ASCO GI 2024|快速口头报告专场肝胆胰研究重磅公布

肿瘤瞭望消化时讯 发表时间:2024-01-30 16:00:12

编者按:2024年美国临床肿瘤学会胃肠道肿瘤研讨会(2024 ASCO Gastrointestinal Cancers Symposium,ASCO GI 2024)于美国当地时间2024年1月18~20日在旧金山召开。昨日ASCO GI官网披露了除LBA之外的所有研究,肝胆胰口头报告专场研究由已经整理发表的2篇中国讲者研究,和两篇尚未公布的LBA,后续将整理LBA研究,故不再整理口头报告专场肝胆胰研究。现整理已经公开的快速口头报告专场的肝胆胰研究,以飨读者。


NABNEC: A randomised phase II study of nab-paclitaxel in combination with carboplatin as first line treatment of gastrointestinal neuroendocrine carcinomas (GI-NECs)

NABNEC: 白蛋白紫杉醇联合卡铂作为胃肠道神经内分泌癌(GI-NECs)一线治疗的随机II期研究

摘要号:589

背景

神经内分泌癌是一种罕见的侵袭性肿瘤。迄今为止,尚无随机试验确定晚期G3胃肠道(GI) NENs的标准治疗方法。从小细胞肺癌的数据推断,标准做法是用依托泊苷和卡铂治疗G3 GI-NENs。紫杉醇在NECs中也有活性,但没有关于白蛋白紫杉醇作用的数据。NABNEC (ANZCTR # 12616000958482)旨在确定卡铂和白蛋白紫杉醇在晚期G3 GI-NENs中的活性、安全性和耐受性,并加深我们对此类NENs生物学和影像学特征的了解。

方法

NABNEC被设计为一项随机非对照II期研究,评估白蛋白紫杉醇在晚期和/或转移性不可切除G3 GI-NENs患者中的活性。统计计划是,实验组的46名可评估患者将给予80%的权重和95%的置信区间来排除30%的RR,以支持6个月时更有趣的50%的RR。修改方案3.0版本,在12例患者后暂停随机分配到对照组。主要终点为RECIST 1.1的客观缓解率(RR)。次要终点为无进展生存期、总生存期、NCI-CTCAE V4.03不良事件和生活质量(EORTC QLQC30、QLQ-GINET21问卷)。转化研究终点包括血液和组织生物标志物,包括循环肿瘤细胞谱、突变谱(全外显子组测序)和与临床终点相关的DNA甲基化谱。18-氟脱氧葡萄糖正电子发射断层扫描(FDG-PET)与早期反应的相关性也被评估。

结果

2016年9月至2021年12月期间,来自16个中心的60名患者,其中58名可评估患者,对照组12名,实验组46名。43例(75%)患者肿瘤Ki-67>55%;其余的Ki-67在20%到55%之间。约20%为胰腺原发;超过一半的患者有肝转移。NABNEC试验达到了主要终点。实验治疗组的RR为53%(38%—69%),对照组为42%(16%—71%)。两种治疗的无进展生存期无差异。实验组的24个月总生存率为25%,对照组为17%。白蛋白紫杉醇组3级毒性更差(52% vs 42%)。正在分析生活质量和第三阶段结果。

结论

卡铂联合白蛋白紫杉醇是治疗G3 GI-NENs的有效方案。需要在III期研究中进行进一步评估。

临床试验信息

ACTRN12616000958482


Alternating application of gemcitabine/nab-paclitaxel (Gem/nab-Pac) and Gem monotherapy or continuous application of Gem/nab-Pac after induction treatment for first-line treatment of metastatic pancreatic cancer (mPC): First results from the randomized phase 2 ALPACA study from the German AIO study group (AIO-PAK-0114)

交替应用吉西他滨/白蛋白紫杉醇(Gem/nab-Pac)和Gem单药治疗或在诱导治疗后继续应用Gem/nab-Pac用于转移性胰腺癌(mPC)的一线治疗:来自德国AIO研究组(AIO-PAK-0114)的随机2期ALPACA研究的初步结果

