Next Article in Journal
mRNA Vaccines Enhance Neutralizing Immunity against SARS-CoV-2 Variants in Convalescent and ChAdOx1-Primed Subjects
Next Article in Special Issue
An Overview of Influenza Viruses and Vaccines
Previous Article in Journal
Changing Attitudes toward the COVID-19 Vaccine among North Carolina Participants in the COVID-19 Community Research Partnership
Previous Article in Special Issue
Novel Surrogate Neutralizing Assay Supports Parvovirus B19 Vaccine Development for Children with Sickle Cell Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Promise and Challenges of Cyclic Dinucleotides as Molecular Adjuvants for Vaccine Development

1
Department of Chemistry, Brock University, St. Catharines, ON L2S 3A1, Canada
2
Human Health and Therapeutics Research Centre, National Research Council Canada, Ottawa, ON K1A 0R6, Canada
3
Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
*
Authors to whom correspondence should be addressed.
Vaccines 2021, 9(8), 917; https://doi.org/10.3390/vaccines9080917
Submission received: 28 June 2021 / Revised: 5 August 2021 / Accepted: 10 August 2021 / Published: 17 August 2021
(This article belongs to the Special Issue Advances in Vaccine Development)

Abstract

:
Cyclic dinucleotides (CDNs), originally discovered as bacterial second messengers, play critical roles in bacterial signal transduction, cellular processes, biofilm formation, and virulence. The finding that CDNs can trigger the innate immune response in eukaryotic cells through the stimulator of interferon genes (STING) signalling pathway has prompted the extensive research and development of CDNs as potential immunostimulators and novel molecular adjuvants for induction of systemic and mucosal innate and adaptive immune responses. In this review, we summarize the chemical structure, biosynthesis regulation, and the role of CDNs in enhancing the crosstalk between host innate and adaptive immune responses. We also discuss the strategies to improve the efficient delivery of CDNs and the recent advance and future challenges in the development of CDNs as potential adjuvants in prophylactic vaccines against infectious diseases and in therapeutic vaccines against cancers.

1. Introduction

Host innate immunity is the first line of defence against invading microbial pathogens and cancers and also plays a critical role in the development of autoimmune diseases. Innate immune cells, such as macrophages, granulocytes, dendritic cells (DCs), natural killer (NK) cells, and mast cells, contain specific pathogen-recognition receptors (PRRs), which sense pathogen-associated molecular patterns (PAMPs) and induce the production of proinflammatory cytokines/chemokines and subsequently activate the adaptive immune response. A variety of PRRs has now been identified and characterized, including the Toll-like receptors (TLRs), nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), and Retinoic acid-inducible gene (RIG)-I-like receptors (RLRs). In addition, cyclic dinucleotides (CDNs), including bacterial second messengers bis-(3′,5′)-cyclic diguanosine monophosphate (c-di-GMP) and bis-(3′,5′)-cyclic diadenosine monophosphate (c-di-AMP), 3′5′-3′5′ cyclic GMP-AMP (3′3′-cGAMP) produced by Vibrio cholerae, and metazoan second messenger 2′5′-3′5′ cyclic GMP-AMP (2′3′-cGAMP, hereafter cGAMP), bind to an endoplasmic reticulum (ER)-associated sensor termed as a stimulator of interferon genes (STING) [1,2,3,4]. The finding that CDNs can trigger the innate immune response in eukaryotic cells through the STING signalling pathway, leading to the type I interferons (IFN) production, has prompted the extensive research and development of CDNs as potential vaccine adjuvants and agents for cancer immunotherapy [5].
In this review, we summarize the structure and chemical synthesis, the biological characteristics, the current status of CDN vaccine adjuvant research, including their superior adjuvant activities, in vivo mode of action, and application in the development of infectious disease and cancer vaccines. We also examine the strategies to improve the efficient delivery of CDNs and future challenges in advancing the lead CDN adjuvant candidate into clinical trials and ultimately in human clinical use.

2. Discovery and Chemical Structure of CDNs

In their investigation of the regulation of cellulose synthesis in Acetobacter xylinum (now classified as Gluconacetobacter xylinus), Benziman and co-workers discovered a cyclic oligonucleotide that appeared to be an activator of cellulose synthase [6,7]. This activator was subsequently identified in the same laboratory as bis-(3′→5′)-cyclic diguanylic acid (also known in the literature as 3′,5′-cyclic diguanylic acid, c-di-GMP, or c-diGMP, Figure 1), with the mass spectrometric and NMR data in agreement with samples generated by enzymatic synthesis from guanosine triphosphate (GTP) catalyzed by diguanylate cyclase [8]. The crystal structure of chemically synthesized c-di-GMP was subsequently resolved by Rich and co-workers [9].
The adenylyl analog of c-di-GMP, c-di-AMP, together with the diadenylate cyclase activity of the nucleotide-binding domains of the DNA-integrity scanning protein A (DisA), were discovered in 2008 [10]. The DisA protein was shown to control a sporulation checkpoint in Bacillus subtilis in response to DNA double-strand breaks. Since this discovery, the roles of c-di-AMP in bacterial physiology were rapidly expanded [11,12].
A guanosine/adenosine hybrid CDN, 3′3′-cGAMP (3′5′-3′5′ cyclic GMP-AMP or c(GpAp)), was also found in a V. cholerae strain expressing dinucleotide cyclase DncV [13], which is involved in intestinal colonization and chemotaxis of V. cholerae.
In two back-to-back publications in Science in 2013, Chen and co-workers reported the roles of cyclic GMP-AMP synthase in the activation of the type I interferon pathway [14], and the identification of 2′2′-cyclic GMP-AMP (2′2′-cGAMP) [15] as the endogenous ligand for cyclic GMP-AMP synthase. Very shortly after the publication of these original results, it was determined that the endogenous ligand is, in fact, 2′3′-cyclic GMP-AMP (2′3′-cGAMP) [16].
In the ensuing few years since its discovery, c-di-GMP attracted very little attention until around 2004/2005 when its roles as a bacterial second messenger were recognized, and the field quickly expanded with the potential of this cyclic dinucleotide and its analogs as immunostimulatory agents and vaccine adjuvants. The rapidly growing interest in cyclic dinucleotides is clearly reflected by the expansion of the number of published records in the last two decades (Figure 2).

3. c-di-GMP as a Universal Bacterial Second Messenger

Intracellular levels of c-di-GMP are regulated through the actions of diguanylate cyclases (DGC) and phosphodiesterases (PDE), usually as a response to an environmental stimulus, i.e., first messenger. In signalling cascades involving c-di-GMP, binding of this second messenger with effector molecules, which are usually proteins and, in some cases, RNA such as riboswitches, allows for the regulation of the processes mediated by these effector molecules. Most DGCs contain a conserved GGDEF domain, although DGC cyclase enzymes with GGEEF, AGDEF, and GGDEM domains have also been identified [17]. PDEs, on the other hand, typically contain EAL (or less frequently HY-GYP) domains. Interestingly, numerous other proteins containing these domains are enzymatically inactive.
In the last 15 years or so, significant progress has been made toward the identification of effector molecules that are key to the biological processes or phenotypes regulated by c-di-GMP. The first such effector protein identified was the cellulose synthase from G. xylinus (formerly A. xylinum) [8,18], where the cellulose synthesis is activated by c-di-GMP. Almost 20 years after the discovery of the regulation of cellulose synthase activity via the allosteric binding with c-di-GMP, the PilZ domain was identified to be responsible for the binding to c-di-GMP [19]. Indeed, PilZ was subsequently found to be a common c-di-GMP binding domain among bacterial species, regulating bacterial motility [20,21,22,23] and virulence [24]. Other proteins such as the PelD protein from Pseudomonas aeruginosa [25], LapD protein from P. fluorescens [26], the transcription factor VpsT from V. cholera, the transcription factor FleQ (flagellar transcriptional regulator) from P. aeruginosa [27], and the Bcam1349 protein from Burkholderia cenocepacia [28] have also been identified as effector proteins in c-di-GMP-mediated processes. Furthermore, riboswitches [29,30] and self-splicing ribozyme [31] have been shown to be another class of effector molecules. Located in the 5′-untranslated upstream of the open reading frames of DGC and PDE proteins, binding of these riboswitches to c-di-GMP can serve as a regulator for the expression of DGC and PDE proteins.
Taken together, c-di-GMP mediated signalling networks have been shown to affect the regulation of a wide range of biological processes and phenotypes in bacteria (Figure 3), enabled by the binding of this second messenger to an effector molecule.

4. c-di-GMP and Analogs Generated to Date

While c-di-GMP can be isolated from bacterial cultures [32], this approach is very limited in the amount of accessible material. Furthermore, the generation of modified c-di-GMP via this approach is usually unfeasible. With this in mind, a number of methods have been described toward the synthesis of c-di-GMP and analogs, allowing for access to a relatively large amount of material for structural, bacterial, and immunological applications.
The vast majority of CDN analogs reported to date are based on phosphate or modified phosphate backbone in nature. To date, CDNs with all possible combinations of canonical nucleobases have been synthesized [33]. Figure S1 summarizes the c-di-GMP analogs reported to date that feature modifications in nucleobases, various sugar residues (e.g., ribose, deoxyribose, and 2′-modified ribose), phosphate, and phosphorothioate. A selection of CDNs containing both 2′-5′ and 3′-5′ internucleoside phosphate/phosphorothioate linkages (Figure S2) have also been made available through chemical synthesis. Among the chemistry reported for the synthesis of c-di-GMP analogs, some features are of important consideration: versatility of the chemistry, ease to scale up the process, and to purify the final product. It is worth noting that c-di-GMP [34,35], c-di-araGMP and c-di-dGMP [36], and other analogs [37], 3′3′-cGAMP and 2′3′-cGAMP [38,39] and their analogs [39,40] have also been generated enzymatically.
Furthermore, CDN analogs where the phosphate backbone is replaced with bis-carbamate, amide, urea, thiourea, carbodiimide, and triazolyl have also been synthesized (Figure S3) to explore their applications in bacterial physiology. In addition, conjugates of c-di-GMP with other molecular entities (Figure S4) have also been synthesized to study biological processes involving this bacterial second messenger. These conjugates are particularly useful to visualize c-di-GMP when a fluorophore is covalently attached or to pull-down molecules that bind to c-di-GMP when biotin is conjugated to c-di-GMP.

5. Immunostimulatory Function of CDNs in Host Innate Immune Responses

It is now well established that CDNs activate innate immunity by directly binding to STING to induce the production of type I IFN and inflammatory cytokines via TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3) pathway (Figure 4) [41,42]. However, alternative pathways also exist. McFarland et al. have identified the oxidoreductase RECON as a cytosolic sensor for bacterial CDNs in modulating the inflammatory gene activation via its antagonistic effects on STING and nuclear factor kappa B (NF-κB) [43]. In addition, it was reported that c-di-GMP could induce antigen-specific antibody and balanced T helper type 1 (Th1)/Th2/Th17 cell responses through a novel IFN-I-independent, STING-NF-κB-tumour necrosis factor (TNF)-α pathway, which is distinct from STING-mediated DNA adjuvant activity that requires IFN-I, but not TNF-α, production [44]. The critical role of TNF-α signalling in this pathway was confirmed in studies using TNFR1(−/−) mice. Moreover, Chang et al. have recently suggested A (2B) adenosine receptors in the intestinal epithelium as a potential pathway for extracellular CDNs to enter intracellular cytosols [45].

5.1. In Vitro Immunostimulatory Functions of CDNs

The in vitro immunostimulatory function of CDNs has been studied in cultured human and murine cells such as immature DCs and macrophages. These studies have identified DCs and macrophages as the major target cells of c-di-GMP and c-di-AMP. Analyses of DC subsets revealed conventional DCs as principal responders to c-di-AMP stimulation [47]. Both c-di-GMP and c-di-AMP were able to promote the activation and maturation of murine and human DCs [48,49] and induce the production of IFN-β and interleukin (IL)-6, but not TNF, in RAW264.7 macrophage cells and mouse primary bone marrow-derived macrophages (BMDMs) [50]. In cultured human immature DCs, c-di-GMP upgraded the surface expression of costimulatory molecules CD80 and CD86, maturation marker CD83, and MHC class II. c-di-GMP treatment also altered the expression of chemokine receptors C-C Motif Chemokine Receptor 1 (CCR1), CCR7, and C-X-C Motif Chemokine Receptor 4 (CXCR4), and increased the production of IL-12, IFN-γ, IL-8, CC chemokine ligand 2 (CCL2), C-X-C motif chemokine ligand 10 (CXCL10), and CCL5 by DCs. Studies on M1- and M2-polarized THP-1 and murine BMDMs showed that both murine and human M1-polarized macrophages showed increased STING expression and treatment with a STING agonist (DMXAA) reprogramed M2 macrophages toward an M1-like subtype [51].
c-di-GMP-matured DCs showed enhanced T-cell stimulatory activity [48,49]. Stimulation of bone marrow-derived DCs (BMDCs) with CDNs induced marked metabolic products (nitric oxide and the cytokine BAFF) that are different from those induced by TLR ligands [52]. The BAFF production appears to be correlated with the improved (i.e., more rapid and persistent) antibody responses in both aged and young mice [52]. Recent studies have also shown that c-di-GMP differentiated lung monocyte-derived DCs (MoDCs) into Bcl6+ mature MoDCs and promoted lung memory T helper cells through a process mediated by soluble TNF. MoDCs are essential for c-di-GMP-induced lung mucosal responses in this model and are responsible for lung IgA responses, but they are dispensable for vaccine-induced systemic immune responses [53]. In addition to type 1 IFN, physiologically relevant levels of c-di-AMP and c-di-GMP also stimulated a robust IL-1β production in murine BMDMs by the activation of NLRP3 inflammasome through a unique pathway associated with the recognition of intracellular infections, but independent of the STING pathway [54].
CDNs (such as c-di-GMP) also synergized with B cell receptor signals to directly activate B cells and promote antibody responses in a STING-dependent manner [55]. Similarly, costimulation of T cells with cGAMP and TCR ligands induced IFN-I production and regulated the growth and function of T cells through the mTORC1 pathway [56]. Studies in unfractionated human peripheral blood mononuclear cell (PBMC) cultures have shown that both 2′3′-cGAMP and 3′3′-cGAMP were highly potent in driving the expansion and maturation of functional antitumor or antiviral antigen-specific CD8+ T cells via the induction of type I IFNs [57,58,59]. 3′3′-cGAMP-primed HIV-1-specific CD8+ T cells produced higher levels of perforin and granzyme B expressions than lipopolysaccharides (LPS)- or TLR7/8 agonist R848-primed ones and were more potent to suppress HIV-1 [58,59]. c-di-AMP and, to a lesser extent, c-di-GMP also played a divergent role in regulating Porphyromonas gingivalis LPS-induced cytokine responses in human gingival keratinocytes [60]. c-di-AMP treatment increased the extracellular MCP-1 and vascular endothelial growth factor (VEGF) levels and intracellular IL-1 receptor antagonist level and restored decreased extracellular IL-8 levels induced by LPS. In the presence of LPS, c-di-GMP increased extracellular VEGF level, whereas c-di-AMP suppressed intracellular IL-1β levels [60].

