scholarly journals STING contributes to anti-glioma immunity via triggering type-I IFN signals in the tumor microenvironment

2014 ◽  
Vol 2 (Suppl 3) ◽  
pp. P228 ◽  
Author(s):  
Takayuki Ohkuri ◽  
Arundhati Ghosh ◽  
Akemi Kosaka ◽  
Jianzhong Zhu ◽  
Maki Ikeura ◽  
...  
2017 ◽  
Vol 114 (51) ◽  
pp. E10981-E10990 ◽  
Author(s):  
Meredith L. Stone ◽  
Katherine B. Chiappinelli ◽  
Huili Li ◽  
Lauren M. Murphy ◽  
Meghan E. Travers ◽  
...  

Ovarian cancer is the most lethal of all gynecological cancers, and there is an urgent unmet need to develop new therapies. Epithelial ovarian cancer (EOC) is characterized by an immune suppressive microenvironment, and response of ovarian cancers to immune therapies has thus far been disappointing. We now find, in a mouse model of EOC, that clinically relevant doses of DNA methyltransferase and histone deacetylase inhibitors (DNMTi and HDACi, respectively) reduce the immune suppressive microenvironment through type I IFN signaling and improve response to immune checkpoint therapy. These data indicate that the type I IFN response is required for effective in vivo antitumorigenic actions of the DNMTi 5-azacytidine (AZA). Through type I IFN signaling, AZA increases the numbers of CD45+ immune cells and the percentage of active CD8+ T and natural killer (NK) cells in the tumor microenvironment, while reducing tumor burden and extending survival. AZA also increases viral defense gene expression in both tumor and immune cells, and reduces the percentage of macrophages and myeloid-derived suppressor cells in the tumor microenvironment. The addition of an HDACi to AZA enhances the modulation of the immune microenvironment, specifically increasing T and NK cell activation and reducing macrophages over AZA treatment alone, while further increasing the survival of the mice. Finally, a triple combination of DNMTi/HDACi plus the immune checkpoint inhibitor α-PD-1 provides the best antitumor effect and longest overall survival, and may be an attractive candidate for future clinical trials in ovarian cancer.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Xuezhi Cao ◽  
Yong Liang ◽  
Zhenxiang Hu ◽  
Huiyu Li ◽  
Jiaming Yang ◽  
...  

AbstractType I interferon is promising in treating different kinds of tumors, but has been limited by its toxicity, lack of tumor targeting, and very short half-life. To target tumors, reduce systemic toxicity, and increase half-life, here we engineer a masked type I IFN-Fc (ProIFN) with its natural receptor connected by a cleavable linker that can be targeted by tumor-associated proteases. ProIFN has a prolonged serum half-life and shows an improved tumor-targeting effect. Interestingly, ProIFN-treated mice show enhanced DC cross-priming and significant increased CD8+ infiltration and effector function in the tumor microenvironment. ProIFN is able to improve checkpoint blockade efficacy in established tumors, as well as radiation efficacy for both primary and metastatic tumors. ProIFN exhibits superior long-term pharmacokinetics with minimal toxicity in monkeys. Therefore, this study demonstrates an effective tumor-activating IFN that can increase targeted immunity against primary tumor or metastasis and reduce periphery toxicity to the host.


2020 ◽  
Vol 3 ◽  
Author(s):  
Abigail Chmiel ◽  
Steven Rhodes ◽  
Steve Angus ◽  
Yongzheng He ◽  
Quingbo Lu ◽  
...  

Background/Objective:  Neurofibromatosis type 1 (NF1) is a cancer predisposition syndrome caused by mutations in the NF1 tumor suppressor gene. Patients with NF1 develop tumors of the peripheral nervous system called plexiform neurofibromas (PNs). These histopathologically complex tumors are composed of various immune and inflammatory cells. Mast cells have previously been identified as one key immune cell lineage underpinning PN initiation and progression, however new technologies leveraging RNA-sequencing (RNAseq) allow for the broad and systematic characterization of the PN tumor microenvironment. Here we utilized these tools to delineate PN cellular composition.  Methods:  RNA seq was performed on murine wild type (n=6) and PN (n=6) tissues. We utilized CIBERSORT to profile the cellular constituents of the PN microenvironment. CIBERSORT is a deconvolution method that uses a reference matrix to estimate the relative proportions of various cell types. Statistical analyses were performed on cell lineage subtypes delineated by CIBERSORT. We further performed a Gene Set Enrichment Analysis (GSEA) to identify which pathways and cytokines might be upregulated in PNs.   Results:  Using a murine reference matrix, the macrophage lineage, M0 (p = 0.072), M1 (p = 0.1), were upregulated in PNs (n=6) compared to WT (n=6). A human reference matrix showed M2 (p=0.025) to be upregulated in PNs. GSEA showed IL-1, IL-6, IL-8, TNF and Type I IFN and cytokine secretion to be upregulated in PNs compared to WT.   Conclusion:  Macrophages were among the most upregulated components of the NF1 tumor microenvironment and upregulation of IL-1, IL-6, IL-8, TNF and Type I IFN production may be contributing to inflammation that is critical in the initiation and progression of PNs.    Scientific/Clinical/Policy Impact and Implications:  Pharmacotherapies that can target the macrophage lineage and/or aforementioned cytokines may have utility in the treatment of PNs. Further studies are necessary to evaluate this hypothesis. 