摘要号:605

背景

ALPACA研究的目的是研究在mPC患者经过3个月的诱导期后,Gem/nab-Pac和Gem单药交替治疗周期,与标准的Gem/nab-Pac连续治疗相比,是否可以改善总体耐受性,同时延长治疗时间和提高疗效。

方法

德国AIO研究组的随机、多中心2期ALPACA试验在一线环境中招募了确诊的mPC患者。在标准剂量Gem/nab-Pac的3个周期诱导期(Gem 1000 mg/m2和nab-Pac 125 mg/m2,28天1周期的第1、8、15天)后,患者被随机(1:1)分配到继续使用标准Gem/nab-Pac治疗,或接受标准剂量Gem/nab-Pac联合治疗和Gem单药治疗的交替使用(Gem 1000 mg/m2,28天1周期的第1、8、15天)。主要研究终点是随机化后的总生存期(OS)。该试验已在ClinicalTrials.gov注册,编号NCT02564146。

结果

2016年5月至2021年5月,来自德国29个中心的325名患者入组。6名入组患者没有在研究中开始治疗。诱导治疗3个周期后,174例(53.5%)患者可随机入组。过早退出研究的主要原因是死亡(24.8%)、疾病进展(22.8%)和不良事件(15.9%)。随机化后交替治疗组的中位OS与标准治疗组相当(10.5个月 vs. 10.4个月;HR 0.903,80%CI:0.723-1.128,P=0.5551)。同样,两组的中位无进展生存期相当(5.5个月 vs. 5.3个月;HR 0.767,95%CI:0.556-1.056,P=0.1017)。与标准治疗相比,交替治疗组的mPC患者的耐受性得到改善,特别是周围神经病变(所有级别,44.7% vs. 52.5%)和感染的发生(所有级别,29.4% vs. 47.5%)。两组随机分组后的治疗时间相似(3.25个月 vs. 3.02个月)。与连续Gem/nab-Pac治疗的患者相比,接受交替治疗的患者因不良事件退出研究的人数较少(14.9% vs 27.5%),这些患者因疾病进展而退出研究的频率略高(48.3% vs. 42.5%)。

结论

ALPACA试验表明,在标准Gem/nab-Pac诱导3个周期后,Gem/nab-Pac和Gem单药交替治疗的减剂量方案是可行的,其OS与标准治疗相当,同时耐受性提高。

临床试验信息NCT02564146


Hepatic arterial infusion pump chemotherapy in patients with advanced intrahepatic cholangiocarcinoma confined to the liver: A multicenter phase II trial

肝动脉灌注泵化疗治疗局限于肝脏的晚期肝内胆管癌:一项多中心II期试验

摘要号:433

背景

在ABC试验中,局限于肝脏的晚期肝内胆管癌(iCCA)患者接受全身吉西他滨联合顺铂治疗的3年总生存率(OS)仅为2.8%。最近的一项荟萃分析显示,肝动脉输注泵(HAIP)联合全身化疗的3年总OS为39.5%。HAIP化疗包括使用皮下泵将氟脲苷(FUDR)直接注入肝动脉。本研究的目的是前瞻性评估HAIP与全身化疗在荷兰局限于肝脏的晚期iCCA患者中的有效性。

方法

我们在荷兰的3个中心进行了一项单臂II期试验。如果以前没有用过氟脲苷,则安排6个周期的氟脲苷HAIP化疗,同时进行8个周期的吉西他滨和顺铂全身化疗。主要终点是OS,次要终点是无进展生存期(PFS)和客观反应。