5.2. In Vivo Immunostimulatory Functions of CDNs

In corroboration with the in vitro results, experimental studies in animal models have confirmed the immunostimulatory function of CDNs in inducing host innate immune responses and enhancing the adaptive immune responses. Systemic (intraperitoneal, i.p.) or mucosal (intranasal, i.n.) administration of mice with c-di-GMP induced an acute and transient local recruitment of inflammatory cells (neutrophils and macrophages) and activation of DCs [48,61,62,63]. Intranasal c-di-GMP administration also induced the production of chemokines CXCL1, CCL2, CCL3, CXCL2, and CCL5 [63]. Similarly, systemic or local administration of cGAMP induced high levels of type I IFNs in mice but minimal inflammatory gene expression [64]. Intranasal administration of c-di-GMP in mice greatly enhanced the antigen uptake by CD11c+ cells through both pinocytosis and receptor-mediated endocytosis, which was dependent on the increased STING expression [65]. In this regard, c-di-GMP selectively activated DCs with high pinocytosis efficiency and induced the production of IL-12p70, IFNγ, IL-5, IL-13, IL-23, IFNλ, and IL-6, but interestingly not IFNβ production, in this model [65].
The immunostimulatory properties of CDNs have been explored for their potential as novel immunotherapeutics for the treatment of infections, cancer, and other immunological disorders. Intranasal, subcutaneous (s.c.), intramuscular (i.m.), i.p., and intramammary administration with CDNs induced robust innate immune responses and enhanced host resistance against infection with a wide range of pathogenic bacteria, such as Bordetella pertussis, Acinetobacter baumannii, Staphylococcus aureus, Klebsiella pneumoniae, Clostridium perfringens, and various serotypes of Streptococcus pneumoniae [48,62,63,66,67,68]. Depletion of neutrophils abolished the protective role of c-di-GMP in the mouse model of Acinetobacter pneumonia, underscoring the importance of neutrophils as the critical effector cell in c-di-GMP-induced protection [63]. In addition, studies in a chicken model of C. perfringens infection have shown the promise of CDNs as an alternative to antibiotics without increasing the selection pressure for some β-lactamase genes or altering the commensal bacterial population [67].
Recent studies have shown that systemic treatment of mice with cGAMP inhibited the replication of genital herpes simplex virus (HSV) 2 and improved the clinical outcome of infection [64]. More importantly, direct application of CDNs, but not TLR agonists, on the genital epithelial surface increased local IFN activity and conferred total protection against disease even in immunocompromised mice without causing overt systemic responses [64]. Moreover, therapeutic administration of encapsulated-cGAMP microparticles (MPs), but not soluble cGAMP, protected mice from the development and relapse of experimental allergic encephalomyelitis in an IFN-I-dependent and -independent mechanism, and the therapeutic outcome was more effective than recombinant IFN-β [69]. In this regard, in vitro studies of PBMCs from relapsing-remitting multiple sclerosis patients showed that cGAMP MPs promoted both IFN-I and the immunoregulatory cytokines IL-27 and IL-10 responses [69].
The identification of the critical contribution of STING to antitumor immunity has generated a great enthusiasm to explore the intratumoral administration of STING agonists as standalone cancer immunotherapy or in combination with other cancer immunotherapeutic modalities such as immune checkpoint inhibitors and chimeric antigen receptor (CAR)-T-cell immunotherapies to enhance their efficacy (Figure 5). Immunologically, intratumoral injection of mice with various soluble or encapsulated synthetic and natural CDNs (such as RR-CDG, the R,R-stereoisomer of the phosphorothioate analog c-di-GMP-S2 or RR-c-di-GMP-S2; RR-cyclic-di-guanine (CDG), R(P),R(P) dithio-c-di-GMP, ADU-S100, the R,R-stereoisomer of the phorphorothioate analog 2′3′-cAAMP-S2 or RR-2′3′-cAAMP or RR-CDA; c-di-GMP, and cGAMP-NPs) induced enhanced type I IFN signalling, potent local inflammatory responses (DCs, macrophages, NK cells, and CXCL10 and CCL5 responses), reprograming of macrophages from protumorigenic M2-like type toward M1-like type, and the activation of adaptive immunity (CD4 and CD8 T cells, and T-cell migration) to increase tumour immunogenicity [70,71,72,73,74,75,76]. As a result, the treatment reprogramed the tumor microenvironment toward a more immunogenic antitumor milieu and induced profound regression and necrosis of established tumours. In many cases, the treatment-induced substantial and durable systemic tumour-specific Th1 adaptive immunity is capable of rejecting distant metastases and providing long-lived immunologic memory [72,73,74,76]. The intratumoral CDN treatment also increased the response rate to T-cell receptor activation by checkpoint-modulating antibodies to CTLA-4, PD-1, 4-1BB, and OX40, resulting in significant increases in median survival time in animal models of melanoma (B16-F10 and YUMM1.7), breast adenocarcinoma (E0771), prostate cancer (TRAMP-C2), head and neck squamous cell cancers (SCCFVII) [72,73], HER-2+ breast tumours [76], glioblastoma multiforme [77], glioma [70], neuroblastoma [75], and 4T1 breast tumour [72,74,78,79,80,81]. In this regard, a single dose of cGAMP-liposomal nanoparticles (cGAMP-NP) was sufficient to modulate the tumour microenvironment for effective control of programmed death-ligand 1 (PD-L1)-insensitive basal-like triple-negative breast cancer and melanoma and, more significantly, prevented the formation of secondary tumours in mice [82]. Moreover, coadministration of c-di-GMP with implantable biopolymer devices to deliver CAR T cells directly to the surfaces of solid tumours improved the effectiveness of CAR T-cell therapy and protected against the emergence of escape variants in immunocompetent orthotopic mouse models of pancreatic cancer and melanoma [83].
Studies in mouse models of breast cancer have shown that intratumoral administration of ADU-S100 (an analog of c-di-AMP) alone was able to induce durable tumour clearance in 100% of parental FVB/N mice that are able to mount potent immune responses to the implanted neu-positive tumour cells, but the treatment only resulted in tumour clearance in 10% of the FVB/N-derived neu/N transgenic mice that have well-established peripheral immune tolerance to the endogenous neu antigen [76,81]. Coadministration of ADU-S100 with anti-PD-L1 and anti-OX40 antibodies significantly enhanced the clearance rate to 40% of neu/N mice and also induced abscopal immunity [76,81]. The efficacy of combination treatment was well correlated with globally enhanced ratios of CD8+ T cells to regulatory T cells, macrophages, and myeloid-derived suppressor cells, and downregulation of the M2 marker CD206 on tumour-associated macrophages in this model [81]. Unfortunately, ADU-S100 failed to demonstrate sufficient clinical benefit in clinical trials.
CDNs have also been used with other immunomodulators, such as CpG oligonucleotides (ODN) and monophosphoryl lipid A (MPLA), for cancer immunotherapy [85,86]. Systemic delivery of c-di-GMP/MPLA-encapsulated immuno-nanoparticles induced extensive upregulation of antigen-presenting cells (APCs) and NK cells in the blood and tumour tissue and resulted in significant therapeutic benefit in a mouse model of metastatic triple-negative breast cancer [85]. Compared with single treatments, intratumoral injection of both CpG ODN and cGAMP significantly reduced tumour size in mouse models of lymphoma and melanoma [86].
Although the precise events induced by CDNs in the tumour microenvironment remain to be determined, mechanistic studies using knockout and bone marrow chimeric mice suggest that both stromal and immune cells (bone marrow-derived TNFα) and their crosstalk are essential to achieve CDN-induced tumour regression and clearance [87]. In this regard, cGAMP-induced STING activation promoted the polarization of tumour-associated macrophages into proinflammatory subtypes in spontaneous gastric cancer in p53(+/−) mice and cell line-based xenografts and induced apoptosis of gastric cancer cells through IL-6R-Janus kinase (JAK)-IL-24 pathway [88]. The potential role of macrophages in the CDN-induced antitumor activities has also been analyzed in detail. Intratumoral injection of cGAMP induced the transient accumulation of macrophages in the tumor tissue in mouse models of 4T1 breast cancer, squamous cell carcinomas, CT26 colon cancer, and B16F10 melanoma. Clodronate liposome-mediated macrophage depletion impaired the antitumor effect of cGAMP treatment [89]. It has also been reported that c-di-AMP-induced NK cell-mediated tumor rejection involved type I IFN-mediated activation of NK cells and the enhanced expression of IL-15 and IL-15 receptors [90].

6. CDNs as Potent Vaccine Adjuvants for Systemic and Mucosal Immunization

The potential adjuvant function of CDNs was first demonstrated in mice with model antigens beta-galactosidase (beta-Gal) and ovalbumin (Ova). Subcutaneous immunization with c-di-GMP or c-di-AMP and beta-Gal or Ova elicited potent antigen-specific serum IgG titers, a balanced Th1/Th2 response, and strong in vivo cytotoxic T lymphocyte (CTL) responses (i.e., 30–60% of antigen-specific lysis) [91]. When immunized by mucosal routes (i.n. or sublingual), these vaccines also induced specific secretory IgA responses in multiple mucosal sites (such as lung and vagina) [49,92]. Similar results were also observed when bis-(3′,5′)-cyclic dimeric inosine monophosphate (c-di-IMP) or cGAMP was used [93,94].
Compared to other licensed and experimental adjuvants such as LPS, CpG ODN, and aluminum salt, c-di-GMP appears to be more potent in inducing both humoral and Th1 immune responses (higher IgG2a, IFN-γ, TNF-α, and CXCL10 responses) in mice and in non-human primate PBMC cultures [95]. Mice immunized with c-di-GMP also showed a more predominant germinal centre formation, indicative of long-term memory, than the mice immunized with LPS or CpG [95]. Similarly, recent studies have shown that s.c. immunization of mice with c-di-AMP and Ova resulted in significantly higher levels of Ova-specific IgG and stronger Th1 and IFNγ-producing CD8+ memory T-cell response than the mice immunized with poly(I:C)/CpG and Ova [96].
The potency and immune profiles induced by different CDNs and their derivatives have not been systemically compared in a head-to-head manner. Overall, it appears that all CDNs are capable of stimulating the host’s innate immune responses but often with subtle differences. Based on the antigen-specific IgG1 to IgG2a ratio and cytokine response profiles, earlier studies suggest that c-di-GMP induces a dominant Th1 response, cGAMP induces a balanced Th1/Th2 response, whereas c-di-AMP and c-di-IMP promotes a balanced Th1/Th2/Th17 response [49,92,93,94]. In murine BMDCs and human DCs, it has also been shown that cGAMP induced fewer IL-17 responses than c-di-AMP [94], but 3′3′-cGAMP stimulated stronger IFN gene activation than c-di-GMP [97]. Molecular dynamics simulation analysis showed that human STING binds more strongly to 3′3′-cGAMP than to c-di-GMP [98], although CDN derivatives capable of activating all human STING alleles and murine STING have been successfully synthesized [71]. In addition, Yan et al. have chemically synthesized several c-di-GMP analogs (mono- and bisphosphorothioate (c-di-GMP-S1 and -S2)) and the 2′-fluoro analog of c-di-GMP (2′-F-c-di-GMP) and compared their immunostimulatory properties in mice. Although the innate immune responses induced by these analogs were generally comparable to those induced by the parental c-di-GMP [99,100], the pulmonary inflammatory responses (neutrophils and chemokine CXCL1, CCL4, and CCL5) induced by the two phosphorothioate analogs (S1 and S2), particularly the c-di-GMP-S1, were much milder than the parent c-di-GMP [99]. On the other hand, 2′-F-c-di-GMP was effective when administered orally [100].

