激环境。IFN-γ表达增加与PFS延长有关,风险比(hazard ratio,HR)为0.37(95%CI:0.16~0.87,P=0.023),而血管内皮生长因子受体3(vascular endothelial growth factor receptor 3,VEGFR3)水平升高与PFS缩短相关(HR=3.22,95%CI:1.23~8.40,P=0.017)。这表明阻断血管内皮生长因子(vascular endothelial growth factor,VEGF)/VEGFR通路对提高PARP抑制剂和PD-L1阻断联合用药的疗效可能是必要的。Lee等[22]2015年6月—2016年5月对26例复发性女性生殖系统恶性肿瘤进行了Durvalumab联合Olaparib或Cediranib(VEGFR抑制剂)治疗的Ⅰ期试验,其中卵巢癌是最多的肿瘤类型(19/26,73%)。治疗方案为Durvalumab以10 mg/kg每2周或1 500 mg每4周1次静脉注射,Olaparib 300 mg每日2次,Cediranib 20 mg连续服用5 d/停用2 d,接受Durvalumab和Olaparib联合治疗的12例患者出现了2例PR(15个月和11个月)和8例SD(4个月),DCR为83%。在接受Durvalumab和Cediranib治疗的14例患者中,有12例可评估患者,其中观察到6例PR(5~8个月)和3例SD(4~8个月),疾病缓解率为50%,DCR为75%。但是治疗反应与PD-L1表达无关。这是首次报道的抗PD -L1与Olaparib、Cediranib联合治疗复发性女性癌症的研究。该课题组后续进行了一项Olaparib、Cediranib与Durvalumab联合治疗女性复发性癌症的Ⅰ期研究,该实验招募了9例患者(7例卵巢癌、1例子宫内膜癌、1例三阴性乳腺癌),其中4例PR(44%),3例SD(33%),持续≥6个月,临床获益率为67%,该研究初步证明,在之前已接受过多种治疗的复发性妇科癌症群体中,3种药物的联合治疗对没有生殖系BRCA突变的患者是可耐受的和有效的[23]。Zamarin等[24]进行了关于Durvalumab与叶酸受体α疫苗TPIV200联合治疗晚期卵巢癌患者的一项Ⅱ期试验,共纳入27例患者。Durvalumab 750 mg患者在1~12个治疗周期的第1天和第15天静脉给药,TPIV200(500 μg肽段)与粒细胞-巨噬细胞集落刺激因子(GM-CSF)(125 μg)混合后在第1~6个治疗周期的第1天肌内注射,28 d为1个治疗周期。9例(33%)SD,DCR为37%(24.4%~100%),PFS为2.8个月(2.5个月~∞), 6个月PFS率(6- month PFS rate)为11.1%(95%CI:4.9%~100%)。由于所有患者最终都经历了疾病进展,因此没有进行PFS分析。尽管联合用药疗效有限,但OS为21个月(13.5个月~∞),中位随访时间为29个月,12个月时的OS为66%(52.9%~100%),在这种已经接受过多种治疗的卵巢癌患者中,持久的存活时间突出了研究叶酸受体α疫苗接种对卵巢癌治疗的必要性。

2.5 Avelumab治疗卵巢癌的临床进展

一项招募了125例铂耐药的晚期卵巢癌患者的Ⅰ期临床试验评估了Avelumab单药的疗效和安全性,所有患者接受Avelumab(每2周10 mg/kg)的中位时间为2.8个月(0.5~27.4个月),中位随访时间为26.6个月(16~38个月),12例客观缓解(9.6%,95%CI:5.1%~16.2%),包括1例CR(0.8%)和11例PR(8.8%)。1年PFS率为10.2%(95%CI:5.4%~16.7%),中位OS为11.2个月(95%CI:8.7~15.4个月)。该研究提示Avelumab在接受过多种前期治疗的复发性或难治性卵巢癌患者中显示出抗肿瘤活性和可接受的安全性[25]。目前还有研究者正在进行一项随机三臂Ⅲ期试验,旨在比较铂耐药/难治性复发性卵巢癌,单独以及联合使用Avelumab、多柔比星脂质体对于铂耐药/难治性复发性卵巢癌的临床疗效[26]。目前的证据表明Avelumab单药治疗或联合化疗均可改善铂类难治性/耐药性患者的PFS或OS,但是仍需要更大样本量的临床资料证实。

