2018
DOI: 10.1158/1078-0432.ccr-17-3573
|View full text |Cite
|
Sign up to set email alerts
|

IDO1 Inhibition Synergizes with Radiation and PD-1 Blockade to Durably Increase Survival Against Advanced Glioblastoma

Abstract: Glioblastoma is the most aggressive primary brain tumor in adults with a median survival of 15-20 months. Numerous approaches and novel therapeutics for treating glioblastoma have been investigated in the setting of phase III clinical trials, including a recent analysis of the immune checkpoint inhibitor, nivolumab (anti-PD-1), which failed to improve recurrent glioblastoma patient survival. However, rather than abandoning immune checkpoint inhibitor treatment for glioblastoma, which has shown promise in other… Show more

Help me understand this report
View preprint versions

Search citation statements

Order By: Relevance

Paper Sections

Select...
3
1
1

Citation Types

4
134
1

Year Published

2018
2018
2024
2024

Publication Types

Select...
7
1

Relationship

1
7

Authors

Journals

citations
Cited by 158 publications
(139 citation statements)
references
References 44 publications
4
134
1
Order By: Relevance
“…High levels of IDO expression in the TME is correlated with poor prognosis and promotes resistance to CTLA-4 blockade [47] and CD19-CAR-T cells [48]. A recent study evidenced the beneficial role of IDO1 inhibitors in long-term survival in combination with radiotherapy and PD-1 blockade in a mouse model of glioblastoma [49]. Similarly, studies performed in the murine B16.SIY melanoma model showed improved tumor control when combining anti-PD-L1 or anti-CTLA-4 with the IDO inhibitor INCB23843 [50].…”
Section: Targeting the Tumor Microenvironment To Improve Immunotherapiesmentioning
confidence: 99%
“…High levels of IDO expression in the TME is correlated with poor prognosis and promotes resistance to CTLA-4 blockade [47] and CD19-CAR-T cells [48]. A recent study evidenced the beneficial role of IDO1 inhibitors in long-term survival in combination with radiotherapy and PD-1 blockade in a mouse model of glioblastoma [49]. Similarly, studies performed in the murine B16.SIY melanoma model showed improved tumor control when combining anti-PD-L1 or anti-CTLA-4 with the IDO inhibitor INCB23843 [50].…”
Section: Targeting the Tumor Microenvironment To Improve Immunotherapiesmentioning
confidence: 99%
“…Immunologically, GBM is characterized by a highly suppressive tumor microenvironment [12], and for most patients, there is scant intratumoral infiltration of effector T cells [27,28]. Well-described barriers to infiltration and/or activation of effector T cells in the GBM tumor microenvironment include immunosuppressive microglial cells and tumor-associated (M2) macrophages [29,30], upregulation of immune checkpoints on functionally exhausted cytotoxic T lymphocytes and other immune cells [31], the presence of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) [32,33], and increased expression and secretion of immune inhibitory molecules by tumor and other cells [2,14]. With each of these immunosuppressive mechanisms involving modulation of different signaling pathways and gene expression levels, as well as the unprecedented number of targeted and immunomodulatory therapies in development or already approved across oncology [34], prioritization of individual targets and combinations of targets for preclinical and clinical evaluation in GBM is a significant challenge.…”
Section: Discussionmentioning
confidence: 99%
“…However, despite promise in preclinical and early phase studies [2,3], single agent programmed death 1 (PD-1) inhibition is ineffective for recurrent GBM in the absence of a rare, markedly hypermutated tumor [46]. This is due, in part, to the relative paucity of intratumoral T cells in GBM compared to cancers with carcinogen-induced mutational signatures and high tumor mutational burden (TMB) [711].…”
Section: Introductionmentioning
confidence: 99%
“…The majority of endogenous AHR ligands are tryptophan metabolites, and the most abundant tryptophan metabolite in glioma is kynurenine, which is produced by enzymes 2,3-dioxygenase (IDO/TDO), binds to AHR and inhibits cytotoxic immune cells activity against glioma cells [6], forming a pathway of Tryptophan-Kynurenine-AHR-Impairment of Immunity. To block this pathway, inhibitors for the IDO/TDO were developed and mixed clinical trial results have been posted [7,8], not meeting expectations. An alternative route is to directly target AHR using small-molecule ligands.…”
Section: Introductionmentioning
confidence: 99%