6.1. CDNs as Adjuvants for the Development of Vaccines against Infectious Diseases

The potential of CDNs as effective vaccine adjuvants for systemic and mucosal immunization against infectious diseases was first demonstrated in the development of vaccines against pneumococcal and S. aureus infections [48,62]. Intraperitoneal immunization of mice with c-di-GMP and pneumolysin toxoid (PdB) or pneumococcal surface protein A (PspA) induced significantly higher antigen-specific antibody responses and increased survival of mice after S. pneumoniae challenges, as compared to alum adjuvant [62]. Moreover, i.n. immunization of mice with pneumococcal surface adhesion A (PsaA) and c-di-GMP elicited strong antigen-specific serum IgG and secretory IgA responses at multiple mucosal surfaces and significantly reduced nasopharyngeal S. pneumoniae colonization in the immunized mice [61]. This study provided the first evidence of c-di-GMP as an effective mucosal adjuvant to induce protective immunity against mucosal bacterial infection. It appears that, in mice, c-di-GMP induces stronger immune responses and better protection against S. pneumoniae challenge than cGAMP-adjuvanted vaccine [65].
Many groups have subsequently explored the possibility of CDNs to improve the vaccine-induced immune profiles and the duration of immunity against a number of bacterial infections of human and veterinary importance (Table 1). Tuberculosis (TB), caused by Mycobacterium tuberculosis, remains a significant public health threat in many regions. Bacillus Calmette-Guerin (BCG), the live attenuated TB vaccine developed more than a century ago, is the most commonly used vaccine worldwide. However, BCG has varied protective efficiency in adults and safety concerns in the immunocompromised population. Several laboratories have attempted to use CDNs to enhance the current BCG vaccines or develop new, safe and effective subunit TB vaccines. Early efforts to introduce the c-di-GMP phosphodiesterase gene Rv1357c into BCG Pasteur failed to provide better protection against M. tuberculosis challenge than conventional BCG in mice [101]. Vaccination with another modified BCG strain (BCGΔBCG1419c) lacking the c-di-GMP phosphodiesterase gene BCG1419c induced a better activation of specific T lymphocytes, moderate protection, and improved resistance to infection reactivation in mice, as compared to vaccination with the conventional BCG vaccine [102]. Two groups have independently generated two recombinant BCG vaccines (rBCG-DisA), which overexpress the endogenous mycobacterial diadenylate cyclase (DisA) gene and produce high levels of c-di-AMP. Compared to conventional BCG vaccines, both rBCG-DisA vaccine candidates induced stronger immune responses, but one failed to provide additional protection against M. tuberculosis infection in mice [103], whereas the other showed significantly reduced lung weights, pathology scores, and M. tuberculosis burdens in guinea pigs [104].
When the CDN is used as an adjuvant for subunit TB vaccines, it enhances antigen-specific Th1 responses and elicits long-lasting protective immunity to M. tuberculosis. Immunization of mice with recombinant Rv3852 (H-NS) and cGAMP resulted in an enhanced antigen-specific IL-2 response by splenic lymphocytes, high serum levels of IL-2, IL-12p40, and TNF-α, and better protective efficacy (reduced bacterial counts in the spleen, but not in the lung) in mice challenged with M. tuberculosis H37Rv [133]. Similarly, s.c. immunization of a subunit TB vaccine consisting of a five M. tuberculosis antigen (antigen-85B, ESAT-6, Rv1733c, Rv2626c, and RpfD) fusion protein and a synthetic CDN (RR-CDG) induced protection equivalent to that induced by BCG vaccine. Interestingly, the protection in this study was STING-dependent but type I IFN-independent and correlated with an increased frequency of a subset of CXCR3-expressing T cells in the lung tissue. More significantly, i.n. immunization of this vaccine candidate induced protection superior to that induced by the BCG vaccine and elicited both Th1 and Th17 immune responses, the latter of which was correlated with enhanced protection [139]. On the other hand, i.n. immunization of mice with ESAT-6 and c-di-AMP failed to reduce the bacterial burdens after an i.v. challenge as compared to the mice immunized with ESTA-6 alone [119].
Failure to induce potent Th1 immune responses and long-term immunity and protect against nasal colonization and transmission of Bordetella pertussis by the current acellular pertussis (aP) vaccines have contributed to the recent global resurgence of pertussis (whooping cough). Intranasal immunization of mice with an experimental aP vaccine formulated with c-di-GMP and LP1569, a TLR2 agonist from B. pertussis (LP-GMP), induced potent antigen-specific Th1 and Th17 responses and conferred long-lasting protection (for 10 months) against nasal colonization and lung infection with B. pertussis [111]. The long-term protection against nasal colonization was correlated to IL-17-secreting respiratory tissue-resident memory CD4 T cells. Others have studied the adjuvant activity of cGAMP for parenteral and mucosal immunization with recombinant Helicobacter pylori urease A, urease B, and neutrophil-activating protein [132]. The gastric mucosal H. pylori colonization was significantly reduced in mice immunized by i.n. and, to a lesser degree, s.c. route, but not by the i.m. route. The protection efficacy appeared to be associated with the level of antigen-specific Th1 and particularly Th17 responses [132]. Moreover, Li et al. have demonstrated the potential of 2′-Fluoro-c-di-GMP as an oral vaccine adjuvant. Oral immunization of mice with H. pylori cell-free sonicate extract or flagellin proteins from C. difficile and Listeria monocytogenes in combination with 2′-Fluoro-c-di-GMP induced antigen-specific serum IgG and mucosal IgA responses and showed a significant reduction in the gastric bacterial colonization when mice were orally challenged with H. pylori [100].
The adjuvant function of CDNs has also been demonstrated in experimental vaccines against other bacterial and parasitic infections, including bacteria of antimicrobial resistance and biodefense significance. Subcutaneous immunization of mice with c-di-GMP and S. aureus antigen, clumping factor A (ClfA), or a nontoxic mutant staphylococcal enterotoxin C (mSEC) significantly reduced the bacterial burdens in the spleen and liver and improved the day 7 survival rates (87.5%) than those of the non-immunized mice (33.3%) after i.v., challenge with methicillin-resistant S. aureus [48,112]. Similarly, sublingual immunization of mice with Bacillus anthracis protective antigen (PA) and 3′3′-cGAMP induced comparable levels of PA-specific serum IgG and saliva IgA responses to those induced after immunization with PA and cholera toxin (CT), the golden standard mucosal adjuvant [138]. A single dose immunization with F1-V antigens of Yersinia pestis and RR-CDG induced 95% protection against a lethal challenge with Y. pestis CO92 within 14 days post-immunization [140]. Encapsulation of F1-V within nanoparticles further improved the efficacy and duration of the protection at a high challenge dose (≥7000 colony-forming units) in that 75% of immunized mice were protected from challenge at 182 days post-immunization [140]. Others have shown that the combination of the lipopeptide-based nano carrier system with c-di-AMP significantly reduced the antigen dose of a peptide vaccine and protected mice against a respiratory challenge with a lethal dose of a heterologous S. pyogenes strain [120]. Moreover, immunization of mice with several different recombinant antigens (a chimeric antigen Traspain, Tc52 or its N- and C-terminal domains) of Trypanosoma cruzi and c-di-AMP induced stronger protection against infection with higher survival rates, lower parasitemia, and reduced weight loss and chronic inflammation, compared to those immunized with other adjuvants [123,124,125]. Finally, c-di-GMP has also been explored for the development of live attenuated vaccines against salmonellosis (ΔXIII) [141,142] and mycoplasmal infections [126]. Under these circumstances, the CDNs were either overexpressed in the live attenuated vaccine or delivered together with cationic liposomes [126].
The promise of CDNs as systemic and mucosal adjuvants for influenza vaccines has been extensively studied with different influenza vaccines and antigens, including hemagglutinin (HA), neuraminidase (NA), matrix protein 2 ectodomain (M2e), and inactivated viruses. Overall, these studies have demonstrated CDNs as effective adjuvants by promoting potent, long-lasting mucosal and systemic humoral (IgA, IgG, IgG isotypes, and hemagglutination inhibition (HI) titers) and cellular immune responses (high frequencies of virus-specific polyfunctional CD4+ T cells producing one or more Th1 cytokines (IFN-γ, IL-2, TNF-α), and protection against challenges with homologous and heterologous viral strains (H1N1, H5N1, H3N2, H5N1, H7N9, and H9N2) in mice and ferrets [105,106,107,108,109,117,118,127].
Several studies have shown that the combination of CDNs with other adjuvants and vaccine delivery systems (such as chitosan, silica nanoparticle, acetylated dextran microparticles, and microneedle) further enhanced the immunogenicity, dose-sparing, and cross-clade protection of the experimental or licensed influenza vaccines [107,108,110,117,127,137]. In this regard, formulation of an H1N1 vaccine with pulmonary surfactant-biomimetic liposomes encapsulating cGAMP elicited cross-protection against distant H1N1 and heterosubtypic H3N2, H5N1, and H7N9 viruses for at least 6 months [130]. Similarly, immunization of mice with cGAMP-adjuvanted inactivated H7N9 vaccine also provided effective cross-protection against H1N1, H3N2, and H9N2 influenza viruses, induced significantly higher nucleoprotein-specific CD4+ and CD8+ T-cell responses in immunized mice and upregulated the IFN-γ and granzyme B expression in the lung tissue of mice in the early stages post a heterosubtypic virus challenge [127]. The adjuvant effect and safety of cGAMP were further confirmed in a pig model of intradermal immunization with the H5N1 influenza vaccine [129]. The extensive studies of CDNs in influenza vaccine development have also demonstrated the broad application of CDNs in multiple animal species (mice, rats, guinea pigs, ferrets, cattle, pigs, and chickens) by different immunization routes (i.n., i.m. intratracheal, intradermal, and sublingual).
However, whether CDNs can effectively enhance specific immune responses in aged or newborns remains to be determined [131,136]. Borriello et al. found that although cGAMP induced a comparable expression of surface maturation markers in newborn and adult BMDCs, cGAMP adjuvantation alone failed to increase HA-specific antibody responses in newborn mice [136]. However, immunization of puppies with cGAMP formulated with alum (Alhydrogel)-adjuvanted trivalent recombinant HA influenza vaccine (Flublok) significantly enhanced HA-specific IgG2a/c titers by six weeks of age [136]. Immunization with cGAMP+alum also enhanced IFNγ production by CD4+ T cells and increased both the absolute numbers and percentages of CD4+CXCR5+PD-1+ T follicular helper cells and GL-7+CD138+ germinal centre B cells. Similarly, immunization of an H1N1 influenza vaccine with cGAMP and saponin (Quil-A) significantly improved the survival (by 80–100%) of vaccinated 20-month-old mice, whereas cGAMP alone showed no significant improvement [143]. In this regard, recent studies found that targeting MoDCs with TNF fusion proteins restored the c-di-GMP adjuvantivity in aged mice [144].
The adjuvant activity of CDNs has also been demonstrated in vaccines against other viral infections, such as hepatitis B surface antigen (cGAMP) [135], HCV rE1E2 (c-di-AMP and archaeosomes) [121], HIV-1 reverse transcriptase (c-di-GMP) [113], mutant human papillomavirus (HPV) 16 E7 (E7GRG) protein (cGAMP and CpG-C ODN) [134], and foot and mouth disease vaccines in cattle and pigs [114]. In this regard, therapeutic immunization of female C57BL/6 mice with E7GRG formulated with cGAMP and CpG-C ODN induced significantly suppressed TC-1 tumour growth in mice [134].

6.2. CDNs as Adjuvants for the Development of Cancer Vaccines

The potential of CDNs in the development of vaccines against chronic, non-communicable diseases, particularly cancer, has also been investigated. It has been well recognized that effective therapeutic cancer vaccines require potent adjuvants to induce robust type I IFN and proinflammatory cytokine responses in the tumour microenvironment. CDNs, such as c-di-GMP, c-di-AMP, and cGAMP, are excellent adjuvants for the development of cancer vaccines because they have the potential to provide “two punches” to eliminate the tumour: activating immunogenic tumour cell death directly and inducing type 1 IFN production to enhance tumour-specific Th1 and CTL responses (Figure 5). Targeting both pathways is likely to result in a nearly complete local tumour regression and elimination of metastases [145]. Despite these rationales, relatively few studies have evaluated CDNs as a cancer vaccine adjuvant. This is perhaps due to the limited numbers of tumour-specific or associated antigens identified and available for cancer vaccine development. Chandra et al. reported that therapeutic immunization of metastatic breast cancer-bearing mice (4T1 model) with an attenuated L. monocytogenes (LM) expressing tumour-associated antigen MAGE-B (LM-Mb), followed by multiple low doses of c-di-GMP (0.2 μmol/L) treatment, resulted in almost complete elimination of all metastases. In this model, it appears that low doses of c-di-GMP significantly increased the production of IL-12 by myeloid-derived suppressor cells and improved antigen-specific T-cell responses, while high doses of c-di-GMP (range: 0.3–3 mmol/L) killed the 4T1 tumour cells directly by activating the tumour cell caspase-3 pathway [146].
CDNs have also been evaluated with a number of other cancer antigens and cancer vaccines, including MUC1 glycopeptide [147], a B16 melanoma peptide [148], and a mouse model of glioma [70] and EG7-Ova tumour [57]. In these applications, c-di-GMP and cGAMP functioned as potent immunostimulants and induced robust antitumor immunity and tumour regression. Shae et al. have recently developed a synthetic cancer nanovaccine platform (nanoSTING-vax) for efficient co-delivery of tumour peptide antigens and cGAMP to the cytosol of APCs [149]. Therapeutic immunization with this platform, in combination with immune checkpoint blockade, induced significant suppression of tumour growth, even complete tumour rejection, and long-lasting antitumor immune memory in multiple murine tumor models.

7. Novel Delivery Strategies to Enhance the Immunostimulatory and Adjuvant Function of CDNs

The in vivo efficacy of CDNs is limited by their physiochemical properties such as negative charges, hydrophilicity, short plasma half-life, and poor cellular targeting and membrane permeability. To overcome these challenges, researchers have developed several innovative strategies to transport CDNs more efficiently to the cell cytosol where STING is located (Table 2). These include in vivo synthesis of CDNs, nanoparticles, and microparticles and combination with other adjuvants and immunostimulators, etc. In this regard, the adjuvant activity of CDNs can be simply further enhanced or tailored by combination with other adjuvants and immunomodulators, particularly those that activate different immune signalling pathways, such as TLR agonists (CpG ODN, lipid A, and Pam3CSK), macrophage inducible C-type lectin (Mincle), saponin, and alum, etc. [97,114,150,151]. Others have attempted to synthesize c-di-GMP in vivo by transducing a diguanylate cyclase gene into the nonreplicating adenovirus serotype 5 (Ad5) vector, which produced enhanced amounts of c-di-GMP when expressed in mammalian cells in vivo [152,153]. Furthermore, cGAMP has been incorporated into viral particles such as lentivirus and herpesvirus virions [154]. The proof of concept of these approaches has been demonstrated in animal models with extracellular protein Ova, HIV-1 Gag peptides, or the C. difficile toxin B [152,153]. Quintana and colleagues have explored the feasibility to engineer a live Lactococcus lactis vector containing a single plasmid with both cdaA and tscf genes under different promoters, respectively, to simultaneously synthesize the adjuvant c-di-AMP as well as a heterologous vaccine antigen of interests in order to develop a simple and economical system for vaccine development [155]. However, control of the level and duration of the CDN expression by this approach can be a challenge since chronic STING activation has been involved and responsible for initiating certain inflammatory and autoimmune diseases due to type 1 IFN overproduction [4,156,157] In addition, sustained gene transcription is rigidly prevented by the host to avoid lethal STING-dependent proinflammatory disease by unknown mechanisms [158,159].
Substantial efforts have been made to develop novel delivery systems (such as different compositions of microparticles, liposomes, and nanoparticles, STINGel, and microneedles) to facilitate the efficient and targeted CDN delivery [162,168,169,170,171,172,173]. Compared with unformulated (free) CNDs, these formulations have a markedly prolonged half-life and local retention of CDNs within the tumour microenvironment, enhanced the local infiltration of CD11c+ DCs, F4/80+ macrophages, CD4+ T cells, and CD8+ T cells, stimulated APC activation (high expression of CD80, CD86 and MHC class I), induced the production of proinflammatory cytokines (type I and type II IFN, IL-1β, and IL-6), dramatically increased expansion and function of antigen-specific CD8+ T cells, and facilitated NK cell-mediated MHC-I non-restricted antitumor immunity [74,77,78,79,160,161,174].