3 结语与展望

PD-1/PD-L1抑制剂疗法是近年卵巢癌治疗的新模式,尤其在复发性/耐药性卵巢癌中显示出一定的抗肿瘤活性。现阶段研究者已经不局限于PD-1/PD-L1单药治疗卵巢癌,联合用药获得较好的进展。主要的联合用药种类有PARP抑制剂、VEGF抑制剂、CTLA-4抑制剂、环磷酰胺、多柔比星脂质体和特殊疫苗制剂等,但如何恰当使用该疗法对于临床医生仍是一个挑战。

大多数联合用药的临床研究表明没有新的与药物有关的不良反应,有研究表明联合用药治疗反应优于单独用药,甚至获得持久反应,这可能为卵巢癌的免疫治疗尤其是PD-1/PD-L1抑制剂的治疗提供了新思路。但是目前的临床研究主要针对于耐药/复发/化疗后的晚期卵巢癌患者,尚未发现对于早期卵巢癌患者的研究,对于这些患者应用PD-1/PD-L1抑制剂可能会有不同的结论。另外,PD-1/PD-L1抑制剂与化疗或者靶向药物的联合治疗用药种类繁多,对于特定的联合治疗药物组合尚无大规模的研究。目前PD-1/PD-L1免疫治疗相关研究正在进行中,相信在不久的将来免疫治疗将在卵巢肿瘤的治疗中发挥越来越重要的作用,为卵巢癌患者带来新的希望。

参考文献

Nash Z, Menon U.

Ovarian cancer screening: Current status and future directions

[J]. Best Pract Res Clin Obstet Gynaecol, 2020,65:32-45. doi: 10.1016/j.bpobgyn.2020.02.010.

DOI:10.1016/j.bpobgyn.2020.02.010      URL     [本文引用: 1]

Bray F, Ferlay J, Soerjomataram I, et al.

Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries

[J]. CA Cancer J Clin, 2018,68(6):394-424. doi: 10.3322/caac.21492.

DOI:10.3322/caac.21492      URL     [本文引用: 1]

Sharma P, Hu-Lieskovan S, Wargo JA, et al.

Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy

[J]. Cell, 2017,168(4):707-723. doi: 10.1016/j.cell.2017.01.017.

DOI:10.1016/j.cell.2017.01.017      URL     [本文引用: 1]

Li J, Stanger BZ.

How Tumor Cell Dedifferentiation Drives Immune Evasion and Resistance to Immunotherapy

[J]. Cancer Res, 2020,80(19):4037-4041. doi: 10.1158/0008-5472.CAN-20-1420.

DOI:10.1158/0008-5472.CAN-20-1420      URL     [本文引用: 1]

Spranger S, Gajewski TF.

Mechanisms of tumor cell-intrinsic immune evasion

[J]. Annual Review Cancer Biology, 2018,2(1):213-228. doi: 10.1146/annurev-cancerbio-030617-050606.

DOI:10.1146/annurev-cancerbio-030617-050606      URL     [本文引用: 1]

Hartnett EG, Knight J, Radolec M, et al.

Immunotherapy Advances for Epithelial Ovarian Cancer

[J]. Cancers (Basel), 2020,12(12):3733. doi: 10.3390/cancers12123733.

DOI:10.3390/cancers12123733      URL     [本文引用: 1]

Duan M, Fang M, Wang C, et al.

LncRNA EMX2OS Induces Proliferation, Invasion and Sphere Formation of Ovarian Cancer Cells via Regulating the miR-654-3p/AKT3/PD-L1 Axis

[J]. Cancer Manag Res, 2020,12:2141-2154. doi: 10.2147/CMAR.S229013.

DOI:10.2147/CMAR.S229013      URL     [本文引用: 1]

Matsuo K, Spragg SE, Ciccone MA, et al.

Nivolumab use for BRCA gene mutation carriers with recurrent epithelial ovarian cancer: A case series

[J]. Gynecol Oncol Rep, 2018,25:98-101. doi: 10.1016/j.gore.2018.06.011.

DOI:10.1016/j.gore.2018.06.011      [本文引用: 1]

Normann MC, Türzer M, Diep LM, et al.

Early experiences with PD-1 inhibitor treatment of platinum resistant epithelial ovarian cancer

[J]. J Gynecol Oncol, 2019,30(4):e56. doi: 10.3802/jgo.2019.30.e56.

