Abstract A071: Immune biomarkers of response in a phase 1 trial of combined MEK/STAT3/PD-1 inhibition in metastatic pancreatic ductal adenocarcinoma (PDAC)

IF 16.6 1区 医学 Q1 ONCOLOGY
Peter Joel. Hosein, Sayan Chakraborty, Anna Bianchi, Yolanda Justal, Nkiruka Ezenwajiaku, Maria Vanessa. Yow, Chloe Brown, Melinda Boone, Edmond Box, Juan Pablo. Vaccario, Paolo Serafini, Jashodeep Datta, Nipun Merchant
{"title":"Abstract A071: Immune biomarkers of response in a phase 1 trial of combined MEK/STAT3/PD-1 inhibition in metastatic pancreatic ductal adenocarcinoma (PDAC)","authors":"Peter Joel. Hosein, Sayan Chakraborty, Anna Bianchi, Yolanda Justal, Nkiruka Ezenwajiaku, Maria Vanessa. Yow, Chloe Brown, Melinda Boone, Edmond Box, Juan Pablo. Vaccario, Paolo Serafini, Jashodeep Datta, Nipun Merchant","doi":"10.1158/1538-7445.pancreatic25-a071","DOIUrl":null,"url":null,"abstract":"Background: PDAC remains resistant to immune checkpoint inhibition (ICI), in part due to a suppressive tumor microenvironment (TME) driven by oncogenic RAS and stromal interactions. Our preclinical data demonstrate that combined MEK and JAK/STAT3 inhibition reprograms the TME, reduces immunosuppressive myeloid cell populations, and restores effector T cell function, facilitating ICI sensitivity. Based on these findings, we conducted a phase 1 trial (NCT05440942) to evaluate the safety, preliminary efficacy, and immune biomarkers of trametinib (MEKi), ruxolitinib (JAK/STAT3i), and retifanlimab (anti–PD-1) (TRRf), in patients with metastatic PDAC as second or third-line therapy. Methods: The trial consisted of a dose escalation phase (Part 1, N=8) and an expansion phase (Part 2, N=20) at the target dose level (trametinib 2 mg PO daily, ruxolitinib 15 mg PO BID, retifanlimab 500 mg IV every 28 days). Peripheral blood and core biopsies were collected at baseline and after Cycle 1 of treatment cycle for biomarker analyses, including high-dimensional flow cytometry, cytokine profiling, and CODEX spatial immune profiling. Results: No dose-limiting toxicities were observed across all dose levels. The most common grade 3/4 adverse events (AE’s) were fatigue (32%), anemia (29%) and transaminase increase (18%). Among 20 patients in part 2, 2 achieved partial responses and 5 had stable disease for over 16 weeks (objective response rate 10%; disease control rate 35%). In-depth immune profiling of 5 responders (defined as having disease control) and 5 non-responders after Cycle 1 revealed that responders exhibited expansion of CD4+ early effector and memory T cells with increased levels of Granzyme B and CD107a, consistent with enhanced cytotoxic potential. Responders also demonstrated a reduction in monocytic myeloid-derived suppressor cells (M-MDSCs) and regulatory B cells, along with increases in mature NK cells. In contrast, non-responders showed expansion of total CD19+ B cells along with their CD20+ subset of memory B cells and downregulation of CD38 on CD4+ T cells, suggestive of impaired activation. Notably, plasma IL-12p70 and IFN-γ levels were significantly elevated after one treatment cycle in all six patients tested, regardless of response status, supporting TRRf-induced systemic Th1 immune polarization. Conclusions: TRRf therapy was well tolerated in patients with metastatic PDAC and induced a distinct immune reprogramming in responders characterized by activation of memory/effector CD4+ T cells, innate cytotoxicity via NK cell expansion, and systemic Th1 cytokine induction. These circulating immune phenotypes and cytokine profiles may serve as predictive biomarkers and provide mechanistic insight into response to MEK/STAT3-targeted immunotherapy in PDAC. Citation Format: Peter Joel. Hosein, Sayan Chakraborty, Anna Bianchi, Yolanda Justal, Nkiruka Ezenwajiaku, Maria Vanessa. Yow, Chloe Brown, Melinda Boone, Edmond Box, Juan Pablo. Vaccario, Paolo Serafini, Jashodeep Datta, Nipun Merchant. Immune biomarkers of response in a phase 1 trial of combined MEK/STAT3/PD-1 inhibition in metastatic pancreatic