8. Conclusions and Future Direction

Most of the recent research on the immunostimulatory and adjuvant function of CDNs has focused on the development of vaccines against bacterial and viral infections, particularly in influenza and tuberculosis. However, it is encouraging to note that CDNs have been increasingly used as vaccine adjuvants for some challenging infections and for their veterinary application. The identification of STING as a crucial pathway in cancer immunology generated a great interest in the research and development of CDNs for cancer immunotherapy and cancer vaccine adjuvants, which has led to several human clinical trials. As adjuvants for prophylactic vaccines for infectious diseases, CDNs induce potent humoral and cellular memory immune responses in the systemic and mucosal compartments. These immune responses often provide long-lasting immunity, cross-protection, and antigen dose-sparing, although more studies are needed to determine if CDNs are able to enhance the immunity in aged or newborns. As adjuvants for therapeutic cancer vaccines, CDNs induce potent antitumor immunity, including the activation of cytotoxic T cells, NK cells, and tumor-associated macrophages, and achieve durable regression in multiple mouse models of tumours.
Preclinical studies have shown that CDNs function in multiple animal species (mice, rats, guinea pigs, ferrets, chicken, pigs, and cattle). These studies also indicate that CDNs can be administered through different parenteral and mucosal routes to induce the desired systemic and local immune responses. Human cell culture and molecular modelling indicate that CDNs bind to human STING and function in vitro. CDNs have several inherent advantages as an ideal vaccine adjuvant, and these include the relative ease to synthesize and formulate, known mode of action, and a large amount of preclinical efficacy data. Several novel strategies and approaches, such as in vivo synthesis, chemical modifications, combination with other vaccine adjuvants and delivery systems, have been developed over the years to improve the cellular uptake of CDNs and facilitate their cytosolic delivery to further enhance their efficacy and application. In addition, several novel methods of enzymatic and microbiological synthesis of CDNs for scale-up have been developed. Among these, the delivery via in vivo synthesis is an attractive strategy, but appropriate control of their expression level and duration to prevent the development of autoimmune diseases will be a challenge.
Although an overwhelming number of studies have convincingly demonstrated the promise of CDNs as an effective and safe adjuvant for mucosal and systemic vaccination, a few studies have shown no or partial effect of CDNs. Intranasal immunization of mice with BtaF trimeric autotransporter of B. suis and c-di-AMP conferred significant protection against intragastric B. suis challenge but failed to protect against the respiratory challenge, despite the presence of vaccine-induced specific mucosal IgA and the production of high IFN-γ levels by lung cells [122]. Furthermore, immunization of pigs with c-di-AMP and naked self-amplifying replicon RNA (RepRNA) encoding influenza virus nucleoprotein and HA induced only low-level immune responses in 20% of pigs [175].
Despite the promising progress and more than 15 years of R&D efforts by the biomedical research communities and pharm/biotech industries, the research on CDNs as vaccine adjuvants seems stalled at the preclinical development stage. To date, there are no licensed CDN products for human clinical use. Several clinical trials of the leading CDNs for cancer immunotherapy have been conducted with disappointing results. The reasons for this remain unknown, but it was suggested that the heterogeneity of the human STING gene and the potential immunocompromised status and immunosenescence in clinical trial participants are possible contributing factors [176]. In this regard, single nucleotide polymorphism analysis showed the common presence of STING variants and mutations in the human population (ranged from 1.5% in R293Q to 20.4% in R71H-G230A-R293Q (HAQ) [177]). Therefore, further studies should be directed to dissect the differences in immune responses induced by different CDNs and their mode of action between human and experimental animal cells. In addition, more stringent preclinical research and development in clinically relevant animal models will be needed to promote the clinical application of these promising molecules. Finally, strategies to design “pan-STING agonists” as prophylactic vaccine adjuvants should be considered to ensure their function in a majority of the population.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/vaccines9080917/s1, Figure S1: Selected CDNs containing 3′-5′ internucleotide phosphate/phosphorothioate linkages reported in the literature, Figure S2: Selected CDNs containing 2′-5′ and both 2′-5′ and 3′-5′ internucleotide phosphate/phosphorothioate linkages reported in the literature, Figure S3: Selected CDN analogs with internucleotide linkages that are not phosphorous based, and Figure S4: c-di-GMP conjugates reported.

Author Contributions

H.Y. and W.C. developed the concept, drafted the manuscript, and agreed to the published version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Natural Sciences and Engineering Research Council of Canada (RGPIN-2020-07040) and the National Research Council Canada (Vaccine Program and Vaccine and Immunotherapeutics Program). The view expressed here (W.C.) is solely the responsibility of the author and does not necessarily represent the official views of the National Research Council Canada.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Hongyan Zhou for her assistance in the literature search and the preparation of Table 1 and Table 2 in this manuscript.

Conflicts of Interest

The authors are inventors of U.S. Patent 10,092,644, which is owned by Brock University and the National Research Council Canada (Government of Canada).