结果

2020年1月至2022年9月,纳入50例晚期iCCA患者。HAIP联合全身化疗38例(76.0%)。11例患者(22.0%)单独接受HAIP化疗,因为他们在入组前接受过全身治疗。1例(2.0%)未开始治疗,因植入泵19天后因COVID-19死亡。中位随访时间为26.4个月(95% CI: 21.7 - 39.0)。中位生存期为22.1个月(95% CI: 19.7 -未达到)。1年OS率为80.0% (95% CI: 69.6% ~ 91.9%);3年OS率为28.6% (95% CI: 16.0% - 51.2%)。中位PFS为10.0个月(95% CI: 8.7 - 12.2)。27例患者(54.0%)获得客观影像学反应(RECIST), 43例患者(86.0%)在6个月时获得疾病控制。4例患者(8.0%)在HAIP化疗后进行了切除,其中2例患者有完全的病理反应。

结论

HAIP联合全身化疗治疗晚期iCCA患者的3年OS为28.6%,而在ABC试验中单独全身化疗患者的3年OS为2.8%。

临床试验信息NL8234


Efficacy and safety results of FGFR1-3 inhibitor, tinengotinib, as monotherapy in patients with advanced, metastatic cholangiocarcinoma: Results from phase II clinical trial

FGFR1-3抑制剂tinengotinib单药治疗晚期转移性胆管癌的疗效和安全性:来自II期临床试验的结果

摘要号:434

背景

Tinengotinib是一种选择性多激酶抑制剂,具有独特的FGFR结合特性,在临床前模型和包括胆管癌(CCA)患者(pts)的I期试验中有效抑制FGFR2融合/重排并获得耐药突变。在这里,我们在一项II期临床试验中介绍了tinengotinib的有效性和安全性。

方法

既往接受过≥1次全身化疗且ECOG PS为0或1分的晚期/转移性CCA患者接受tinengotinib 10 mg QD治疗。四个队列包括:队列A1: FGFR2融合,既往FGFR抑制剂(FGFRi)初始进展(原发性耐药),A2: FGFR2融合,既往FGFRi反应后进展(获得性耐药);B:非融合型FGFR改变;C: FGFR野生型(FGFRwt)。主要终点是RECIST v1.1的客观缓解率(ORR)。采用CTCAE V5.0进行安全性评价。

结果

截至2023年8月7日,纳入48例CCA患者,A1队列13例,A2队列10例,B队列12例,C队列13例。中位年龄61.5岁[范围25-81],41.7%为男性,58.3%既往治疗≥3线。47.9%为ECOG PS 0分。在35名FGFR改变的患者中,80.0%既往接受过≥1次FGFRi治疗,97.1%既往接受过化疗。40例患者的疗效可评估。在A1中,11名患者中有1名(9.1%)达到PR,肿瘤缩小31.8%。在A2中,8名患者中有3名(37.5%)达到了PR,肿瘤缩小率分别为40.7%、47.0%和54.6%。在B组,9名患者中有3名(33.3%)达到PR,肿瘤缩小率分别为36.5%、48.6%和60.6%。C组没有观察到PR。整体DCR为94.7% (18/19)在FGFR2融合/重组患者(A1 + A2), 88.9%(8/9)在其他FGFR改变患者(B),75% (9/12)在FGFRwt患者(C)。A1+ A2组的中位无进展生存期(mPFS)是5.26个月(95%CI:2.86-9.10),B组为5.98个月(95%CI:1.87-NA) 和C组3.84个月(95%CI:1.84-4.80)。48例患者中,45例(93.8%)患者出现治疗相关的AEs (TRAEs),14(29.2%)例患者为G1-2,29(60.4%)例患者为G3和2例(4.2%)为G4。最常见的G3 TRAEs是高血压12例(25%),掌跖感觉丧失性红斑综合征3例(6.3%),腹泻3例(6.3%)和口腔炎3例(6.3%)。G4 TRAEs中,一名患者为促甲状腺激素升高和脂肪酶升高,另一例患者为后部可逆性脑病综合征。未见G5 TRAE。

结论

Tinengotinib对于既往FGFRi经治的FGFR2融合CCA和非融合FGFR改变后CCA具有良好的临床益处。Tinengotinib相关的毒性是可控的。一项正在进行的随机对照III期研究将评估Tinengotinib对比医生选择的方案治疗既往化疗和FGFRi治疗后的FGFR2改变的难治性/复发性CCA患者的临床疗效、安全性和药效学效应。

临床试验信息NCT04919642


Atezolizumab plus chemotherapy with or without bevacizumab in advanced biliary tract cancer: Results from a randomized proof-of-concept phase II trial (IMbrave151).