ductal adenocarcinoma (PDAC) [abstract]. In: Proceedings of the AACR Special Conference in Cancer Research: Advances in Pancreatic Cancer Research—Emerging Science Driving Transformative Solutions; Boston, MA; 2025 Sep 28-Oct 1; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2025;85(18_Suppl_3): nr A071.","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":"120 1","pages":""},"PeriodicalIF":16.6000,"publicationDate":"2025-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1158/1538-7445.pancreatic25-a071","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: PDAC remains resistant to immune checkpoint inhibition (ICI), in part due to a suppressive tumor microenvironment (TME) driven by oncogenic RAS and stromal interactions. Our preclinical data demonstrate that combined MEK and JAK/STAT3 inhibition reprograms the TME, reduces immunosuppressive myeloid cell populations, and restores effector T cell function, facilitating ICI sensitivity. Based on these findings, we conducted a phase 1 trial (NCT05440942) to evaluate the safety, preliminary efficacy, and immune biomarkers of trametinib (MEKi), ruxolitinib (JAK/STAT3i), and retifanlimab (anti–PD-1) (TRRf), in patients with metastatic PDAC as second or third-line therapy. Methods: The trial consisted of a dose escalation phase (Part 1, N=8) and an expansion phase (Part 2, N=20) at the target dose level (trametinib 2 mg PO daily, ruxolitinib 15 mg PO BID, retifanlimab 500 mg IV every 28 days). Peripheral blood and core biopsies were collected at baseline and after Cycle 1 of treatment cycle for biomarker analyses, including high-dimensional flow cytometry, cytokine profiling, and CODEX spatial immune profiling. Results: No dose-limiting toxicities were observed across all dose levels. The most common grade 3/4 adverse events (AE’s) were fatigue (32%), anemia (29%) and transaminase increase (18%). Among 20 patients in part 2, 2 achieved partial responses and 5 had stable disease for over 16 weeks (objective response rate 10%; disease control rate 35%). In-depth immune profiling of 5 responders (defined as having disease control) and 5 non-responders after Cycle 1 revealed that responders exhibited expansion of CD4+ early effector and memory T cells with increased levels of Granzyme B and CD107a, consistent with enhanced cytotoxic potential. Responders also demonstrated a reduction in monocytic myeloid-derived suppressor cells (M-MDSCs) and regulatory B cells, along with increases in mature NK cells. In contrast, non-responders showed expansion of total CD19+ B cells along with their CD20+ subset of memory B cells and downregulation of CD38 on CD4+ T cells, suggestive of impaired activation. Notably, plasma IL-12p70 and IFN-γ levels were significantly elevated after one treatment cycle in all six patients tested, regardless of response status, supporting TRRf-induced systemic Th1 immune polarization. Conclusions: TRRf therapy was well tolerated in patients with metastatic PDAC and induced a distinct immune reprogramming in responders characterized by activation of memory/effector CD4+ T cells, innate cytotoxicity via NK cell expansion, and systemic Th1 cytokine induction. These circulating immune phenotypes and cytokine profiles may serve as predictive biomarkers and provide mechanistic insight into response to MEK/STAT3-targeted immunotherapy in PDAC. Citation Format: Peter Joel. Hosein, Sayan Chakraborty, Anna Bianchi, Yolanda Justal, Nkiruka Ezenwajiaku, Maria Vanessa. Yow, Chloe Brown, Melinda Boone, Edmond Box, Juan Pablo. Vaccario, Paolo Serafini, Jashodeep Datta, Nipun Merchant. Immune biomarkers of response in a phase 1 trial of combined MEK/STAT3/PD-1 inhibition in metastatic pancreatic ductal adenocarcinoma (PDAC) [abstract]. In: Proceedings of the AACR Special Conference in Cancer Research: Advances in Pancreatic Cancer Research—Emerging Science Driving Transformative Solutions; Boston, MA; 2025 Sep 28-Oct 1; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2025;85(18_Suppl_3): nr A071.