DOI:10.3802/jgo.2019.30.e56      URL     [本文引用: 1]

Hamanishi J, Mandai M, Ikeda T, et al.

Safety and Antitumor Activity of Anti-PD-1 Antibody, Nivolumab, in Patients With Platinum-Resistant Ovarian Cancer

[J]. J Clin Oncol, 2015,33(34):4015-4022. doi: 10.1200/JCO.2015.62.3397.

DOI:10.1200/JCO.2015.62.3397      PMID:26351349      [本文引用: 1]

Programmed death-1 (PD-1), a coinhibitory immune signal receptor expressed in T cells, binds to PD-1 ligand and regulates antitumor immunity. Nivolumab is an anti-PD-1 antibody that blocks PD-1 signaling. We assessed the safety and antitumor activity of nivolumab in patients with platinum-resistant ovarian cancer.Twenty patients with platinum-resistant ovarian cancer were treated with an intravenous infusion of nivolumab every 2 weeks at a dose of 1 or 3 mg/kg (constituting two 10-patient cohorts) from October 21, 2011. This phase II trial defined the primary end point as the best overall response. Patients received up to six cycles (four doses per cycle) of nivolumab treatment or received doses until disease progression occurred. Twenty nivolumab-treated patients were evaluated at the end of the trial on December 7, 2014.Grade 3 or 4 treatment-related adverse events occurred in eight (40%) of 20 patients. Two patients had severe adverse events. In the 20 patients in whom responses could be evaluated, the best overall response was 15%, which included two patients who had a durable complete response (in the 3-mg/kg cohort). The disease control rate in all 20 patients was 45%. The median progression-free survival time was 3.5 months (95% CI, 1.7 to 3.9 months), and the median overall survival time was 20.0 months (95% CI, 7.0 months to not reached) at study termination.This study, to our knowledge, is the first to explore the effects of nivolumab against ovarian cancer. The encouraging safety and clinical efficacy of nivolumab in patients with platinum-resistant ovarian cancer indicate the merit of additional large-scale investigations (UMIN Clinical Trials Registry UMIN000005714).© 2015 by American Society of Clinical Oncology.

Zamarin D, Burger RA, Sill MW, et al.

Randomized Phase II Trial of Nivolumab Versus Nivolumab and Ipilimumab for Recurrent or Persistent Ovarian Cancer: An NRG Oncology Study

[J]. J Clin Oncol, 2020,38(16):1814-1823. doi: 10.1200/JCO.19.02059.

DOI:10.1200/JCO.19.02059      URL     [本文引用: 1]

Liu JF, Herold C, Gray KP, et al.

Assessment of Combined Nivolumab and Bevacizumab in Relapsed Ovarian Cancer: A Phase 2 Clinical Trial

[J]. JAMA Oncol, 2019,5(12):1731-1738. doi: 10.1001/jamaoncol.2019.3343.

DOI:10.1001/jamaoncol.2019.3343      URL     [本文引用: 1]

Matulonis UA, Shapira-Frommer R, Santin AD, et al.

Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: results from the phase II KEYNOTE-100 study

[J]. Ann Oncol, 2019,30(7):1080-1087. doi: 10.1093/annonc/mdz135.

DOI:S0923-7534(19)31241-4      PMID:31987374      [本文引用: 1]

Advanced recurrent ovarian cancer (ROC) is the leading cause of gynecologic cancer-related death in developed countries and new treatments are needed. Previous studies of immune checkpoint blockade showed low objective response rates (ORR) in ROC with no identified predictive biomarker.This phase II study of pembrolizumab (NCT02674061) examined two patient cohorts with ROC: cohort A received one to three prior lines of treatment with a platinum-free interval (PFI) or treatment-free interval (TFI) between 3 and 12months and cohort B received four to six prior lines with a PFI/TFI of ≥3months. Pembrolizumab 200mg was administered intravenously every 3weeks until cancer progression, toxicity, or completion of 2years. Primary end points were ORR by Response Evaluation Criteria in Solid Tumors version 1.1 per blinded independent central review by cohort and by PD-L1 expression measured as combined positive score (CPS). Secondary end points included duration of response (DOR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and safety.Cohort A enrolled 285 patients; the first 100 served as the training set for PD-L1 biomarker analysis. Cohort B enrolled 91 patients. ORR was 7.4% for cohort A and 9.9% for cohort B. Median DOR was 8.2months for cohort A and not reached for cohort B. DCR was 37.2% and 37.4%, respectively, in cohorts A and B. Based on the training set analysis, CPS 1 and 10 were selected for evaluation in the confirmation set. In the confirmation set, ORR was 4.1% for CPS<1, 5.7% CPS ≥1, and 10.0% for CPS ≥10. PFS was 2.1months for both cohorts. Median OS was not reached for cohort A and was 17.6months for cohort B. Toxicities were consistent with other single-agent pembrolizumab trials.Single-agent pembrolizumab showed modest activity in patients with ROC. Higher PD-L1 expression was correlated with higher response.Clinicaltrials.gov, NCT02674061.© 2019 THE AUTHORS. Published by Elsevier Ltd on behalf of the European Society for Medical Oncology. This is an open access article under the CC-BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Varga A, Piha-Paul S, Ott PA, et al.