阿替利珠单抗联合化疗加或不加贝伐珠单抗治疗晚期胆道癌:一项随机概念验证II期试验的结果(IMbrave151)

摘要号:435

背景

VEGF阻断联合细胞毒性化疗可以促进免疫允许的肿瘤微环境,增强对PD-L1抑制的反应。IMbrave151 (NCT04677504)是一项随机、双盲、全球概念验证II期研究,评估阿替利珠单抗(atezo)、贝伐单抗(bev)和顺铂和吉西他滨(CisGem)作为晚期胆道癌(aBTC)的一线治疗。在这里,我们报告最新的临床数据与反应和耐药的分子相关性。

方法

先前未治疗的aBTC患者(pts)按1:1随机分配,接受atezo(1200 mg,q3w) + bev(15 mg/kg,q3w)或安慰剂(plb) + CisGem (Cis 25 mg/m2和Gem 1000 mg/m2,第1天和第8天,q3w)长达8个周期,随后接受atezo (1200 mg q3w) + bev(15 mg/kg q3w)或plb,直到疾病进展或不可接受的毒性。主要终点为无进展生存期(PFS)。次要终点包括总生存期(OS)、客观缓解率(ORR)、缓解持续时间(DOR)和安全性。当死亡人数达到90例或所有患者随访时间≥2年时,进行最终OS分析。对基线肿瘤样本进行转录组分析(n=98)和突变谱分析(n=103)。这是一项寻求信号的试验,旨在评估每组的治疗效果,没有正式的假设检验。

结果

总共有162名患者被随机分配到atezo + bev + CisGem组(atezo/bev;n=79)或atezo + plb + CisGem组(atezo/plb;n= 83)。更新后的PFS HR为0.67 (95%CI:0.46~0.95),支持使用atezo/bev。atezo/bev组的更新中位PFS为8.35个月,atezo/plb组的更新中位PFS为7.9个月,6个月生存率分别为78%和63%。更新后的OS HR为0.97 (95%CI:0.64~1.47),atezo/bev组的中位OS为14.9个月,atezo/plb组的中位OS为14.6个月。atezo/bev组和atezo/plb组的确认ORR分别为26.6%和26.5%,中位DORs分别为10.28个月(95%CI: 6.7~16.7)和6.18个月(95%CI:4.3~6.7)。atezo/bev组3/4级不良事件发生率为73%,atezo/plb组为74%。VEGFA基因高表达与PFS(HR 0.43;95%CI:0.22~0.83)和OS(HR 0.66;95%CI:0.31~1.4)改善相关,支持使用atezo/bev。肝细胞高基因标记也与PFS(HR 0.47;95%CI: 0.24~0.92)和OS (HR 0.66;95%CI: 0.31~1.4)的改善相关,支持使用atezo/bev。在atezo/bev组,PI3k/AKT通路突变的患者的OS似乎比没有突变的患者更差(HR 3.7;95%CI:1.5~9.1)。

结论

IMbrave151研究的最终分析表明,在意向治疗患者中,atezo联合bev和化疗有适度的PFS获益。该试验受限于样本数量少和非比较性设计。相关生物标志物的探索性分析表明,高VEGF-A基因表达和肝细胞高基因标记可能是atezo/bev获益的预测标志物,值得进一步研究。

临床试验信息NCT04677504

摘要原文

(一)

