STING pathway and modulation for cancer immunotherapy

2022 ◽  
pp. 353-373
Author(s):  
Ting Su ◽  
Nadia Tasnim Ahmed ◽  
Shurong Zhou ◽  
Xiang Liu ◽  
Guizhi Zhu
Biomedicines ◽  
2021 ◽  
Vol 10 (1) ◽  
pp. 33
Author(s):  
Hee Ra Jung ◽  
Seongman Jo ◽  
Min Jae Jeon ◽  
Hyelim Lee ◽  
Yeonjeong Chu ◽  
...  

In cancer immunotherapy, the cyclic GMP–AMP synthase–stimulator of interferon genes (STING) pathway is an attractive target for switching the tumor immunophenotype from ‘cold’ to ‘hot’ through the activation of the type I interferon response. To develop a new chemical entity for STING activator to improve cyclic GMP-AMP (cGAMP)-induced innate immune response, we identified KAS-08 via the structural modification of DW2282, which was previously reported as an anti-cancer agent with an unknown mechanism. Further investigation revealed that direct STING binding or the enhanced phosphorylation of STING and downstream effectors were responsible for DW2282-or KAS-08-mediated STING activity. Furthermore, KAS-08 was validated as an effective STING pathway activator in vitro and in vivo. The synergistic effect of cGAMP-mediated immunity and efficient anti-cancer effects successfully demonstrated the therapeutic potential of KAS-08 for combination therapy in cancer treatment.


2020 ◽  
Vol 9 (10) ◽  
pp. 3323
Author(s):  
Leila Motedayen Aval ◽  
James E. Pease ◽  
Rohini Sharma ◽  
David J. Pinato

Immune checkpoint inhibitors (ICI) have revolutionised cancer therapy. However, they have been effective in only a small subset of patients and a principal mechanism underlying immune-refractoriness is a ‘cold’ tumour microenvironment, that is, lack of a T-cell-rich, spontaneously inflamed phenotype. As such, there is a demand to develop strategies to transform the tumour milieu of non-responsive patients to one supporting T-cell-based inflammation. The cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) pathway is a fundamental regulator of innate immune sensing of cancer, with potential to enhance tumour rejection through the induction of a pro-inflammatory response dominated by Type I interferons. Recognition of these positive immune-modulatory properties has rapidly elevated the STING pathway as a putative target for immunotherapy, leading to a myriad of preclinical and clinical studies assessing natural and synthetic cyclic dinucleotides and non-nucleotidyl STING agonists. Despite pre-clinical evidence of efficacy, clinical translation has resulted into disappointingly modest efficacy. Poor pharmacokinetic and physiochemical properties of cyclic dinucleotides are key barriers to the development of STING agonists, most of which require intra-tumoral dosing. Development of systemically administered non-nucleotidyl STING agonists, or conjugation with liposomes, polymers and hydrogels may overcome pharmacokinetic limitations and improve drug delivery. In this review, we summarise the body of evidence supporting a synergistic role of STING agonists with currently approved ICI therapies and discuss whether, despite the numerous obstacles encountered to date, the clinical development of STING agonist as novel anti-cancer therapeutics may still hold the promise of broadening the reach of cancer immunotherapy.


2020 ◽  
Vol 12 (556) ◽  
pp. eaaz6606 ◽  
Author(s):  
Xueguang Lu ◽  
Lei Miao ◽  
Wenting Gao ◽  
Ziqi Chen ◽  
Kevin J. McHugh ◽  
...  

Activation of the stimulator of interferon gene (STING) pathway within the tumor microenvironment has been shown to generate a strong antitumor response. Although local administration of STING agonists has promise for cancer immunotherapy, the dosing regimen needed to achieve efficacy requires frequent intratumoral injections over months. Frequent dosing for cancer treatment is associated with poor patient adherence, with as high as 48% of patients failing to comply. Multiple intratumoral injections also disrupt the tumor microenvironment and vascular networks and therefore increase the risk of metastasis. Here, we developed microfabricated polylactic-co-glycolic acid (PLGA) particles that remain at the site of injection and release encapsulated STING agonist as a programmable sequence of pulses at predetermined time points that mimic multiple injections over days to weeks. A single intratumoral injection of STING agonist–loaded microparticles triggered potent local and systemic antitumor immune responses, inhibited tumor growth, and prolonged survival as effectively as multiple soluble doses, but with reduced metastasis in several mouse tumor models. STING agonist–loaded microparticles improved the response to immune checkpoint blockade therapy and substantially decreased the tumor recurrence rate from 100 to 25% in mouse models of melanoma when administered during surgical resection. In addition, we demonstrated the therapeutic efficacy of STING microparticles on an orthotopic pancreatic cancer model in mice that does not allow multiple intratumoral injections. These findings could directly benefit current STING agonist therapy by decreasing the number of injections, reducing risk of metastasis, and expanding its applicability to hard-to-reach cancers.


2015 ◽  
Vol 216 ◽  
pp. 149-157 ◽  
Author(s):  
Takashi Nakamura ◽  
Hiroko Miyabe ◽  
Mamoru Hyodo ◽  
Yusuke Sato ◽  
Yoshihiro Hayakawa ◽  
...  

2019 ◽  
Vol 12 (1) ◽  
Author(s):  
Anping Li ◽  
Ming Yi ◽  
Shuang Qin ◽  
Yongping Song ◽  
Qian Chu ◽  
...  

2021 ◽  
pp. 2100065
Author(s):  
Min Gao ◽  
Yu‐Qing Xie ◽  
Kewen Lei ◽  
Yu Zhao ◽  
Armand Kurum ◽  
...  

2019 ◽  
Vol 25 (5) ◽  
pp. 412-427 ◽  
Author(s):  
Gilles Berger ◽  
Mickaël Marloye ◽  
Sean E. Lawler

Cytokine ◽  
2016 ◽  
Vol 77 ◽  
pp. 245-247 ◽  
Author(s):  
Leticia Corrales ◽  
Thomas F. Gajewski

Sign in / Sign up

Export Citation Format

Share Document