References

  1. Ahn, J.; Barber, G.N. STING signaling and host defense against microbial infection. Exp. Mol. Med. 2019, 51, 1–10. [Google Scholar] [CrossRef]
  2. Burdette, D.L.; Monroe, K.M.; Sotelo-Troha, K.; Iwig, J.S.; Eckert, B.; Hyodo, M.; Hayakawa, Y.; Vance, R.E. STING is a direct innate immune sensor of cyclic di-GMP. Nature 2011, 478, 515–518. [Google Scholar] [CrossRef]
  3. Ouyang, S.; Song, X.; Wang, Y.; Ru, H.; Shaw, N.; Jiang, Y.; Niu, F.; Zhu, Y.; Qiu, W.; Parvatiyar, K.; et al. Structural Analysis of the STING Adaptor Protein Reveals a Hydrophobic Dimer Interface and Mode of Cyclic di-GMP Binding. Immunity 2012, 36, 1073–1086. [Google Scholar] [CrossRef] [Green Version]
  4. Barber, G.N. STING-dependent cytosolic DNA sensing pathways. Trends Immunol. 2014, 35, 88–93. [Google Scholar] [CrossRef] [PubMed]
  5. Wu, J.-J.; Zhao, L.; Hu, H.; Li, W.; Li, Y. Agonists and inhibitors of the STING pathway: Potential agents for immunotherapy. Med. Res. Rev. 2020, 40, 1117–1141. [Google Scholar] [CrossRef] [PubMed]
  6. Ross, P.; Aloni, Y.; Weinhouse, C.; Michaeli, D.; Weinberger-Ohana, P.; Meyer, R.; Benziman, M. An unusual guanyl oligonucleotide regulates cellulose synthesis in Acetobacter xylinum. FEBS Lett. 1985, 186, 191–196. [Google Scholar] [CrossRef] [Green Version]
  7. Ross, P.; Aloni, Y.; Weinhouse, H.; Michaeli, D.; Weinberger-Ohana, P.; Mayer, R.; Benziman, M. Control of cellulose synthesis Acetobacter xylinum. A unique guanyl oligonucleotide is the immediate activator of the cellulose synthase. Carbohydr. Res. 1986, 149, 101–117. [Google Scholar] [CrossRef]
  8. Ross, P.; Weinhouse, H.; Aloni, Y.; Michaeli, D.; Weinberger-Ohana, P.; Mayer, R.; Braun, S.; de Vroom, E.; van der Marel, G.A.; van Boom, J.H.; et al. Regulation of cellulose synthesis in Acetobacter xylinum by cyclic diguanylic acid. Nature 1987, 325, 279–281. [Google Scholar] [CrossRef]
  9. Egli, M.; Gessner, R.V.; Williams, L.D.; Quigley, G.J.; van der Marel, G.A.; van Boom, J.H.; Rich, A.; Frederick, C.A. Atomic-resolution structure of the cellulose synthase regulator cyclic diguanylic acid. Proc. Natl. Acad. Sci. USA 1990, 87, 3235–3239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Witte, G.; Hartung, S.; Büttner, K.; Hopfner, K.-P. Structural Biochemistry of a Bacterial Checkpoint Protein Reveals Diadenylate Cyclase Activity Regulated by DNA Recombination Intermediates. Mol. Cell 2008, 30, 167–178. [Google Scholar] [CrossRef]
  11. Stülke, J.; Krüger, L. Cyclic di-AMP Signaling in Bacteria. Annu. Rev. Microbiol. 2020, 74, 159–179. [Google Scholar] [CrossRef] [PubMed]
  12. Yin, W.; Cai, X.; Ma, H.; Zhu, L.; Zhang, Y.; Chou, S.-H.; Galperin, M.Y.; He, J. A decade of research on the second messenger c-di-AMP. FEMS Microbiol. Rev. 2020, 44, 701–724. [Google Scholar] [CrossRef] [PubMed]
  13. Davies, B.; Bogard, R.W.; Young, T.S.; Mekalanos, J.J. Coordinated Regulation of Accessory Genetic Elements Produces Cyclic Di-Nucleotides for V. cholerae Virulence. Cell 2012, 149, 358–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Sun, L.; Wu, J.; Du, F.; Chen, X.; Chen, Z.J. Cyclic GMP-AMP Synthase Is a Cytosolic DNA Sensor That Activates the Type I Interferon Pathway. Science 2013, 339, 786–791. [Google Scholar] [CrossRef] [Green Version]
  15. Wu, J.; Sun, L.; Chen, X.; Du, F.; Shi, H.; Chen, C.; Chen, Z.J. Cyclic GMP-AMP Is an Endogenous Second Messenger in Innate Immune Signaling by Cytosolic DNA. Science 2013, 339, 826–830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Zhang, X.; Shi, H.; Wu, J.; Zhang, X.; Sun, L.; Chen, C.; Chen, Z.J. Cyclic GMP-AMP containing mixed phosphodiester linkages is an endogenous high-affinity ligand for STING. Mol. Cell. 2013, 51, 226–235. [Google Scholar] [CrossRef] [Green Version]
  17. Dahlstrom, K.M.; O’Toole, G.A. A Symphony of Cyclases: Specificity in Diguanylate Cyclase Signaling. Annu. Rev. Microbiol. 2017, 71, 179–195. [Google Scholar] [CrossRef]
  18. Ross, P.; Mayer, R.; Weinhouse, H.; Amikam, D.; Huggirat, Y.; Benziman, M.; de Vroom, E.; Fidder, A.; de Paus, P.; Sliedregt, L.A. The cyclic diguanylic acid regulatory system of cellulose synthesis in Acetobacter xylinum. Chemical synthesis and biological activity of cyclic nucleotide dimer, trimer, and phosphothioate derivatives. J. Biol. Chem. 1990, 265, 18933–18943. [Google Scholar] [CrossRef]
  19. Amikam, D.; Galperin, M. PilZ domain is part of the bacterial c-di-GMP binding protein. Bioinformatics 2005, 22, 3–6. [Google Scholar] [CrossRef]
  20. Ryjenkov, D.A.; Simm, R.; Romling, U.; Gomelsky, M. The PilZ domain is a receptor for the second messenger c-di-GMP: The PilZ domain protein YcgR controls motility in enterobacteria. J. Biol. Chem. 2006, 281, 30310–30314. [Google Scholar] [CrossRef] [Green Version]
  21. Alm, R.A.; Bodero, A.J.; Free, P.D.; Mattick, J. Identification of a novel gene, pilZ, essential for type 4 fimbrial biogenesis in Pseudomonas aeruginosa. J. Bacteriol. 1996, 178, 46–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Christen, M.; Christen, B.; Allan, M.G.; Folcher, M.; Jenö, P.; Grzesiek, S.; Jenal, U. DgrA is a member of a new family of cyclic diguanosine monophosphate receptors and controls flagellar motor function in Caulobacter crescentus. Proc. Natl. Acad. Sci. USA 2007, 104, 4112–4117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Martínez-Granero, F.; Navazo, A.; Barahona, E.; Redondo-Nieto, M.; de Heredia, E.G.; Baena, I.; Martín-Martín, I.; Rivilla, R.; Martín, M. Identification of flgZ as a Flagellar Gene Encoding a PilZ Domain Protein That Regulates Swimming Motility and Biofilm Formation in Pseudomonas. PLoS ONE 2014, 9, e87608. [Google Scholar] [CrossRef] [Green Version]
  24. Pratt, J.T.; Tamayo, R.; Tischler, A.; Camilli, A. PilZ Domain Proteins Bind Cyclic Diguanylate and Regulate Diverse Processes in Vibrio cholerae. J. Biol. Chem. 2007, 282, 12860–12870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Li, Z.; Chen, J.-H.; Hao, Y.; Nair, S.K. Structures of the PelD Cyclic Diguanylate Effector Involved in Pellicle Formation in Pseudomonas aeruginosa PAO1. J. Biol. Chem. 2012, 287, 30191–30204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Newell, P.D.; Monds, R.D.; O’Toole, G.A. LapD is a bis-(3′,5′)-cyclic dimeric GMP-binding protein that regulates surface attachment by Pseudomonas fluorescens Pf0-1. Proc. Natl. Acad. Sci. USA 2009, 106, 3461–3466. [Google Scholar] [CrossRef] [Green Version]
  27. Hickman, J.W.; Harwood, C.S. Identification of FleQ from Pseudomonas aeruginosa as a c-di-GMP-responsive transcription factor. Mol. Microbiol. 2008, 69, 376–389. [Google Scholar] [CrossRef] [Green Version]
  28. Fazli, M.M.; O’Connell, A.; Nilsson, M.; Niehaus, K.; Dow, J.M.; Givskov, M.; Ryan, R.P.; Tolker-Nielsen, T. The CRP/FNR family protein Bcam1349 is a c-di-GMP effector that regulates biofilm formation in the respiratory pathogen Burkholderia cenocepacia. Mol. Microbiol. 2011, 82, 327–341. [Google Scholar] [CrossRef]
  29. Smith, K.D.; Shanahan, C.A.; Moore, E.L.; Simon, A.C.; Strobel, S.A. Structural basis of differential ligand recognition by two classes of bis-(3′-5′)-cyclic dimeric guanosine monophosphate-binding riboswitches. Proc. Natl. Acad. Sci. USA 2011, 108, 7757–7762. [Google Scholar] [CrossRef] [Green Version]
  30. Sudarsan, N.; Lee, E.R.; Weinberg, Z.; Moy, R.; Kim, J.N.; Link, K.H.; Breaker, R.R. Riboswitches in Eubacteria Sense the Second Messenger Cyclic Di-GMP. Science 2008, 321, 411–413. [Google Scholar] [CrossRef] [Green Version]
  31. Lee, E.R.; Baker, J.L.; Weinberg, Z.; Sudarsan, N.; Breaker, R.R. An Allosteric Self-Splicing Ribozyme Triggered by a Bacterial Second Messenger. Science 2010, 329, 845–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Roy, A.B.; Petrova, O.E.; Sauer, K. Extraction and Quantification of Cyclic Di-GMP from P. aeruginosa. Bio Protoc. 2013, 3, e828. [Google Scholar] [CrossRef] [Green Version]
  33. Wang, C.; Sinn, M.; Stifel, J.; Heiler, A.C.; Sommershof, A.; Hartig, J.S. Synthesis of All Possible Canonical (3′-5′-Linked) Cyclic Di-nucleotides and Evaluation of Riboswitch Interactions and Immune-Stimulatory Effects. J. Am. Chem. Soc. 2017, 139, 16154–16160. [Google Scholar] [CrossRef] [Green Version]
  34. Rao, F.; Pasunooti, S.; Ng, Y.; Zhuo, W.; Lim, L.; Liu, A.W.; Liang, Z.-X. Enzymatic synthesis of c-di-GMP using a thermophilic diguanylate cyclase. Anal. Biochem. 2009, 389, 138–142. [Google Scholar] [CrossRef] [PubMed]
  35. Korovashkina, A.S.; Rymko, A.N.; Kvach, S.V.; Zinchenko, A. Enzymatic synthesis of c-di-GMP using inclusion bodies of Ther-motoga maritima full-length diguanylate cyclase. J. Biotechnol. 2012, 164, 276–280. [Google Scholar] [CrossRef] [PubMed]
  36. Shchokolova, A.S.; Rymko, A.N.; Kvach, S.V.; Shabunya, P.S.; Fatykhava, S.A.; Zinchenko, A.I. Enzymatic synthesis of 2′-ara and 2′-deoxy analogues of c-di-GMP. Nucleosides Nucleotides Nucleic Acids 2015, 34, 416–423. [Google Scholar] [CrossRef]
  37. Launer-Felty, K.D.; Strobel, S.A. Enzymatic synthesis of cyclic dinucleotide analogs by a promiscuous cyclic-AMP-GMP syn-thetase and analysis of cyclic dinucleotide responsive riboswitches. Nucleic Acids Res. 2018, 46, 2765–2776. [Google Scholar] [CrossRef] [PubMed]
  38. Becker, M.; Nikel, P.; Andexer, J.; Lütz, S.; Rosenthal, K. A Multi-Enzyme Cascade Reaction for the Production of 2′3′-cGAMP. Biomolecules 2021, 11, 590. [Google Scholar] [CrossRef]
  39. Li, L.; Yin, Q.; Kuss, P.; Maliga, Z.; Millan, J.L.; Wu, H.; Mitchison, T.J. Hydrolysis of 2′3′-cGAMP by ENPP1 and design of nonhydrolyzable analogs. Nat. Chem. Biol. 2014, 10, 1043–1048. [Google Scholar] [CrossRef] [Green Version]
  40. Rosenthal, K.; Becker, M.; Rolf, J.; Siedentop, R.; Hillen, M.; Nett, M.; Lütz, S. Catalytic promiscuity of cGAS: A facile enzymatic synthesis of 2′-3′-Linked cyclic dinucleotides. Chembiochem Eur. J. Chem. Biol. 2020, 21, 3225–3228. [Google Scholar] [CrossRef]
  41. Dubensky, J.T.W.; Kanne, D.B.; Leong, M.L. Rationale, progress and development of vaccines utilizing STING-activating cyclic dinucleotide adjuvants. Ther. Adv. Vaccines 2013, 1, 131–143. [Google Scholar] [CrossRef]
  42. Parvatiyar, K.; Zhang, Z.; Teles, R.; Ouyang, S.; Jiang, Y.; Iyer, S.S.; Zaver, S.A.; Schenk, M.; Zeng, S.; Zhong, W.; et al. The helicase DDX41 recognizes the bacterial secondary messengers cyclic di-GMP and cyclic di-AMP to activate a type I interferon immune response. Nat. Immunol. 2012, 13, 1155–1161. [Google Scholar] [CrossRef] [Green Version]
  43. McFarland, A.P.; Luo, S.; Ahmed-Qadri, F.; Zuck, M.; Thayer, E.F.; Goo, Y.A.; Hybiske, K.; Tong, L.; Woodward, J.J. Sensing of Bacterial Cyclic Dinucleotides by the Oxidoreductase RECON Promotes NF-kappaB Activation and Shapes a Proinflammatory Antibacterial State. Immunity 2017, 46, 433–445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Blaauboer, S.M.; Gabrielle, V.D.; Jin, L. MPYS/STING-Mediated TNF-α, Not Type I IFN, Is Essential for the Mucosal Adjuvant Activity of (3′–5′)-Cyclic-Di-Guanosine-Monophosphate In Vivo. J. Immunol. 2013, 192, 492–502. [Google Scholar] [CrossRef] [Green Version]
  45. Chang, D.; Whiteley, A.T.; Gwilt, K.B.; Lencer, W.I.; Mekalanos, J.J.; Thiagarajah, J.R. Extracellular cyclic dinucleotides induce polarized responses in barrier epithelial cells by adenosine signaling. Proc. Natl. Acad. Sci. USA 2020, 117, 27502–27508. [Google Scholar] [CrossRef] [PubMed]
  46. Shu, C.; Li, X.; Li, P. The mechanism of double-stranded DNA sensing through the cGAS-STING pathway. Cytokine Growth Factor Rev. 2014, 25, 641–648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Škrnjug, I.; Rueckert, C.; Libanova, R.; Lienenklaus, S.; Weiss, S.; Guzmán, C. The Mucosal Adjuvant Cyclic di-AMP Exerts Immune Stimulatory Effects on Dendritic Cells and Macrophages. PLoS ONE 2014, 9, e95728. [Google Scholar] [CrossRef]
  48. Karaolis, D.K.R.; Means, T.K.; Yang, D.; Takahashi, M.; Yoshimura, T.; Muraille, E.; Philpott, D.; Schroeder, J.T.; Hyodo, M.; Hayakawa, Y.; et al. Bacterial c-di-GMP Is an Immunostimulatory Molecule. J. Immunol. 2007, 178, 2171–2181. [Google Scholar] [CrossRef] [Green Version]
  49. Ebensen, T.; Libanova, R.; Schulze, K.; Yevsa, T.; Morr, M.; Guzman, C.A. Bis-(3′,5′)-cyclic dimeric adenosine monophosphate: Strong Th1/Th2/Th17 promoting mucosal adjuvant. Vaccine 2011, 29, 5210–5220. [Google Scholar] [CrossRef]
  50. Jin, L.; Hill, K.K.; Filak, H.; Mogan, J.; Knowles, H.; Zhang, B.; Perraud, A.-L.; Cambier, J.C.; Lenz, L.L. MPYS Is Required for IFN Response Factor 3 Activation and Type I IFN Production in the Response of Cultured Phagocytes to Bacterial Second Messengers Cyclic-di-AMP and Cyclic-di-GMP. J. Immunol. 2011, 187, 2595–2601. [Google Scholar] [CrossRef] [Green Version]
  51. Martin, G.R.; Blomquist, C.M.; Henare, K.L.; Jirik, F.R. Stimulator of interferon genes (STING) activation exacerbates experimental colitis in mice. Sci. Rep. 2019, 9, 1–14. [Google Scholar] [CrossRef] [Green Version]
  52. Darling, R.; Senapati, S.; Kelly, S.M.; Kohut, M.L.; Narasimhan, B.; Wannemuehler, M.J. STING pathway stimulation results in a differentially activated innate immune phenotype associated with low nitric oxide and enhanced antibody titers in young and aged mice. Vaccine 2019, 37, 2721–2730. [Google Scholar] [CrossRef]