NABNEC: A randomised phase II study of nab-paclitaxel in combination with carboplatin as first line treatment of gastrointestinal neuroendocrine carcinomas (GI-NECs)

Presenter:Lorraine A. Chantrill, PhD, MBBS, BS, FRACP | Wollongong Hospital, NSW

Abstract:589

Background:Neuroendocrine carcinomas (NEC) are rare and aggressive cancers. There are no randomised trials to date to establish standard therapy for advanced G3 gastrointestinal (GI) NENs. Extrapolating from small cell lung cancer data, standard practice is to treat G3 GI-NENs with etoposide and carboplatin. Paclitaxel is also active in NECs however there is no data on the role of nab-paclitaxel. NABNEC (ANZCTR # 12616000958482) aimed to determine the activity, safety and tolerability of carboplatin and nab-paclitaxel in advanced G3 GI-NENs and to enhance our understanding of the biology and imaging characteristics of such NENs.

Methods:NABNEC was designed as a randomised non-comparative phase II study, evaluating the activity of nab-paclitaxel in patients with advanced and/or metastatic non-resectable G3 GI-NENs. The statistical plan was that 46 evaluable patients in the experimental arm would give 80% power and 95% confidence to rule out a 30% RR in favour of a more interesting RR of 50% at 6 months. Protocol version 3.0 was amended and randomisation to the control arm was suspended after 12 patients. Primary endpoint was objective response rate (RR) by RECIST 1.1. Secondary endpoints were progression free survival, overall survival, adverse events by NCI-CTCAE V4.03 and quality of life (EORTC QLQC30, QLQ-GINET21 questionnaires). Translational research endpoints include blood and tissue biomarkers including circulating tumour cells profiling, mutation profile (whole exome sequencing) and DNA methylation profile correlated with clinical endpoints. Correlation of 18-fluoro-deoxyglucose positron emission tomography (FDG-PET) to early response has also been evaluated.

Results:60 patients were accrued between Sep 2016 and Dec 2021 from 16 centres, resulting in 58 evaluable patients, 12 (control arm), 46 (experimental arm). 43 patients (75%) had tumours with Ki-67>55%; the remainder had Ki-67 between 20 and 55%. About 20% had pancreatic primaries; more than half had liver metastases. The NABNEC trial met its primary endpoint. RR was 53% (38-69%) in the experimental treatment arm compared to 42% (16-71%) in the control group. Progression free survival was not different between the 2 treatments. 24m overall survival was 25% in the experimental group compared to 17% in the control. Grade 3 toxicity was worse in the nab-paclitaxel arms (52% v 42%). Quality of life and tertiary outcomes are being analysed.

Conclusions:The combination of carboplatin and nab-paclitaxel is an active regimen for G3 GI-NENs. Further evaluation in a Phase III study is warranted. Clinical trial information: ACTRN12616000958482.

(二)

Alternating application of gemcitabine/nab-paclitaxel (Gem/nab-Pac) and Gem monotherapy or continuous application of Gem/nab-Pac after induction treatment for first-line treatment of metastatic pancreatic cancer (mPC): First results from the randomized phase 2 ALPACA study from the German AIO study group (AIO-PAK-0114)

Presenter:Frank Kullmann, MD | Department of Medicine I, Klinikum Weiden

Abstract:605

Background:The aim of the ALPACA study was to investigate whether improved overall tolerability accompanied by prolonged treatment duration and increased efficacy can be achieved by alternating treatment cycles of Gem/nab-Pac and Gem monotherapy compared to standard continuous Gem/nab-Pac after a 3-month induction phase in patients with mPC.