摘要:MEK/STAT3/PD-1联合抑制转移性胰腺导管腺癌(PDAC)的1期试验免疫生物标志物反应
背景:PDAC仍然抵抗免疫检查点抑制(ICI),部分原因是由致癌RAS和基质相互作用驱动的抑制性肿瘤微环境(TME)。我们的临床前数据表明,MEK和JAK/STAT3联合抑制可重编程TME,减少免疫抑制的骨髓细胞群,恢复效应T细胞功能,促进ICI敏感性。基于这些发现,我们开展了一项i期试验(NCT05440942),以评估trametinib (MEKi)、ruxolitinib (JAK/STAT3i)和retifanlimab(抗pd -1) (TRRf)作为转移性PDAC患者二线或三线治疗的安全性、初步疗效和免疫生物标志物。方法:试验分为剂量递增期(第1部分,N=8)和扩展期(第2部分,N=20),目标剂量水平为(舒美替尼每日2mg PO,鲁索替尼每日15mg PO BID,瑞替利单抗500mg IV,每28天)。在基线和治疗周期第1周期后收集外周血和核心活检,用于生物标志物分析,包括高维流式细胞术、细胞因子谱和CODEX空间免疫谱。结果:所有剂量水平均未观察到剂量限制性毒性。最常见的3/4级不良事件(AE)是疲劳(32%)、贫血(29%)和转氨酶升高(18%)。第2部分20例患者中,2例部分缓解,5例病情稳定16周以上(客观缓解率10%,疾病控制率35%)。在第1周期后,对5名应答者(定义为疾病控制)和5名无应答者的深入免疫分析显示,应答者表现出CD4+早期效应细胞和记忆T细胞的扩张,颗粒酶B和CD107a水平升高,与增强的细胞毒性潜力一致。应答者还表现出单核髓源性抑制细胞(M-MDSCs)和调节性B细胞的减少,以及成熟NK细胞的增加。相比之下,无应答者表现出CD19+ B细胞总数和CD20+记忆B细胞亚群的增加,CD4+ T细胞上CD38的下调,提示活化受损。值得注意的是,在一个治疗周期后,所有6名患者的血浆IL-12p70和IFN-γ水平均显著升高,无论反应状态如何,这支持了trrf诱导的全身Th1免疫极化。结论:TRRf治疗在转移性PDAC患者中耐受性良好,并在应答者中诱导了明显的免疫重编程,其特征是激活记忆/效应CD4+ T细胞,通过NK细胞扩增产生先天细胞毒性,以及系统性Th1细胞因子诱导。这些循环免疫表型和细胞因子谱可以作为预测性生物标志物,并为PDAC患者对MEK/ stat3靶向免疫治疗的反应提供机制见解。引文格式:Peter Joel。Hosein, Sayan Chakraborty, Anna Bianchi, Yolanda Justal, Nkiruka Ezenwajiaku, Maria Vanessa。你,克洛伊·布朗,梅林达·布恩,埃德蒙·博克斯,胡安·巴勃罗。Vaccario, Paolo Serafini, Jashodeep Datta, Nipun Merchant。MEK/STAT3/PD-1联合抑制转移性胰腺导管腺癌(PDAC)的1期试验中的免疫生物标志物反应[摘要]。摘自:AACR癌症研究特别会议论文集:胰腺癌研究进展-新兴科学驱动变革解决方案;波士顿;2025年9月28日至10月1日;波士顿,MA。费城(PA): AACR;癌症研究2025;85(18_Suppl_3): nr A071。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Cancer research
Cancer research 医学-肿瘤学
CiteScore
16.10
自引率
0.90%
发文量
7677
审稿时长
2.5 months
期刊介绍: Cancer Research, published by the American Association for Cancer Research (AACR), is a journal that focuses on impactful original studies, reviews, and opinion pieces relevant to the broad cancer research community. Manuscripts that present conceptual or technological advances leading to insights into cancer biology are particularly sought after. The journal also places emphasis on convergence science, which involves bridging multiple distinct areas of cancer research. With primary subsections including Cancer Biology, Cancer Immunology, Cancer Metabolism and Molecular Mechanisms, Translational Cancer Biology, Cancer Landscapes, and Convergence Science, Cancer Research has a comprehensive scope. It is published twice a month and has one volume per year, with a print ISSN of 0008-5472 and an online ISSN of 1538-7445. Cancer Research is abstracted and/or indexed in various databases and platforms, including BIOSIS Previews (R) Database, MEDLINE, Current Contents/Life Sciences, Current Contents/Clinical Medicine, Science Citation Index, Scopus, and Web of Science.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信