Pembrolizumab in patients with programmed death ligand 1-positive advanced ovarian cancer: Analysis of KEYNOTE-028

[J]. Gynecol Oncol, 2019,152(2):243-250. doi: 10.1016/j.ygyno.2018.11.017.

DOI:10.1016/j.ygyno.2018.11.017      URL     [本文引用: 1]

Zsiros E, Lynam S, Attwood KM, et al.

Efficacy and Safety of Pembrolizumab in Combination With Bevacizumab and Oral Metronomic Cyclophosphamide in the Treatment of Recurrent Ovarian Cancer: A Phase 2 Nonrandomized Clinical Trial

[J]. JAMA Oncol, 2021,7(1):78-85. doi: 10.1001/jamaoncol.2020.5945.

DOI:10.1001/jamaoncol.2020.5945      URL     [本文引用: 1]

Konstantinopoulos PA, Waggoner S, Vidal GA, et al.

Single-Arm Phases 1 and 2 Trial of Niraparib in Combination With Pembrolizumab in Patients With Recurrent Platinum-Resistant Ovarian Carcinoma

[J]. JAMA Oncol, 2019,5(8):1141-1149. doi: 10.1001/jamaoncol.2019.1048.

DOI:10.1001/jamaoncol.2019.1048      [本文引用: 1]

ImportancePatients with recurrent ovarian carcinoma frequently develop resistance to platinum-based chemotherapy, at which time treatment options become limited. ObjectiveTo evaluate the poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor niraparib combined with pembrolizumab in patients with recurrent ovarian carcinoma. Design, Setting, and ParticipantsThe TOPACIO/KEYNOTE-162 (Niraparib in Combination With Pembrolizumab in Patients With Triple-Negative Breast Cancer or Ovarian Cancer) trial, an open-label, single-arm phases 1 and 2 study enrolled women with advanced or metastatic triple-negative breast cancer (TNBC) or recurrent ovarian carcinoma, irrespective of BRCA mutation status. Median follow-up was 12.4 months (range, 1.2 to >= 23.0 months). Data were collected from April 15, 2016, through September 4, 2018, with September 4, 2018, as a data cutoff, and analyzed from September 4, 2018, through January 30, 2019. InterventionsThe recommended phase 2 dose (RP2D) was 200 mg of oral niraparib once daily and 200 mg of intravenous pembrolizumab on day 1 of each 21-day cycle. Main Outcomes and MeasuresThe primary objectives of phase 1 were to evaluate dose-limiting toxic effects and establish the RP2D and dosing schedule. The primary objective of phase 2 was to assess objective response rate (ORR; complete plus partial responses). Results from the phase 1 ovarian carcinoma and TNBC cohorts and phase 2 ovarian carcinoma cohort are reported. Because of the similarity in the phase 1 and 2 ovarian carcinoma populations, the data were pooled to perform an integrated efficacy analysis. ResultsFourteen patients (9 with ovarian carcinoma and 5 with TNBC) in phase 1 and 53 patients with ovarian carcinoma in phase 2 were enrolled, for a pooled ovarian carcinoma cohort of 62 patients (median age, 60 years [range, 46-83 years]). In the integrated efficacy phases 1 and 2 ovarian carcinoma population (60 of 62 evaluable patients), ORR was 18% (90% CI, 11%-29%), with a disease control rate of 65% (90% CI, 54%-75%), including 3 (5%) with confirmed complete responses, 8 (13%) with confirmed partial responses, 28 (47%) with stable disease, and 20 (33%) with progressive disease. The ORRs were consistent across subgroups based on platinum-based chemotherapy sensitivity, previous bevacizumab treatment, or tumor BRCA or homologous recombination deficiency (HRD) biomarker status. Median duration of response was not reached (range, 4.2 to >= 14.5 months). At data cutoff, 2 patients with a response and 1 patient with stable disease continued to receive treatment. Conclusions and RelevanceNiraparib in combination with pembrolizumab is tolerable, with promising antitumor activity for patients with ovarian carcinoma who have limited treatment options regardless of platinum status, biomarker status, or prior treatment with bevacizumab. Responses in patients without tumor BRCA mutations or non-HRD cancers were higher than expected with either agent as monotherapy. Trial RegistrationClinicalTrials.gov identifier: NCT02657889