Methods:The randomized, multicenter phase 2 ALPACA trial of the German AIO study group enrolled patients with confirmed mPC in the first-line setting. After an induction phase with 3 cycles of standard dose Gem/nab-Pac (Gem 1000 mg/m2 and nab-Pac 125 mg/m2 on d1, 8, 15 of each 28-day cycle), patients were randomized (1:1) either to continue treatment with standard Gem/nab-Pac, or to receive alternating cycles of standard dose Gem/nab-Pac combination therapy and Gem monotherapy (Gem 1000 mg/m2 on d1, 8, 15 of each 28-day cycle). Primary study endpoint was overall survival (OS) after randomization. The trial was registered with ClinicalTrials.gov, NCT02564146.

Results:Between May 2016 and May 2021, 325 patients were enrolled from 29 German centers. 6 of the enrolled patients did not start treatment within the study. Following 3 cycles of induction treatment, 174 patients (53.5%) could be randomized. Main reasons for premature dropout were death (24.8%), progression of disease (22.8%), and adverse events (15.9%). Median OS after randomization in the alternating treatment arm was comparable to the standard treatment arm (10.5 vs. 10.4 months; HR 0.903, 80% CI 0.723-1.128, p=0.5551). Likewise, median progression-free survival was comparable in both arms (5.5 vs. 5.3 months; HR 0.767, 95% CI 0.556-1.056, p=0.1017). Tolerability was improved for mPC patients treated with alternating cycles compared to standard therapy, especially regarding peripheral neuropathy (all grades, 44.7% vs. 52.5%), and occurrence of infections (all grades, 29.4% vs. 47.5%). Treatment duration after randomization was similar in both arms (3.25 vs. 3.02 months). Fewer patients receiving alternating treatment cycles dropped out of the study due to adverse events compared to patients treated with continuous Gem/nab-Pac (14.9% vs 27.5%). In return, these patients terminated the study slightly more often due to progression of disease (48.3% vs 42.5%).

Conclusions:The ALPACA trial suggests that a dose-reduced regimen with alternating cycles of Gem/nab-Pac and Gem monotherapy after 3 induction cycles of standard Gem/nab-Pac is feasible and associated with an OS comparable to standard treatment while resulting in improved tolerability. Clinical trial information: NCT02564146.

(三)

Hepatic arterial infusion pump chemotherapy in patients with advanced intrahepatic cholangiocarcinoma confined to the liver: A multicenter phase II trial

Presenter:Bas Groot Koerkamp, MD, PhD | Department of Surgery, Erasmus MC Cancer Institute

Abstract:433

Background:In the ABC-trials, the 3-year overall survival (OS) was only 2.8% for patients with advanced intrahepatic cholangiocarcinoma (iCCA) confined to the liver who received systemic gemcitabine with cisplatin. Hepatic arterial infusion pump (HAIP) combined with systemic chemotherapy had a pooled 3-year OS of 39.5% in a recent meta-analysis. HAIP chemotherapy involves continuous administration of floxuridine (FUDR) directly into the hepatic artery using a subcutaneous pump. The aim of this study was to prospectively assess the effectiveness of HAIP with systemic chemotherapy in patients with advanced iCCA confined to the liver in the Netherlands.

Methods:We performed a single arm phase II trial in 3 centers in the Netherlands. Six cycles of HAIP chemotherapy with floxuridine were scheduled with 8 cycles of concurrent systemic chemotherapy with gemcitabine and cisplatin, if not administered previously. The primary endpoint was OS, secondary endpoints were progression-free survival (PFS) and objective response.

Results:From January 2020 until September 2022, 50 patients with advanced iCCA were included. Combined HAIP and systemic chemotherapy was administered to 38 patients (76.0%). Eleven patients (22.0%) received HAIP chemotherapy alone, because they had received systemic treatment before enrollment. One patient (2.0%) didn’t start treatment, because he died 19 days after pump implantation due to COVID-19. The median follow-up was 26.4 months (95% CI: 21.7 – 39.0). The median OS was 22.1 months (95% CI: 19.7 – not reached). The 1-year OS rate was 80.0% (95% CI: 69.6% – 91.9%); the 3-year OS rate was 28.6% (95% CI: 16.0% – 51.2%). The median PFS was 10.0 months (95% CI: 8.7 – 12.2). An objective response on imaging (RECIST) was achieved in 27 patients (54.0%) and disease control at 6 months in 43 patients (86.0%). Four patients (8.0%) underwent a resection after HAIP chemotherapy of whom 2 patients had a complete pathologic response.