  53. Mansouri, S.; Katikaneni, D.S.; Gogoi, H.; Jin, L. Monocyte-Derived Dendritic Cells (moDCs) Differentiate into Bcl6+ Mature moDCs to Promote Cyclic di-GMP Vaccine Adjuvant–Induced Memory TH Cells in the Lung. J. Immunol. 2021, 206, 2233–2245. [Google Scholar] [CrossRef] [PubMed]
  54. Abdul-Sater, A.A.; Tattoli, I.; Jin, L.; Grajkowski, A.; Levi, A.; Koller, B.H.; Allen, I.C.; Beaucage, S.L.; Fitzgerald, K.A.; Ting, J.P.Y.; et al. Cyclic-di-GMP and cyclic-di-AMP activate the NLRP3 inflammasome. EMBO Rep. 2013, 14, 900–906. [Google Scholar] [CrossRef] [Green Version]
  55. Walker, M.M.; Crute, B.W.; Cambier, J.C.; Getahun, A. B Cell–Intrinsic STING Signaling Triggers Cell Activation, Synergizes with B Cell Receptor Signals, and Promotes Antibody Responses. J. Immunol. 2018, 201, 2641–2653. [Google Scholar] [CrossRef] [Green Version]
  56. Imanishi, T.; Unno, M.; Kobayashi, W.; Yoneda, N.; Matsuda, S.; Ikeda, K.; Hoshii, T.; Hirao, A.; Miyake, K.; Barber, G.N.; et al. Reciprocal regulation of STING and TCR signaling by mTORC1 for T-cell activation and function. Life Sci. Alliance 2019, 2, e201800282. [Google Scholar] [CrossRef] [PubMed]
  57. Gutjahr, A.; Papagno, L.; Nicoli, F.; Kanuma, T.; Kuse, N.; Cabral-Piccin, M.P.; Rochereau, N.; Gostick, E.; Lioux, T.; Perouzel, E.; et al. The STING ligand cGAMP potentiates the efficacy of vaccine-induced CD8+ T cells. JCI Insight 2019, 4. [Google Scholar] [CrossRef] [PubMed]
  58. Kuse, N.; Sun, X.; Akahoshi, T.; Lissina, A.; Yamamoto, T.; Appay, V.; Takiguchi, M. Priming of HIV-1-specific CD8+ T cells with strong functional properties from naïve T cells. EBioMedicine 2019, 42, 109–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Yamamoto, T.; Kanuma, T.; Takahama, S.; Okamura, T.; Moriishi, E.; Ishii, K.J.; Terahara, K.; Yasutomi, Y. STING agonists activate latently infected cells and enhance SIV-specific responses ex vivo in naturally SIV controlled cynomolgus macaques. Sci. Rep. 2019, 9, 1–11. [Google Scholar] [CrossRef]
  60. Elmanfi, S.; Zhou, J.; Sintim, H.O.; Könönen, E.; Gürsoy, M.; Gürsoy, U.K. Regulation of gingival epithelial cytokine response by bacterial cyclic dinucleotides. J. Oral Microbiol. 2018, 11, 1538927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Yan, H.; KuoLee, R.; Tram, K.; Qiu, H.; Zhang, J.; Patel, G.B.; Chen, W. 3′,5′-Cyclic diguanylic acid elicits mucosal immunity against bacterial infection. Biochem. Biophys. Res. Commun. 2009, 387, 581–584. [Google Scholar] [CrossRef] [PubMed]
  62. Ogunniyi, A.D.; Paton, J.C.; Kirby, A.C.; McCullers, J.A.; Cook, J.; Hyodo, M.; Hayakawa, Y.; Karaolis, D.K. c-di-GMP is an effective immunomodulator and vaccine adjuvant against pneumococcal infection. Vaccine 2008, 26, 4676–4685. [Google Scholar] [CrossRef] [Green Version]
  63. Zhao, L.; KuoLee, R.; Harris, G.; Tram, K.; Yan, H.; Chen, W. c-di-GMP protects against intranasal Acinetobacter baumannii infection in mice by chemokine induction and enhanced neutrophil recruitment. Int. Immunopharmacol. 2011, 11, 1378–1383. [Google Scholar] [CrossRef]
  64. Skouboe, M.K.; Knudsen, A.; Reinert, L.; Boularan, C.; Lioux, T.; Pérouzel, E.; Thomsen, M.K.; Paludan, S.R. STING agonists enable antiviral cross-talk between human cells and confer protection against genital herpes in mice. PLoS Pathog. 2018, 14, e1006976. [Google Scholar] [CrossRef] [PubMed]
  65. Blaauboer, S.M.; Mansouri, S.; Tucker, H.R.; Wang, H.L.; Gabrielle, V.D.; Jin, L. The mucosal adjuvant cyclic di-GMP enhances antigen uptake and selectively activates pinocytosis-efficient cells in vivo. eLife 2015, 4, e06670. [Google Scholar] [CrossRef]
  66. Elahi, S.; van Kessel, J.; Kiros, T.G.; Strom, S.; Hayakawa, Y.; Hyodo, M.; Babiuk, L.A.; Gerdts, V. c-di-GMP Enhances Protective Innate Immunity in a Murine Model of Pertussis. PLoS ONE 2014, 9, e109778. [Google Scholar] [CrossRef] [Green Version]
  67. Fatima, M.; Rempel, H.; Kuang, X.T.; Allen, K.J.; Cheng, K.M.; Malouin, F.; Diarra, M.S. Effect of 3’,5’-cyclic diguanylic acid in a broiler Clostridium perfringens infection model. Poult. Sci. 2013, 92, 2644–2650. [Google Scholar] [CrossRef]
  68. Karaolis, D.K.R.; Newstead, M.W.; Zeng, X.; Hyodo, M.; Hayakawa, Y.; Bhan, U.; Liang, H.; Standiford, T.J. Cyclic Di-GMP Stimulates Protective Innate Immunity in Bacterial Pneumonia. Infect. Immun. 2007, 75, 4942–4950. [Google Scholar] [CrossRef] [Green Version]
  69. Johnson, B.M.; Uchimura, T.; Gallovic, M.D.; Thamilarasan, M.; Chou, W.-C.; Gibson, S.A.; Deng, M.; Tam, J.W.; Batty, C.J.; Williams, J.; et al. STING Agonist Mitigates Experimental Autoimmune Encephalomyelitis by Stimulating Type I IFN–Dependent and –Independent Immune-Regulatory Pathways. J. Immunol. 2021, 206, 2015–2028. [Google Scholar] [CrossRef] [PubMed]
  70. Ohkuri, T.; Ghosh, A.; Kosaka, A.; Zhu, J.; Ikeura, M.; David, M.; Watkins, S.C.; Sarkar, S.N.; Okada, H. STING contributes to antiglioma immunity via triggering type I IFN signals in the tumor microenvironment. Cancer Immunol. Res. 2014, 2, 1199–1208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Corrales, L.; Glickman, L.H.; McWhirter, S.M.; Kanne, D.B.; Sivick, K.E.; Katibah, G.E.; Woo, S.-R.; Lemmens, E.; Banda, T.; Leong, J.J.; et al. Direct Activation of STING in the Tumor Microenvironment Leads to Potent and Systemic Tumor Regression and Immunity. Cell Rep. 2015, 11, 1018–1030. [Google Scholar] [CrossRef] [Green Version]
  72. Gadkaree, S.K.; Fu, J.; Sen, R.; Korrer, M.J.; Allen, C.; Kim, Y.J. Induction of tumor regression by intratumoral STING agonists combined with anti-programmed death-L1 blocking antibody in a preclinical squamous cell carcinoma model. Head Neck 2017, 39, 1086–1094. [Google Scholar] [CrossRef]
  73. Moore, E.; Clavijo, P.E.; Davis, R.; Cash, H.; van Waes, C.; Kim, Y.; Allen, C. Established T Cell–Inflamed Tumors Rejected after Adaptive Resistance Was Reversed by Combination STING Activation and PD-1 Pathway Blockade. Cancer Immunol. Res. 2016, 4, 1061–1071. [Google Scholar] [CrossRef] [Green Version]
  74. Wehbe, M.; Wang-Bishop, L.; Becker, K.W.; Shae, D.; Baljon, J.J.; He, X.; Christov, P.; Boyd, K.L.; Balko, J.M.; Wilson, J.T. Nanoparticle delivery improves the pharmacokinetic properties of cyclic dinucleotide STING agonists to open a therapeutic window for intravenous administration. J. Control. Release 2021, 330, 1118–1129. [Google Scholar] [CrossRef]
  75. Wang-Bishop, L.; Wehbe, M.; Shae, D.; James, J.; Hacker, B.C.; Garland, K.; Chistov, P.P.; Rafat, M.; Balko, J.M.; Wilson, J.T. Potent STING activation stimulates immunogenic cell death to enhance antitumor immunity in neuroblastoma. J. Immunother. Cancer 2019, 8, e000282. [Google Scholar] [CrossRef] [Green Version]
  76. Foote, J.B.; Kok, M.; Leatherman, J.M.; Armstrong, T.D.; Marcinkowski, B.; Ojalvo, L.S.; Kanne, D.B.; Jaffee, E.; Dubensky, T.W.; Emens, L.A. A STING Agonist Given with OX40 Receptor and PD-L1 Modulators Primes Immunity and Reduces Tumor Growth in Tolerized Mice. Cancer Immunol. Res. 2017, 5, 468–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Bielecki, P.A.; Lorkowski, M.E.; Becicka, W.M.; Atukorale, P.U.; Moon, T.J.; Zhang, Y.; Wiese, M.; Covarrubias, G.; Ravichandran, S.; Karathanasis, E. Immunostimulatory silica nanoparticle boosts innate immunity in brain tumors. Nanoscale Horiz. 2021, 6, 156–167. [Google Scholar] [CrossRef]
  78. Chen, Y.P.; Xu, L.; Tang, T.W.; Chen, C.H.; Zheng, Q.H.; Liu, T.P.; Mou, C.Y.; Wu, C.H.; Wu, S.H. STING Activator c-di-GMP-Loaded Mesoporous Silica Nano-particles Enhance Immunotherapy Against Breast Cancer. ACS Appl. Mater. Interfaces 2020, 12, 56741–56752. [Google Scholar] [CrossRef] [PubMed]
  79. An, M.; Yu, C.; Xi, J.; Reyes, J.; Mao, G.; Wei, W.Z.; Liu, H. Induction of necrotic cell death and activation of STING in the tumor mi-croenvironment via cationic silica nanoparticles leading to enhanced antitumor immunity. Nanoscale 2018, 10, 9311–9319. [Google Scholar] [CrossRef] [PubMed]
  80. Wilson, D.R.; Sen, R.; Sunshine, J.; Pardoll, D.M.; Green, J.J.; Kim, Y.J. Biodegradable STING agonist nanoparticles for enhanced cancer immunotherapy. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 237–246. [Google Scholar] [CrossRef] [PubMed]
  81. Ager, C.R.; Reilley, M.J.; Nicholas, C.; Bartkowiak, T.; Jaiswal, A.; Curran, M.A. Intratumoral STING Activation with T-cell Checkpoint Modulation Generates Systemic Antitumor Immunity. Cancer Immunol. Res. 2017, 5, 676–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Cheng, N.; Watkins-Schulz, R.; Junkins, R.D.; David, C.N.; Johnson, B.M.; Montgomery, S.A.; Peine, K.J.; Darr, D.B.; Yuan, H.; McKinnon, K.P.; et al. A nanoparticle-incorporated STING activator enhances antitumor immunity in PD-L1–insensitive models of triple-negative breast cancer. JCI Insight 2018, 3. [Google Scholar] [CrossRef] [PubMed]
  83. Smith, T.T.; Moffett, H.; Stephan, S.B.; Opel, C.F.; Dumigan, A.; Jiang, X.; Pillarisetty, V.G.; Pillai, S.P.S.; Wittrup, K.D.; Stephan, M. Biopolymers codelivering engineered T cells and STING agonists can eliminate heterogeneous tumors. J. Clin. Investig. 2017, 127, 2176–2191. [Google Scholar] [CrossRef] [PubMed]
  84. Zhu, Y.; An, X.; Zhang, X.; Qiao, Y.; Zheng, T.; Li, X. STING: A master regulator in the cancer-immunity cycle. Mol. Cancer 2019, 18, 1–15. [Google Scholar] [CrossRef] [Green Version]
  85. Atukorale, P.U.; Raghunathan, S.P.; Raguveer, V.; Moon, T.J.; Zheng, C.; Bielecki, P.A.; Wiese, M.L.; Goldberg, A.L.; Covarrubias, G.; Hoimes, C.J.; et al. Nanoparticle Encapsulation of Synergistic Immune Agonists Enables Systemic Codelivery to Tumor Sites and IFNbeta-Driven Antitumor Immunity. Cancer Res. 2019, 79, 5394–5406. [Google Scholar] [CrossRef] [Green Version]
  86. Temizoz, B.; Kuroda, E.; Ohata, K.; Jounai, N.; Ozasa, K.; Kobiyama, K.; Aoshi, T.; Ishii, K.J. TLR9 and STING agonists synergistically induce innate and adaptive type-II IFN. Eur. J. Immunol. 2015, 45, 1159–1169. [Google Scholar] [CrossRef]
  87. Francica, B.J.; Ghasemzadeh, A.; Desbien, A.L.; Theodros, D.; Sivick, K.E.; Reiner, G.L.; Glickman, L.H.; Marciscano, A.E.; Sharabi, A.B.; Leong, M.L.; et al. TNFalpha and Radioresistant Stromal Cells Are Essential for Therapeutic Efficacy of Cyclic Dinucleotide STING Agonists in Nonimmunogenic Tumors. Cancer Immunol. Res. 2018, 6, 422–433. [Google Scholar] [CrossRef] [Green Version]
  88. Miao, L.; Qi, J.; Zhao, Q.; Wu, Q.-N.; Wei, D.-L.; Wei, X.-L.; Liu, J.; Chen, J.; Zeng, Z.-L.; Ju, H.-Q.; et al. Targeting the STING pathway in tumor-associated macrophages regulates innate immune sensing of gastric cancer cells. Theranostics 2020, 10, 498–515. [Google Scholar] [CrossRef]
  89. Ohkuri, T.; Kosaka, A.; Ishibashi, K.; Kumai, T.; Hirata, Y.; Ohara, K.; Nagato, T.; Oikawa, K.; Aoki, N.; Harabuchi, Y.; et al. Intratumoral administration of cGAMP transiently ac-cumulates potent macrophages for anti-tumor immunity at a mouse tumor site. Cancer Immunol. Immunother. 2017, 66, 705–716. [Google Scholar] [CrossRef]
  90. Nicolai, C.J.; Wolf, N.; Chang, I.-C.; Kirn, G.; Marcus, A.; Ndubaku, C.O.; McWhirter, S.M.; Raulet, D.H. NK cells mediate clearance of CD8+ T cell–resistant tumors in response to STING agonists. Sci. Immunol. 2020, 5, eaaz2738. [Google Scholar] [CrossRef] [PubMed]
  91. Ebensen, T.; Schulze, K.; Riese, P.; Link, C.; Morr, M.; Guzmán, C.A. The bacterial second messenger cyclic diGMP exhibits potent adjuvant properties. Vaccine 2007, 25, 1464–1469. [Google Scholar] [CrossRef]
  92. Ebensen, T.; Schulze, K.; Riese, P.; Morr, M.; Guzmán, C.A. The Bacterial Second Messenger cdiGMP Exhibits Promising Activity as a Mucosal Adjuvant. Clin. Vaccine Immunol. 2007, 14, 952–958. [Google Scholar] [CrossRef] [Green Version]
  93. Libanova, R.; Ebensen, T.; Schulze, K.; Bruhn, D.; Nörder, M.; Yevsa, T.; Morr, M.; Guzmán, C.A. The member of the cyclic di-nucleotide family bis-(3′, 5′)-cyclic dimeric inosine monophosphate exerts potent activity as mucosal adjuvant. Vaccine 2010, 28, 2249–2258. [Google Scholar] [CrossRef] [PubMed]
  94. Škrnjug, I.; Guzmán, C.; Ruecker, C. Cyclic GMP-AMP Displays Mucosal Adjuvant Activity in Mice. PLoS ONE 2014, 9, e110150. [Google Scholar] [CrossRef] [PubMed]
  95. Gray, P.M.; Forrest, G.; Wisniewski, T.; Porter, G.; Freed, D.C.; DeMartino, J.A.; Zaller, D.M.; Guo, Z.; Leone, J.; Fu, T.-M.; et al. Evidence for cyclic diguanylate as a vaccine adjuvant with novel immunostimulatory activities. Cell. Immunol. 2012, 278, 113–119. [Google Scholar] [CrossRef] [PubMed]
  96. Volckmar, J.; Knop, L.; Stegemann-Koniszewski, S.; Schulze, K.; Ebensen, T.; Guzman, C.A.; Bruder, D. The STING activator c-di-AMP exerts superior adjuvant properties than the formulation poly(I:C)/CpG after subcutaneous vaccination with soluble protein antigen or DEC-205-mediated antigen targeting to dendritic cells. Vaccine 2019, 37, 4963–4974. [Google Scholar] [CrossRef] [PubMed]
  97. Yildiz, S.; Alpdundar, E.; Gungor, B.; Kahraman, T.; Bayyurt, B.; Gursel, I.; Gursel, M. Enhanced immunostimulatory activity of cyclic dinucleotides on mouse cells when complexed with a cell-penetrating peptide or combined with CpG. Eur. J. Immunol. 2015, 45, 1170–1179. [Google Scholar] [CrossRef]