Lee EK, Xiong N, Cheng SC, et al.

Combined pembrolizumab and pegylated liposomal doxorubicin in platinum resistant ovarian cancer: A phase 2 clinical trial

[J]. Gynecol Oncol, 2020,159(1):72-78. doi: 10.1016/j.ygyno.2020.07.028.

DOI:10.1016/j.ygyno.2020.07.028      [本文引用: 1]

Liu JF, Gordon M, Veneris J, et al.

Safety, clinical activity and biomarker assessments of atezolizumab from a Phase I study in advanced/recurrent ovarian and uterine cancers

[J]. Gynecol Oncol, 2019,154(2):314-322. doi: 10.1016/j.ygyno.2019.05.021.

DOI:10.1016/j.ygyno.2019.05.021      URL     [本文引用: 1]

Moroney JW, Powderly J, Lieu CH, et al.

Safety and Clinical Activity of Atezolizumab Plus Bevacizumab in Patients with Ovarian Cancer: A Phase Ib Study

[J]. Clin Cancer Res, 2020,26(21):5631-5637. doi: 10.1158/1078-0432.CCR-20-0477.

DOI:10.1158/1078-0432.CCR-20-0477      URL     [本文引用: 1]

Zhang L, Chen Y, Li F, et al.

Atezolizumab and Bevacizumab Attenuate Cisplatin Resistant Ovarian Cancer Cells Progression Synergistically via Suppressing Epithelial-Mesenchymal Transition

[J]. Front Immunol, 2019,10:867. doi: 10.3389/fimmu.2019.00867.

DOI:10.3389/fimmu.2019.00867      PMID:31105696      [本文引用: 1]

The AURELIA trial demonstrated that adding Bevacizumab to chemotherapy significantly improved progression-free survival (PFS) for platinum resistant recurrent ovarian cancer. Recently, immunotherapy also presented potential anti-tumor effects in several malignant solid tumors. This study aimed to investigate whether combining anti-PD-L1 Atezolizumab with BEV may have a synergistic effect and enhance the efficacy of both treatments in cisplatin resistant epithelial ovarian cancer (CREOC). We retrospectively analyzed 124 epithelial ovarian cancer (EOC) patients from Gynecologic Oncology Department of Tianjin Cancer Hospital between January 2013 and June 2018, who all were diagnosed with cisplatin resistance due to progressing <6 months after completing platinum-based therapy. Based on responding to at least 2 cycles of Bevacizumab-containing chemotherapy (BC), these Patients were divided into BC response group and BC non-response group. Immunohistochemistry was used to detect that PD-L1 expression and tumor angiogenesis-related proteins (VEGF and Semaphorin4D) in tissues from 124 patients with CREOC. The positive expressions of PD-L1, VEGF, and Semaphorin4D (SEMA4D) were found in 58.73, 50.79, and 71.43% of the 63 cases CREOC tissues with BC response, respectively, which were significantly higher than that in the 61 cases BC non-response group (< 0.05). PD-L1 expression correlated with SEMA4D and VEGF positively ( = 0.344 and 0.363, < 0.001). Over-expressions of PD-L1, VEGF and SEMA4D are associated with more malignant clinicopathologic characteristics of CREOC Patients. In survival analysis, patients' response to BC was the independent factor for evaluation of PFS and overall survival (OS). Cell functional assays showed that Atezolizumab in combination with Bevacizumab inhibited the proliferation, migration, and invasion of cisplatin resistant ovarian cancer cell line A2780cis synergistically, which maybe associate with Bevacizumab suppressing the epithelial-mesenchymal transition (EMT) and PD-L1 expression by targeting STAT3. Furthermore, Bevacizumab and Atezolizumab induced synergistic anti-tumor effect. These findings suggest a novel therapeutic strategy for cisplatin resistant recurrent EOC and its mechanism warrants further study.

