4.8 Article

Combined MEK and STAT3 Inhibition Uncovers Stromal Plasticity by Enriching for Cancer-Associated Fibroblasts With Mesenchymal Stem Cell-Like Features to Overcome Immunotherapy Resistance in Pancreatic Cancer

期刊

GASTROENTEROLOGY
卷 163, 期 6, 页码 1593-1612

出版社

W B SAUNDERS CO-ELSEVIER INC
DOI: 10.1053/j.gastro.2022.07.076

关键词

Pancreatic Ductal Adenocarcinoma; MEK; STAT3; Cancer-Associated Fibroblast; Immunotherapy; Immune Checkpoint Inhibition; Stromal Plasticity; Heterogeneity; Mesenchymal Stem Cell

资金

  1. National Institutes of Health, National Cancer Institute [R21CA209536, 2R01CA161976, T32CA211034]
  2. American Association for Cancer Research Translational Research [15-65-25-MER]
  3. University of Miami Sylvester Comprehensive Cancer Center P30 CCSG under National Institutes of Health National Cancer Institute Award [P30CA240139]
  4. Miami Clinical and Translational Science Institute under National Institutes of Health National Center for Advancing Translational Sciences Award [UL1TR002736]
  5. University of Miami Stanley Glaser Foundation
  6. American College of Surgeons Franklin Martin Career Development Award
  7. Association for Academic Surgery Joel J. Roslyn Faculty Award
  8. American Cancer Society grant [IRG 98-277-13]

向作者/读者索取更多资源

Combined MEK and STAT3 inhibition can reprogram cancer-associated fibroblasts (CAF) and the immune microenvironment to overcome resistance to immune checkpoint inhibition in pancreatic ductal adenocarcinoma (PDAC).
BACKGROUND & AIMS: We have shown that reciprocally activated rat sarcoma (RAS)/mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) and Janus kinase/signal transducer and activator of transcription 3 (STAT3) pathways mediate therapeutic resistance in pancreatic ductal adenocarcinoma (PDAC), while combined MEK and STAT3 inhibition (MEKithornSTAT3i) overcomes such resistance and alters stromal architecture. We now determine whether MEKithornSTAT3i reprograms the cancer-associated fibroblast (CAF) and immune microenvironment to overcome resistance to immune checkpoint inhibition in PDAC. METHODS: CAF and immune cell transcriptomes in MEKi (trametinib)thornSTAT3i (ruxolitinib)-treated vs vehicle-treated Ptf1a(Cre/+);LSL-Kras(G12D/+); Tgfbr2(flox/flox) (PKT) tumors were examined via single-cell RNA sequencing (scRNAseq). Clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeats associated protein 9 silencing of CAF-restricted Map2k1/Mek1 or Stat3, or both, enabled interrogation of CAF-dependent effects on immunologic remodeling in orthotopic models. Tumor growth, survival, and immune profiling via mass cytometry by time-of-flight were examined in PKT mice treated with vehicle, anti-programmed cell death protein 1 (PD-1) monotherapy, and MEKithornSTAT3i combined with anti-PD1. RESULTS: MEKithornSTAT3i attenuates Il6/Cxcl1-expressing proinflammatory and Lrrc15-expressing myofibroblastic CAF phenotypes while enriching for Ly6a/Cd34-expressing CAFs exhibiting mesenchymal stem cell-like features via scRNAseq in PKT mice. This CAF plasticity is associated with M2to-M1 reprogramming of tumor-associated macrophages, and enhanced trafficking of cluster of differentiation 8(+) T cells, which exhibit distinct effector transcriptional programs. These MEKi(+) STAT3i-induced effects appear CAF-dependent, because CAF-restricted Mek1/Stat3 silencing mitigates inflammatory-CAF polarization and myeloid infiltration in vivo. Addition of MEKithornSTAT3i to PD-1 blockade not only dramatically improves antitumor responses and survival in PKT mice but also augments recruitment of activated/memory T cells while improving their degranulating and cytotoxic capacity compared with anti-PD-1 monotherapy. Importantly, treatment of a patient who has chemotherapy-refractory metastatic PDAC with MEKi (trametinib), STAT3i (ruxolitinib), and PD-1 inhibitor (nivolumab) yielded clinical benefit. CONCLUSIONS: Combined MEKithornSTAT3i mitigates stromal inflammation and enriches for CAF phenotypes with mesenchymal stem cell-like properties to overcome immunotherapy resistance in PDAC.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.8
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据