  98. Guo, J.; Wang, J.; Fan, J.; Zhang, Y.; Dong, W.; Chen, C.P. Distinct Dynamic and Conformational Features of Human STING in Re-sponse to 2′3′-cGAMP and c-di-GMP. Chembiochem Eur. J. Chem. Biol. 2019, 20, 1838–1847. [Google Scholar]
  99. Yan, H.; Wang, X.; KuoLee, R.; Chen, W. Synthesis and immunostimulatory properties of the phosphorothioate analogues of cdiGMP. Bioorganic Med. Chem. Lett. 2008, 18, 5631–5634. [Google Scholar] [CrossRef]
  100. Li, J.; Lee, R.K.; Chen, W.; Yan, H. 2′-Fluoro-c-di-GMP as an oral vaccine adjuvant. RSC Adv. 2019, 9, 41481–41489. [Google Scholar] [CrossRef] [Green Version]
  101. Flores-Valdez, M.A.; Aceves-Sánchez, M.D.J.; Montero-Pérez, S.A.; Sánchez-López, A.D.; Gutiérrez-Pabello, J.A.; Hernández-Pando, R. Vaccination of mice with recombinant bacille Calmette-Guérin harboring Rv1357c protects similarly to native BCG. Int. J. Tuberc. Lung Dis. 2012, 16, 774–776. [Google Scholar] [CrossRef]
  102. Pedroza-Roldan, C.; Guapillo, C.; Barrios-Payán, J.; Mata-Espinosa, D.; Aceves-Sánchez, M.D.J.; Marquina-Castillo, B.; Hernández-Pando, R.; Flores-Valdez, M.A. The BCGΔBCG1419c strain, which produces more pellicle in vitro, improves control of chronic tuberculosis in vivo. Vaccine 2016, 34, 4763–4770. [Google Scholar] [CrossRef] [PubMed]
  103. Ning, H.; Wang, L.; Zhou, J.; Lu, Y.; Kang, J.; Ding, T.; Shen, L.; Xu, Z.; Bai, Y. Recombinant BCG With Bacterial Signaling Molecule Cyclic di-AMP as Endogenous Adjuvant Induces Elevated Immune Responses After Mycobacterium tuberculosis Infection. Front. Immunol. 2019, 10, 1519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Dey, R.J.; Dey, B.; Singh, A.K.; Praharaj, M.; Bishai, W. Bacillus Calmette-Guérin Overexpressing an Endogenous Stimulator of Interferon Genes Agonist Provides Enhanced Protection Against Pulmonary Tuberculosis. J. Infect. Dis. 2020, 221, 1048–1056. [Google Scholar] [CrossRef] [Green Version]
  105. Madhun, A.S.; Haaheim, L.R.; Nostbakken, J.K.; Ebensen, T.; Chichester, J.; Yusibov, V.; Guzmán, C.A.; Cox, R.J. Intranasal c-di-GMP-adjuvanted plant-derived H5 influenza vaccine induces multifunctional Th1 CD4+ cells and strong mucosal and systemic antibody re-sponses in mice. Vaccine 2011, 29, 4973–4982. [Google Scholar] [CrossRef] [PubMed]
  106. Pedersen, G.K.; Ebensen, T.; Gjeraker, I.H.; Svindland, S.; Bredholt, G.; Guzmán, C.A.; Cox, R.J. Evaluation of the Sublingual Route for Administration of Influenza H5N1 Virosomes in Combination with the Bacterial Second Messenger c-di-GMP. PLoS ONE 2011, 6, e26973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Svindland, S.C.; Pedersen, G.; Pathirana, R.D.; Bredholt, G.; Nøstbakken, J.K.; Jul-Larsen, Å.; Guzmán, C.; Montomoli, E.; Lapini, G.; Piccirella, S.; et al. A study of Chitosan and c-di-GMP as mucosal adjuvants for intranasal influenza H5N1 vaccine. Influenza Other Respir. Viruses 2012, 7, 1181–1193. [Google Scholar] [CrossRef] [Green Version]
  108. Neuhaus, V.; Chichester, J.A.; Ebensen, T.; Schwarz, K.; Hartman, C.E.; Shoji, Y.; Guzmán, C.A.; Yusibov, V.; Sewald, K.; Braun, A. A new adjuvanted nanoparticle-based H1N1 influenza vaccine induced antigen-specific local mucosal and systemic immune responses after administration into the lung. Vaccine 2014, 32, 3216–3222. [Google Scholar] [CrossRef]
  109. Major, D.; Chichester, J.A.; Pathirana, R.D.; Guilfoyle, K.; Shoji, Y.; Guzmán, C.; Yusibov, V.; Cox, R.J. Intranasal vaccination with a plant-derived H5 HA vaccine protects mice and ferrets against highly pathogenic avian influenza virus challenge. Hum. Vaccines Immunother. 2015, 11, 1235–1243. [Google Scholar] [CrossRef] [Green Version]
  110. Shin, J.-H.; Lee, J.-H.; Jeong, S.D.; Noh, J.-Y.; Lee, H.W.; Song, C.-S.; Kim, Y.-C. C-di-GMP with influenza vaccine showed enhanced and shifted immune responses in microneedle vaccination in the skin. Drug Deliv. Transl. Res. 2020, 10, 815–825. [Google Scholar] [CrossRef]
  111. Allen, A.C.; Wilk, M.; Misiak, A.; Borkner, L.; Murphy, D.; Mills, K.H.G. Sustained protective immunity against Bordetella pertussis nasal colonization by intranasal immunization with a vaccine-adjuvant combination that induces IL-17-secreting TRM cells. Mucosal Immunol. 2018, 11, 1763–1776. [Google Scholar] [CrossRef] [Green Version]
  112. Hu, D.-L.; Narita, K.; Hyodo, M.; Hayakawa, Y.; Nakane, A.; Karaolis, D.K. c-di-GMP as a vaccine adjuvant enhances protection against systemic methicillin-resistant Staphylococcus aureus (MRSA) infection. Vaccine 2009, 27, 4867–4873. [Google Scholar] [CrossRef] [PubMed]
  113. Latanova, A.A.; Petkov, S.; Kilpelainen, A.; Jansons, J.; Latyshev, O.E.; Kuzmenko, Y.V.; Hinkula, J.; Abakumov, M.A.; Valuev-Eliston, V.T.; Gomelsy, M.; et al. Codon optimization and improved delivery/immunization regimen enhance the immune response against wild-type and drug-resistant HIV-1 reverse tran-scriptase, preserving its Th2-polarity. Sci. Rep. 2018, 8, 8078. [Google Scholar] [CrossRef]
  114. Lee, M.J.; Jo, H.; Shin, S.H.; Kim, S.M.; Kim, B.; Shim, H.S.; Park, J.H. Mincle and STING-Stimulating Adjuvants Elicit Robust Cellular Immunity and Drive Long-Lasting Memory Responses in a Foot-and-Mouth Disease Vaccine. Front. Immunol. 2019, 10, 2509. [Google Scholar] [CrossRef]
  115. Azegami, T.; Yuki, Y.; Sawada, S.; Mejima, M.; Ishige, K.; Akiyoshi, K.; Itoh, H.; Kiyono, H. Nanogel-based nasal ghrelin vaccine prevents obesity. Mucosal Immunol. 2017, 10, 1351–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Azegami, T.; Yuki, Y.; Hayashi, K.; Hishikawa, A.; Sawada, S.-I.; Ishige, K.; Akiyoshi, K.; Kiyono, H.; Itoh, H. Intranasal vaccination against angiotensin II type 1 receptor and pneumococcal surface protein A attenuates hypertension and pneumococcal infection in rodents. J. Hypertens. 2018, 36, 387–394. [Google Scholar] [CrossRef] [PubMed]
  117. Ebensen, T.; Debarry, J.; Pedersen, G.K.; Blazejewska, P.; Weissmann, S.; Schulze, K.; McCullough, K.C.; Cox, R.J.; Guzman, C.A. Mucosal Administration of Cy-cle-Di-Nucleotide-Adjuvanted Virosomes Efficiently Induces Protection against Influenza H5N1 in Mice. Front. Immunol. 2017, 8, 1223. [Google Scholar] [CrossRef] [PubMed]
  118. Sanchez, M.; Ebensen, T.; Schulze, K.; Cargnelutti, D.E.; Blazejewska, P.; Scodeller, E.A.; Guzmán, C.A. Intranasal Delivery of Influenza rNP Adjuvanted with c-di-AMP Induces Strong Humoral and Cellular Immune Responses and Provides Protection against Virus Challenge. PLoS ONE 2014, 9, e104824. [Google Scholar] [CrossRef] [PubMed]
  119. Ning, H.; Zhang, W.; Kang, J.; Ding, T.; Liang, X.; Lu, Y.; Guo, C.; Sun, W.; Wang, H.; Bai, Y.; et al. Subunit Vaccine ESAT-6:c-di-AMP Delivered by Intranasal Route Elicits Immune Responses and Protects Against Mycobacterium tuberculosis Infection. Front. Cell. Infect. Microbiol. 2021, 11. [Google Scholar] [CrossRef] [PubMed]
  120. Schulze, K.; Ebensen, T.; Chandrudu, S.; Skwarczynski, M.; Toth, I.; Olive, C.; Guzman, C.A. Bivalent mucosal peptide vaccines administered using the LCP carrier system stimulate protective immune responses against Streptococcus pyogenes infection. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 2463–2474. [Google Scholar] [CrossRef]
  121. Landi, A.; Law, J.; Hockman, D.; Logan, M.; Crawford, K.; Chen, C.; Kundu, J.; Ebensen, T.; Guzman, C.; Deschatelets, L.; et al. Superior immunogenicity of HCV envelope glycoproteins when adjuvanted with cyclic-di-AMP, a STING activator or archaeosomes. Vaccine 2017, 35, 6949–6956. [Google Scholar] [CrossRef] [PubMed]
  122. Munoz Gonzalez, F.; Sycz, G.; Alonso Paiva, I.M.; Linke, D.; Zorreguieta, A.; Baldi, P.C.; Ferero, M.C. The BtaF Adhesin Is Necessary for Full Virulence during Respiratory Infection by Brucella suis and Is a Novel Immunogen for Nasal Vaccination Against Brucella Infection. Front. Immunol. 2019, 10, 1775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Matos, M.N.; Cazorla, S.I.; Schulze, K.; Ebensen, T.; Guzmán, C.A.; Malchiodi, E. Immunization with Tc52 or its amino terminal domain adjuvanted with c-di-AMP induces Th17+Th1 specific immune responses and confers protection against Trypanosoma cruzi. PLoS Negl. Trop. Dis. 2017, 11, e0005300. [Google Scholar] [CrossRef] [PubMed]
  124. Sanchez Alberti, A.; Bivona, A.E.; Cerny, N.; Schulze, K.; Weissmann, S.; Ebensen, T.; Morales, C.; Padilla, A.M.; Cazorla, S.I.; Tarleton, R.L.; et al. Engineered trivalent immunogen adju-vanted with a STING agonist confers protection against Trypanosoma cruzi infection. NPJ Vaccines 2017, 2, 9. [Google Scholar] [CrossRef]
  125. Sanchez Alberti, A.; Bivona, A.E.; Matos, M.N.; Cerny, N.; Schulze, K.; Weissmann, S.; Ebensen, T.; Gonzalez, G.; Morales, C.; Cardoso, A.C.; et al. Mucosal Heterologous Prime/Boost Vaccination Induces Polyfunctional Systemic Immunity, Improving Protection Against Trypanosoma cruzi. Front. Immunol. 2020, 11, 128. [Google Scholar] [CrossRef] [Green Version]
  126. Matthijs, A.M.F.; Auray, G.; Jakob, V.; García-Nicolás, O.; Braun, R.O.; Keller, I.; Bruggman, R.; Devriendt, B.; Boyen, F.; Guzman, C.A.; et al. Systems Immunology Characterization of Novel Vaccine Formulations for Mycoplasma hyopneumoniae Bacterins. Front. Immunol. 2019, 10, 1087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Luo, J.; Liu, X.-P.; Xiong, F.-F.; Gao, F.-X.; Yi, Y.-L.; Zhang, M.; Chen, Z.; Tan, W.-S. Enhancing Immune Response and Heterosubtypic Protection Ability of Inactivated H7N9 Vaccine by Using STING Agonist as a Mucosal Adjuvant. Front. Immunol. 2019, 10, 2274. [Google Scholar] [CrossRef] [Green Version]
  128. Takaki, H.; Takashima, K.; Oshiumi, H.; Ainai, A.; Suzuki, T.; Hasegawa, H.; Matsumoto, M.; Seya, T. cGAMP Promotes Germinal Center Formation and Production of IgA in Nasal-Associated Lymphoid Tissue. Med. Sci. 2017, 5, 35. [Google Scholar] [CrossRef] [Green Version]
  129. Wang, J.; Li, P.; Wu, M.X. Natural STING Agonist as an “Ideal” Adjuvant for Cutaneous Vaccination. J. Investig. Dermatol. 2016, 136, 2183–2191. [Google Scholar] [CrossRef] [Green Version]
  130. Wang, J.; Li, P.; Yu, Y.; Fu, Y.; Jiang, H.; Lu, M.; Sun, Z.; Jiang, S.; Lu, L.; Wu, M.X. Pulmonary surfactant–biomimetic nanoparticles potentiate heterosubtypic influenza immunity. Science 2020, 367, eaau0810. [Google Scholar] [CrossRef]
  131. Vassilieva, E.V.; Li, S.; Korniychuk, H.; Taylor, D.M.; Wang, S.; Prausnitz, M.R.; Compans, R.W. cGAMP/Saponin Adjuvant Combination Improves Protective Response to Influenza Vaccination by Microneedle Patch in an Aged Mouse Model. Front. Immunol. 2021, 11, 583251. [Google Scholar] [CrossRef]
  132. Chen, J.; Zhong, Y.; Liu, Y.; Tang, C.; Zhang, Y.; Wei, B.; Chen, W.; Liu, M. Parenteral immunization with a cyclic guanosine monophos-phate-adenosine monophosphate (cGAMP) adjuvanted Helicobacter pylori vaccine induces protective immunity against H. pylori infection in mice. Hum. Vaccines Immunother. 2020, 16, 2849–2854. [Google Scholar] [CrossRef]
  133. Liu, Y.; Chen, S.; Pan, B.; Guan, Z.; Yang, Z.; Duan, L.; Cai, H. A subunit vaccine based on rH-NS induces protection against Mycobac-terium tuberculosis infection by inducing the Th1 immune response and activating macrophages. Acta Biochim. Biophys. Sin. 2016, 48, 909–922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Dorostkar, F.; Arashkia, A.; Roohvand, F.; Shoja, Z.; Navari, M.; Mashhadi Abolghasem Shirazi, M. Co-administration of 2’3’-cGAMP STING activator and CpG-C adjuvants with a mutated form of HPV 16 E7 protein leads to tumor growth inhibition in the mouse model. Infect. Agents Cancer 2021, 16, 7. [Google Scholar] [CrossRef] [PubMed]
  135. Ito, H.; Kanbe, A.; Hara, A.; Ishikawa, T. Induction of humoral and cellular immune response to HBV vaccine can be up-regulated by STING ligand. Virology 2019, 531, 233–239. [Google Scholar] [CrossRef] [PubMed]
  136. Borriello, F.; Pietrasanta, C.; Lai, J.C.Y.; Walsh, L.M.; Sharma, P.; O’Driscoll, D.N.; Ramirez, J.; Brightman, S.; Pugni, L.; Mosca, F.; et al. Identification and Characterization of Stimulator of Interferon Genes as a Robust Adjuvant Target for Early Life Immunization. Front. Immunol. 2017, 8, 1772. [Google Scholar] [CrossRef] [Green Version]
  137. Chen, N.; Gallovic, M.D.; Tiet, P.; Ting, J.P.-Y.; Ainslie, K.; Bachelder, E.M. Investigation of tunable acetalated dextran microparticle platform to optimize M2e-based influenza vaccine efficacy. J. Control. Release 2018, 289, 114–124. [Google Scholar] [CrossRef] [PubMed]
  138. Martin, T.; Jee, J.; Kim, E.; Steiner, H.E.; Cormet-Boyaka, E.; Boyaka, P.N. Sublingual targeting of STING with 3′3′-cGAMP promotes systemic and mucosal immunity against anthrax toxins. Vaccine 2017, 35, 2511–2519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Van Dis, E.; Sogi, K.M.; Rae, C.S.; Sivick, K.E.; Surh, N.H.; Leong, M.L. Faculty Opinions recommendation of STING-Activating Adjuvants Elicit a Th17 Immune Response and Protect against Mycobacterium tuberculosis Infection. Fac. Opin. 2018, 23, 1435–1447. [Google Scholar] [CrossRef] [Green Version]