Lampert EJ, Zimmer A, Padget M, et al.

Combination of PARP Inhibitor Olaparib, and PD-L1 Inhibitor Durvalumab, in Recurrent Ovarian Cancer: a Proof-of-Concept Phase II Study

[J]. Clin Cancer Res, 2020,26(16):4268-4279. doi: 10.1158/1078-0432.CCR-20-0056.

DOI:10.1158/1078-0432.CCR-20-0056      PMID:32398324      [本文引用: 1]

Preclinical studies suggest PARP inhibition (PARPi) induces immunostimulatory micromilieu in ovarian cancer thus complementing activity of immune checkpoint blockade. We conducted a phase II trial of PARPi olaparib and anti-PD-L1 durvalumab and collected paired fresh core biopsies and blood samples to test this hypothesis.In a single-center, proof-of-concept phase II study, we enrolled women aged ≥18 with recurrent ovarian cancer. All patients were immune checkpoint inhibitor-naïve and had measurable disease per RECISTv1.1, ECOG performance status 0-2, and adequate organ and marrow function. Patients received olaparib 300 mg twice daily and durvalumab 1,500 mg intravenously every 4 weeks until disease progression, unacceptable toxicity, or withdrawal of consent. Primary endpoint was overall response rate (ORR). Secondary objectives were safety and progression-free survival (PFS). Translational objectives included biomarker evaluation for relationships with clinical response and immunomodulatory effects by treatment.Thirty-five patients with ovarian cancer [median, four prior therapies (IQR, 2-5.5), predominantly platinum-resistant (86%), wild-type (77%)] received at least one full cycle of treatment. ORR was 14% [5/35; 95% confidence interval (CI), 4.8%-30.3%]. Disease control rate (PR+SD) was 71% (25/35; 95% CI, 53.7%-85.4%). Treatment enhanced and expression, systemic IFNγ/TNFα production, and tumor-infiltrating lymphocytes, indicating an immunostimulatory environment. Increased IFNγ production was associated with improved PFS [HR, 0.37 (95% CI, 0.16-0.87), = 0.023], while elevated VEGFR3 levels were associated with worse PFS (HR, 3.22 (95% CI, 1.23-8.40), = 0.017].The PARPi and anti-PD-L1 combination showed modest clinical activity in recurrent ovarian cancer. Our correlative study results suggest immunomodulatory effects by olaparib/durvalumab in patients and indicate that VEGF/VEGFR pathway blockade would be necessary for improved efficacy of the combination.©2020 American Association for Cancer Research.

Lee JM, Cimino-Mathews A, Peer CJ, et al.

Safety and Clinical Activity of the Programmed Death-Ligand 1 Inhibitor Durvalumab in Combination With Poly (ADP-Ribose) Polymerase Inhibitor Olaparib or Vascular Endothelial Growth Factor Receptor 1-3 Inhibitor Cediranib in Women′s Cancers: A Dose-Escalation, Phase I Study

[J]. J Clin Oncol, 2017,35(19):2193-2202. doi: 10.1200/JCO.2016.72.1340.

DOI:10.1200/JCO.2016.72.1340      URL     [本文引用: 1]

Zimmer AS, Nichols E, Cimino-Mathews A, et al.

A phase I study of the PD-L1 inhibitor, durvalumab, in combination with a PARP inhibitor, olaparib, and a VEGFR1-3 inhibitor, cediranib, in recurrent women′s cancers with biomarker analyses

[J]. J Immunother Cancer, 2019,7(1):197. doi: 10.1186/s40425-019-0680-3.

DOI:10.1186/s40425-019-0680-3      URL     [本文引用: 1]

Zamarin D, Walderich S, Holland A, et al.

Safety, immunogenicity, and clinical efficacy of durvalumab in combination with folate receptor alpha vaccine TPIV200 in patients with advanced ovarian cancer: a phase II trial

[J]. J Immunother Cancer, 2020,8(1):e000829. doi: 10.1136/jitc-2020-000829.