Conclusions:Combined HAIP with systemic chemotherapy for patients with advanced iCCA was associated with a favorable 3-year OS of 28.6% compared with 2.8% after systemic chemotherapy alone in the ABC trials. Clinical trial information: NL8234.

(四)

Efficacy and safety results of FGFR1-3 inhibitor, tinengotinib, as monotherapy in patients with advanced, metastatic cholangiocarcinoma: Results from phase II clinical trial

Presenter:Milind M. Javle, MD | Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center

Abstract:434

Background:Tinengotinib is a spectrum-selective multi-kinase inhibitor with unique binding properties to FGFR, potently inhibited FGFR2 fusion/rearrangement and acquired resistant mutations in pre-clinical models and in phase I trials that included cholangiocarcinoma (CCA) patients (pts). Here we present the efficacy and safety of tinengotinib in a phase II clinical trial.

Methods:Eligible pts with advanced/metastatic CCA who had received ≥ 1 prior systemic chemotherapy therapy and ECOG PS 0 or 1 were treated with tinengotinib 10 mg QD. Four cohorts included: Cohort A1:FGFR2 fusion(s) with primary progression on previous FGFR inhibitor (FGFRi), A2:FGFR2 fusion(s) with progression after prior response to FGFRi (acquired resistance); B: non-fusion FGFR alteration(s): C:FGFR wild-type (FGFRwt). Primary endpoint was objective response rate (ORR) per RECIST v1.1. CTCAE V5.0 was used for safety assessments.

Results:As of 7Aug2023, 48 pts with CCA were enrolled, 13 in Cohort A1, 10 in A2, 12 in B, 13 in C. Median age 61.5 [range 25-81] years old, 41.7% male, 58.3% had ≥ 3 lines of prior therapy. ECOG PS 0 in 47.9% pts. Among 35 pts with FGFR alterations, 80.0% had ≥ 1 prior FGFRi therapy, and 97.1% had prior chemotherapy. Forty (40) pts were efficacy evaluable. In A1, 1 out of 11 pts (9.1%) achieved PR with tumor reduction of 31.8%. In A2, 3 out of 8 pts (37.5%) achieved PR with tumor reduction of 40.7%, 47.0% and 54.6%. In B, 3 out of 9 pts (33.3%) achieved PR with tumor reduction of 36.5%, 48.6%, and 60.6%. No PR was observed in C. Overall DCR was 94.7% (18/19) in FGFR2 fusion/rearrangement pts (A1+A2), 88.9% (8/9) in other FGFR alterations pts (B), and 75% (9/12) in FGFRwt pts (C). Median progression-free survival (mPFS) was 5.26 months (95%CI, 2.86-9.10) in A1+A2, 5.98 months (95%CI, 1.87-NA) in B and 3.84 months (95% CI, 1.84-4.80) in C. Among 48 treated pts, treatment-related AEs (TRAEs) occurred in 45 (93.8%) pts, 14 (29.2%) in G1-2, 29 (60.4%) in G3 and 2 (4.2%) in G4. The most common G3 TRAEs were hypertension in 12 (25%), palmar-plantar erythrodysesthesia syndrome in 3 (6.3%), diarrhea in 3 (6.3%) and stomatitis in 3 (6.3%). One subject had G4 increased TSH and G4 increased lipase, and another subject had G4 posterior reversible encephalopathy syndrome. No G5 TRAE was observed.