Cytokine ◽  
2011 ◽  
Vol 56 (1) ◽  
pp. 87
Author(s):  
Marcel Renn ◽  
Tobias Bald ◽  
Alexander Sporleder ◽  
Sandra Mikus ◽  
Jennifer Landsberg ◽  
...  

2014 ◽  
Vol 16 (suppl 5) ◽  
pp. v108-v108
Author(s):  
T. Ohkuri ◽  
A. Ghosh ◽  
A. Kosaka ◽  
J. Zhu ◽  
M. Ikeura ◽  
...  

2014 ◽  
Vol 2 (12) ◽  
pp. 1199-1208 ◽  
Author(s):  
Takayuki Ohkuri ◽  
Arundhati Ghosh ◽  
Akemi Kosaka ◽  
Jianzhong Zhu ◽  
Maki Ikeura ◽  
...  

Cell ◽  
2021 ◽  
Author(s):  
Khiem C. Lam ◽  
Romina E. Araya ◽  
April Huang ◽  
Quanyi Chen ◽  
Martina Di Modica ◽  
...  

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A907-A907
Author(s):  
Kristy Kuplast-Barr ◽  
Kristy Kuplast-Barr ◽  
Melissa Johnson ◽  
Manish Patel ◽  
Timothy Yap ◽  
...  

BackgroundPARP7 is a mono-ART that is upregulated in response to cellular stress (e.g., viral infection, cigarette smoke), and suppresses the Type I interferon (IFN) response following cytosolic nucleic acid sensing. RBN-2397 is a first-in-class PARP7 inhibitor, inducing cancer cell autonomous and immune stimulatory effects in preclinical models through enhanced Type I IFN signaling in cancer cells. Moreover, RBN-2397 induces CD8 T cell-dependent tumor-specific immune memory in an immunocompetent mouse cancer model.1 RBN-2397 is currently being tested in an ongoing Phase I clinical study (NCT04053673).2 Here we aimed to compare biomarker results from preclinical models and patient samples.MethodsIn preclinical models, interferon-stimulated gene (ISG) expression was assessed by qPCR, NanoString, or ELISA. Plasma CXCL10 from patients was measured by MSD while ISG expression in PBMCs was measured by NanoString. Baseline and on-treatment patient tumor biopsies were analyzed by NanoString, CD8/GZMB IHC, and MIBI-TOF to characterize immune changes in the tumor microenvironment.ResultsRBN-2397 potently restored tumoral Type I IFN signaling in preclinical models as demonstrated by increases in ISGs, namely CXCL10, which were not observed in non-tumor tissue (e.g. spleen, PBMCs). In peripheral blood from patients treated with RBN-2397, neither plasma nor PBMC CXCL10 increased more than 2-fold over baseline. Expression of 42 ISGs was not consistently induced in a dose-dependent manner in PBMCs. However, in tumor types of interest (e.g. cancers of the upper aerodigestive tract), CXCL10 expression increased 1.5 to 8-fold, with similar effects observed for a subset of ISGs in 5 evaluable paired biopsy samples.Confirming preclinical studies [1], up to 8-fold increases in CD8 T cell infiltration along with induction of granzyme B expression were observed in 4 of 5 paired patient tumor biopsies by immunohistochemistry. Using the MIBI-TOF technology, we observed up to 50-fold increases in intratumoral activated T cells as well as monocytes and M1 macrophages, most strikingly in two NSCLC patients.ConclusionsInhibition of PARP7 with RBN-2397 restores tumor-intrinsic Type I IFN signaling in preclinical models leading to enhanced adaptive immunity, resulting in CD8 T cell-dependent durable tumor regressions. These observations are mirrored in samples from patients treated with RBN-2397 in that pharmacodynamic effects of RBN-2397 were preferentially observed in tumor tissue relative to the periphery, including an increase in immune infiltration into the tumor microenvironment. These data provide evidence for induction of an adaptive immune response and confirm the tumor-intrinsic, immunomodulatory mechanism of action of RBN-2397 in patients.ReferencesGozgit, et al. PARP7 negatively regulates the type I interferon response in cancer cells and its inhibition triggers antitumor immunity. Cancer Cell 2021; In press.Falchook, et al. A first-in-human phase 1 study of a novel PARP7 inhibitor RBN-2397 in patients with advanced solid tumors. ASCO 2021; oral presentation.


Sign in / Sign up

Export Citation Format

Share Document