DOI:10.1136/jitc-2020-000829      URL     [本文引用: 1]

Disis ML, Taylor MH, Kelly K, et al.

Efficacy and Safety of Avelumab for Patients With Recurrent or Refractory Ovarian Cancer: Phase 1b Results From the JAVELIN Solid Tumor Trial

[J]. JAMA Oncol, 2019,5(3):393-401. doi: 10.1001/jamaoncol.2018.6258.

DOI:10.1001/jamaoncol.2018.6258      PMID:30676622      [本文引用: 1]

Current treatment options for progressive ovarian cancer provide limited benefit, particularly in patients whose disease has become resistant to platinum-based chemotherapy.To assess the efficacy and safety of avelumab, an anti-programmed death-ligand 1 agent, in a cohort of patients with previously treated recurrent or refractory ovarian cancer.In an expansion cohort of a phase 1b, open-label study (JAVELIN Solid Tumor), 125 patients with advanced ovarian cancer who had received chemotherapy including a platinum agent were enrolled between November 6, 2013, and August 27, 2015. Statistical analysis was performed from December 31, 2016, to October 9, 2018.Patients received avelumab, 10 mg/kg, every 2 weeks until disease progression, unacceptable toxic effects, or withdrawal from the study.Prespecified end points in this cohort included confirmed best overall response (per Response Evaluation Criteria In Solid Tumors, version 1.1), immune-related best overall response, duration of response, progression-free survival, overall survival, results of programmed death-ligand 1 expression-based analyses, and safety.A total of 125 women (median age, 62.0 years [range, 27-84 years]) who had received a median of 3 prior lines of treatment (range, 0-10) for advanced disease were enrolled in the study. Patients received avelumab for a median of 2.8 months (range, 0.5-27.4 months), with a median follow-up of 26.6 months (range, 16-38 months). A confirmed objective response occurred in 12 patients (9.6%; 95% CI, 5.1%-16.2%), including a complete response in 1 patient (0.8%) and a partial response in 11 patients (8.8%). The 1-year progression-free survival rate was 10.2% (95% CI, 5.4%-16.7%) and median overall survival was 11.2 months (95% CI, 8.7-15.4 months). Infusion-related reactions occurred in 25 patients (20.0%). Other frequent treatment-related adverse events (any grade event occurring in ≥10% of patients) were fatigue (17 [13.6%]), diarrhea (15 [12.0%]), and nausea (14 [11.2%]). Grade 3 or higher treatment-related adverse events occurred in 9 patients (7.2%), of which only the level of lipase increased (3 [2.4%]) occurred in more than 1 patient. Twenty-one patients (16.8%) had an immune-related adverse event of any grade. No treatment-related deaths occurred.Avelumab demonstrated antitumor activity and acceptable safety in heavily pretreated patients with recurrent or refractory ovarian cancer.ClinicalTrials.gov identifier: NCT01772004.

Pujade-Lauraine E, Fujiwara K, Dychter SS, et al.

Avelumab (anti-PD-L1) in platinum-resistant/refractory ovarian cancer: JAVELIN Ovarian 200 Phase III study design

[J]. Future Oncol, 2018,14(21):2103-2113. doi: 10.2217/fon-2018-0070.

DOI:10.2217/fon-2018-0070      PMID:29584456      [本文引用: 1]

Avelumab is a human anti-PD-L1 checkpoint inhibitor with clinical activity in multiple solid tumors. Here, we describe the rationale and design for JAVELIN Ovarian 200 (NCT02580058), the first randomized Phase III trial to evaluate the role of checkpoint inhibition in women with ovarian cancer. This three-arm trial is comparing avelumab administered alone or in combination with pegylated liposomal doxorubicin versus pegylated liposomal doxorubicin alone in patients with platinum-resistant/refractory recurrent ovarian, fallopian tube or peritoneal cancer. Eligible patients are not preselected based on PD-L1 expression and may have received up to three prior lines of chemotherapy for platinum-sensitive disease, but none for resistant disease. Overall survival and progression-free survival are primary end points, and secondary end points include biomarker evaluations and pharmacokinetics.

/

津ICP备05004273
版权所有 © 天津市医学科学技术信息研究所
地址:天津市和平区贵州路94号 邮编:300070 电话:022-23337521 E-mail:fuchanfence@gjfckx.ac.cn
本系统由北京玛格泰克科技发展有限公司设计开发