Conclusions:Tinengotinib has promising clinical benefit for FGFR2 fusion CCA after prior FGFRi and for non-fusion FGFR alterations. Tinengotinib-related toxicities were manageable. An ongoing randomized, controlled phase III study will evaluate the clinical efficacy, safety, and pharmacodynamic effect of Tinengotinib vs Physicians’ choice in subjects with FGFR2-altered refractory/relapsed CCA after prior chemotherapy and FGFRi therapy. Clinical trial information: NCT04919642.

(五)

Atezolizumab plus chemotherapy with or without bevacizumab in advanced biliary tract cancer: Results from a randomized proof-of-concept phase II trial (IMbrave151).

Presenter:Anthony B. El-Khoueiry, MD | USC Norris Comprehensive Cancer Center

Abstract:435

Background:VEGF blockade coupled with cytotoxic chemotherapy can promote an immune-permissive tumor microenvironment that augments response to PD-L1 inhibition. IMbrave151 (NCT04677504) is a randomized, double-blinded, global proof-of-concept Phase II study evaluating atezolizumab (atezo), bevacizumab (bev) and cisplatin and gemcitabine (CisGem) as first-line treatment for advanced biliary tract cancer (aBTC). Here we report updated clinical data and molecular correlates of response and resistance.

Methods:Patients (pts) with previously untreated aBTC were randomized 1:1 to receive atezo (1200 mg every 3 weeks [q3w]) + bev (15 mg/kg q3w) or placebo (plb) + CisGem (Cis 25 mg/m2 and Gem 1000 mg/m2 on Days 1 and 8 q3w) for up to 8 cycles, followed by atezo (1200 mg q3w) + bev (15 mg/kg q3w) or plb until disease progression or unacceptable toxicity. The primary endpoint was progression-free survival (PFS). Secondary endpoints included overall survival (OS), objective response rate (ORR), duration of response (DOR), and safety. The final OS analysis was done when ≈90 deaths occurred or when all pts had ≥2 years of follow-up. Transcriptome analysis (n=98) and mutation profiling (n=103) were done on baseline tumor samples. This was a signal-seeking trial to estimate the treatment effect in each arm with no formal hypothesis testing.

Results:In total, 162 pts were randomized to either atezo + bev + CisGem (atezo/bev; n=79) or atezo + plb + CisGem (atezo/plb; n=83). The updated PFS HR was 0.67 (95% CI: 0.46, 0.95) in favor of atezo/bev. Updated median PFS was 8.35 mo for atezo/bev and 7.9 mo for atezo/plb, with 6-mo rates of 78% and 63%, respectively. The updated OS HR was 0.97 (95% CI: 0.64, 1.47), with a median of 14.9 mo for atezo/bev and 14.6 mo for atezo/plb. Confirmed ORR was 26.6% for atezo/bev and 26.5% for atezo/plb, with median DORs of 10.28 (95% CI: 6.7, 16.7) and 6.18 mo (95% CI: 4.3, 6.7), respectively. The incidence of Grade 3/4 adverse events was 73% with atezo/bev and 74% with atezo/plb. High VEGFA gene expression was associated with an improved PFS (HR, 0.43; 95% CI: 0.22, 0.83) and OS (HR, 0.66; 95% CI: 0.31, 1.4) in favor of atezo/bev. Hepatocytes high gene signature also was associated with an improved PFS (HR, 0.47; 95% CI: 0.24, 0.92) and OS (HR, 0.66; 95% CI: 0.31, 1.4) in favor of atezo/bev. Pts with PI3k/AKT pathway mutations appeared to have worse OS than pts without mutations (HR, 3.7; 95% CI: 1.5, 9.1) with atezo/bev.

Conclusions:Final analysis of the IMbrave151 study indicates a modest PFS benefit in intent-to-treat pts combining atezo with bev and chemotherapy. The trial is limited by small numbers and a non-comparative design. Exploratory analysis of correlative biomarkers suggests that high VEGF-A gene expression and hepatocytes high gene signature may be predictive markers of benefit with atezo/bev, warranting further investigation. Clinical trial information: NCT04677504.

发表评论

提交评论
  • 推荐文章