  140. Wagner, D.A.; Kelly, S.M.; Petersen, A.C.; Peroutka-Bigus, N.; Darling, R.; Bellaire, B.H.; Wannemuehler, M.J.; Narasimhan, B. Single-dose combination nanovaccine induces both rapid and long-lived protection against pneumonic plague. Acta Biomater. 2019, 100, 326–337. [Google Scholar] [CrossRef]
  141. Latasa, C.; Echeverz, M.; Garcia, B.; Gil, C.; García-Ona, E.; Burgui, S.; Casares, N.; Hervas-Stubbs, S.; Lasarte, J.J.; Lasa, I.; et al. Evaluation of a Salmonella Strain Lacking the Secondary Messenger C-di-GMP and RpoS as a Live Oral Vaccine. PLoS ONE 2016, 11, e0161216. [Google Scholar] [CrossRef] [Green Version]
  142. Gil, C.; Latasa, C.; García-Ona, E.; Lázaro, I.; Labairu, J.; Echeverz, M.; Burgui, S.; García, B.; Lasa, I.; Solano, C. A DIVA vaccine strain lacking RpoS and the secondary messenger c-di-GMP for protection against salmonellosis in pigs. Veter. Res. 2020, 51, 1–10. [Google Scholar] [CrossRef] [Green Version]
  143. Vassilieva, E.V.; Taylor, D.W.; Compans, R.W. Combination of STING Pathway Agonist with Saponin Is an Effective Adjuvant in Immunosenescent Mice. Front. Immunol. 2019, 10, 3006. [Google Scholar] [CrossRef] [Green Version]
  144. Gogoi, H.; Mansouri, S.; Katikaneni, D.S.; Jin, L. New MoDC-Targeting TNF Fusion Proteins Enhance Cyclic Di-GMP Vaccine Adjuvanticity in Middle-Aged and Aged Mice. Front. Immunol. 2020, 11, 1674. [Google Scholar] [CrossRef]
  145. Gravekamp, C.; Chandra, D. Targeting STING pathways for the treatment of cancer. OncoImmunology 2015, 4, e988463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Chandra, D.; Quispe-Tintaya, W.; Jahangir, A.; Asafu-Adjei, D.; Ramos, I.; Sintim, H.O.; Zhou, J.; Hayakawa, Y.; Karaolis, D.K.; Gravekamp, C. STING Ligand c-di-GMP Improves Cancer Vaccination against Metastatic Breast Cancer. Cancer Immunol. Res. 2014, 2, 901–910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Wu, J.-J.; Li, W.-H.; Chen, P.-G.; Zhang, B.-D.; Hu, H.-G.; Li, Q.-Q.; Zhao, L.; Chen, Y.-X.; Zhao, Y.-F.; Li, Y.-M. Targeting STING with cyclic di-GMP greatly augmented immune responses of glycopeptide cancer vaccines. Chem. Commun. 2018, 54, 9655–9658. [Google Scholar] [CrossRef] [PubMed]
  148. Wang, Z.; Celis, E. STING activator c-di-GMP enhances the anti-tumor effects of peptide vaccines in melanoma-bearing mice. Cancer Immunol. Immunother. 2015, 64, 1057–1066. [Google Scholar] [CrossRef]
  149. Shae, D.; Baljon, J.J.; Wehbe, M.; Christov, P.P.; Becker, K.W.; Kumar, A.; Suryadevara, N.; Carson, C.S.; Palmer, C.R.; Knight, F.C.; et al. Co-delivery of Peptide Neoantigens and Stimulator of Interferon Genes Agonists Enhances Response to Cancer Vaccines. ACS Nano 2020, 14, 9904–9916. [Google Scholar] [CrossRef]
  150. Ebensen, T.; Delandre, S.; Prochnow, B.; Guzmán, C.A.; Schulze, K. The Combination Vaccine Adjuvant System Alum/c-di-AMP Results in Quantitative and Qualitative Enhanced Immune Responses Post Immunization. Front. Cell. Infect. Microbiol. 2019, 9, 31. [Google Scholar] [CrossRef] [Green Version]
  151. Hu, H.G.; Wu, J.J.; Zhang, B.D.; Li, W.H.; Li, Y.M. Pam3CSK4-CDG(SF) Augments Antitumor Immunotherapy by Synergistically Activating TLR1/2 and STING. Bioconjug. Chem. 2020, 31, 2499–2503. [Google Scholar] [CrossRef]
  152. Alyaqoub, F.S.; Aldhamen, Y.A.; Koestler, B.J.; Bruger, E.L.; Seregin, S.S.; Pereira-Hicks, C.; Godbehere, S.; Waters, C.M.; Amalfitano, A. In Vivo Synthesis of Cyclic-di-GMP Using a Recombinant Adenovirus Preferentially Improves Adaptive Immune Responses against Extracellular Antigens. J. Immunol. 2016, 196, 1741–1752. [Google Scholar] [CrossRef]
  153. Koestler, B.; Seregin, S.S.; Rastall, D.P.W.; Aldhamen, Y.A.; Godbehere, S.; Amalfitano, A.; Waters, C.M. Stimulation of Innate Immunity byIn VivoCyclic di-GMP Synthesis Using Adenovirus. Clin. Vaccine Immunol. 2014, 21, 1550–1559. [Google Scholar] [CrossRef] [Green Version]
  154. Bridgeman, A.; Maelfait, J.; Davenne, T.; Partridge, T.; Peng, Y.; Mayer, A.; Dong, T.; Kaever, V.; Borrow, P.; Rehwinkel, J. Viruses transfer the antiviral second messenger cGAMP between cells. Science 2015, 349, 1228–1232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Quintana, I.; Espariz, M.; Villar, S.R.; Gonzalez, F.B.; Pacini, M.F.; Cabrera, G.; Bontempi, I.; Prochetto, E.; Stulke, J.; Perez, A.R.; et al. Genetic Engineering of Lactococcus lactis Co-producing Antigen and the Mucosal Adjuvant 3′ 5′- cyclic di Adenosine Monophosphate (c-di-AMP) as a Design Strategy to Develop a Mucosal Vaccine Prototype. Front. Microbiol. 2018, 9, 2100. [Google Scholar] [CrossRef]
  156. Konno, H.; Chinn, I.K.; Hong, D.; Orange, J.S.; Lupski, J.R.; Mendoza, A.; Pedroza, L.A.; Barber, G.N. Pro-inflammation Associated with a Gain-of-Function Mutation (R284S) in the Innate Immune Sensor STING. Cell Rep. 2018, 23, 1112–1123. [Google Scholar] [CrossRef] [Green Version]
  157. Ozasa, K.; Temizoz, B.; Kusakabe, T.; Kobari, S.; Momota, M.; Coban, C.; Ito, S.; Kobiyama, K.; Kuroda, E.; Ishii, K.J. Cyclic GMP-AMP Triggers Asthma in an IL-33-Dependent Manner That Is Blocked by Amlexanox, a TBK1 Inhibitor. Front. Immunol. 2019, 10. [Google Scholar] [CrossRef] [PubMed]
  158. Konno, H.; Konno, K.; Barber, G.N. Cyclic Dinucleotides Trigger ULK1 (ATG1) Phosphorylation of STING to Prevent Sustained Innate Immune Signaling. Cell 2013, 155, 688–698. [Google Scholar] [CrossRef] [Green Version]
  159. Rueckert, C.; Rand, U.; Roy, U.; Kasmapour, B.; Strowig, T.; Guzmán, C.A. Cyclic dinucleotides modulate induced type I IFN responses in innate immune cells by degradation of STING. FASEB J. 2017, 31, 3107–3115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Miyabe, H.; Hyodo, M.; Nakamura, T.; Sato, Y.; Hayakawa, Y.; Harashima, H. A new adjuvant delivery system ‘cyclic di-GMP/YSK05 liposome’ for cancer immunotherapy. J. Control. Release 2014, 184, 20–27. [Google Scholar] [CrossRef] [Green Version]
  161. Kocabas, B.B.; Almacioglu, K.; Bulut, E.A.; Gucluler, G.; Tincer, G.; Bayik, D.; Gursel, M.; Gursel, I. Dual-adjuvant effect of pH-sensitive liposomes loaded with STING and TLR9 agonists regress tumor development by enhancing Th1 immune response. J. Control. Release 2020, 328, 587–595. [Google Scholar] [CrossRef]
  162. Hanson, M.C.; Crespo, M.P.; Abraham, W.; Moynihan, K.D.; Szeto, G.L.; Chen, S.H.; Melo, M.B.; Mueller, S.; Irvine, D.J. Nanoparticulate STING agonists are potent lymph node–targeted vaccine adjuvants. J. Clin. Investig. 2015, 125, 2532–2546. [Google Scholar] [CrossRef] [Green Version]
  163. Shakya, A.K.; Lee, C.H.; Uddin, J.; Gill, H.S. Assessment of Th1/Th2 Bias of STING Agonists Coated on Microneedles for Possible Use in Skin Allergen Immunotherapy. Mol. Pharm. 2018, 15, 5437–5443. [Google Scholar] [CrossRef] [PubMed]
  164. Mittal, A.; Schulze, K.; Ebensen, T.; Weissmann, S.; Hansen, S.; Guzmán, C.A.; Lehr, C.-M. Inverse micellar sugar glass (IMSG) nanoparticles for transfollicular vaccination. J. Control. Release 2015, 206, 140–152. [Google Scholar] [CrossRef] [Green Version]
  165. Lee, E.; Jang, H.-E.; Kang, Y.Y.; Kim, J.; Ahn, J.-H.; Mok, H. Submicron-sized hydrogels incorporating cyclic dinucleotides for selective delivery and elevated cytokine release in macrophages. Acta Biomater. 2016, 29, 271–281. [Google Scholar] [CrossRef]
  166. Koshy, S.T.; Cheung, A.; Gu, L.; Graveline, A.; Mooney, D.J. Liposomal Delivery Enhances Immune Activation by STING Agonists for Cancer Immunotherapy. Adv. Biosyst. 2017, 1. [Google Scholar] [CrossRef] [PubMed]
  167. Chen, J.; Qiu, M.; Ye, Z.; Nyalile, T.; Li, Y.; Glass, Z.; Zhao, X.; Yang, L.; Chen, J.; Xu, Q. In situ cancer vaccination using lipidoid nanoparticles. Sci. Adv. 2021, 7, eabf1244. [Google Scholar] [CrossRef]
  168. Collier, M.A.; Junkins, R.D.; Gallovic, M.D.; Johnson, B.M.; Johnson, M.M.; Macintyre, A.N.; Sempowski, G.D.; Bachelder, E.M.; Ting, J.P.Y.; Ainslie, K.M. Acetalated Dextran Microparticles for Codelivery of STING and TLR7/8 Agonists. Mol. Pharm. 2018, 15, 4933–4946. [Google Scholar] [CrossRef]
  169. Junkins, R.D.; Gallovic, M.D.; Johnson, B.M.; Collier, M.A.; Watkins-Schulz, R.; Cheng, N.; David, C.N.; McGee, C.E.; Sempowski, G.D.; Shterev, I.; et al. A robust microparticle platform for a STING-targeted adjuvant that enhances both humoral and cellular immunity during vaccination. J. Control. Release 2018, 270, 1–13. [Google Scholar] [CrossRef] [PubMed]
  170. Kelly, S.H.; Cossette, B.J.; Varadhan, A.K.; Wu, Y.; Collier, J.H. Titrating Polyarginine into Nanofibers Enhances Cyclic-Dinucleotide Adjuvanticity in Vitro and after Sublingual Immunization. ACS Biomater. Sci. Eng. 2021, 7, 1876–1888. [Google Scholar] [CrossRef]
  171. Shae, D.; Becker, K.W.; Christov, P.; Yun, D.S.; Lytton-Jean, A.K.R.; Sevimli, S.; Ascano, M.; Kelley, M.; Johnson, D.B.; Balko, J.M.; et al. Endosomolytic polymersomes increase the activity of cyclic dinucleotide STING agonists to enhance cancer immunotherapy. Nat. Nanotechnol. 2019, 14, 269–278. [Google Scholar] [CrossRef]
  172. Schulze, K.; Ebensen, T.; Babiuk, L.A.; Gerdts, V.; Guzman, C.A. Intranasal vaccination with an adjuvanted polyphosphazenes na-noparticle-based vaccine formulation stimulates protective immune responses in mice. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 2169–2178. [Google Scholar] [CrossRef]
  173. Leach, D.G.; Dharmaraj, N.; Piotrowski, S.L.; Lopez-Silva, T.L.; Lei, Y.; Sikora, A.G.; Young, S.; Hartgerink, J.D. STINGel: Controlled release of a cyclic dinucleotide for enhanced cancer immunotherapy. Biomaterials 2018, 163, 67–75. [Google Scholar] [CrossRef] [PubMed]
  174. Nakamura, T.; Miyabe, H.; Hyodo, M.; Sato, Y.; Hayakawa, Y.; Harashima, H. Liposomes loaded with a STING pathway ligand, cyclic di-GMP, enhance cancer immunotherapy against metastatic melanoma. J. Control. Release 2015, 216, 149–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Demoulins, T.; Ruggli, N.; Gerber, M.; Thomann-Harwood, L.J.; Ebensen, T.; Schulze, K.; Guzman, C.A.; McCullough, K.C. Self-Amplifying Pestivirus Replicon RNA Encoding Influenza Virus Nucleoprotein and Hemagglutinin Promote Humoral and Cellular Immune Responses in Pigs. Front. Immunol. 2020, 11, 622385. [Google Scholar] [CrossRef]
  176. Gogoi, H.; Mansouri, S.; Jin, L. The Age of Cyclic Dinucleotide Vaccine Adjuvants. Vaccines 2020, 8, 453. [Google Scholar] [CrossRef] [PubMed]
  177. Yi, G.; Brendel, V.P.; Shu, C.; Li, P.; Palanathan, S.; Kao, C.C. Single Nucleotide Polymorphisms of Human STING Can Affect Innate Immune Response to Cyclic Dinucleotides. PLoS ONE 2013, 8, e77846. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of naturally occurring cyclic dinucleotides known to date. The structure of 2′3′-cGAMP was originally misassigned as 2′2′-cGAMP shown in brackets.
Figure 1. Chemical structures of naturally occurring cyclic dinucleotides known to date. The structure of 2′3′-cGAMP was originally misassigned as 2′2′-cGAMP shown in brackets.
Vaccines 09 00917 g001
Figure 2. The number of published records on c-di-GMP and analogs retrieved (as of 20 May 2021) from the Web of Sciences with either topic or title containing one of the following terms: c-diGMP, c-di-GMP, cyclic di-GMP, cyclic diguanylic acid, c-GAMP, cGAMP, c-di-AMP, c-di-AMP, cyclic GMP-AMP, or cyclic dinucleotide, with duplicates removed.
Figure 2. The number of published records on c-di-GMP and analogs retrieved (as of 20 May 2021) from the Web of Sciences with either topic or title containing one of the following terms: c-diGMP, c-di-GMP, cyclic di-GMP, cyclic diguanylic acid, c-GAMP, cGAMP, c-di-AMP, c-di-AMP, cyclic GMP-AMP, or cyclic dinucleotide, with duplicates removed.
Vaccines 09 00917 g002
Figure 3. Processes regulated by c-di-GMP signalling networks.
Figure 3. Processes regulated by c-di-GMP signalling networks.
Vaccines 09 00917 g003
Figure 4. Innate immune sensing of bacterial cyclic dinucleotides and microbial DNA through the cGAS-STING pathway. Reproduced from Figure 1 of the work of [46] with permission from Copyright Clearance Center, Inc. ER: endoplasmic reticulum; TBK-1: TANK-binding kinase 1; IRF3: interferon regulatory factor 3; NF-κB: nuclear factor kappa B; IFN-β: interferon-β.
Figure 4. Innate immune sensing of bacterial cyclic dinucleotides and microbial DNA through the cGAS-STING pathway. Reproduced from Figure 1 of the work of [46] with permission from Copyright Clearance Center, Inc. ER: endoplasmic reticulum; TBK-1: TANK-binding kinase 1; IRF3: interferon regulatory factor 3; NF-κB: nuclear factor kappa B; IFN-β: interferon-β.
Vaccines 09 00917 g004
Figure 5. Activation of STING positively regulates each step of the cancer immunity cycle. Reproduced from Figure 2 of [84] with permission under the terms of the Creative Commons Attribution 4.0 International Licence (http://creativecommons.org/licenses/by/4.0/, accessed on 5 August 2021).
Figure 5. Activation of STING positively regulates each step of the cancer immunity cycle. Reproduced from Figure 2 of [84] with permission under the terms of the Creative Commons Attribution 4.0 International Licence (http://creativecommons.org/licenses/by/4.0/, accessed on 5 August 2021).
Vaccines 09 00917 g005
Table 1. Selected application of cyclic dinucleotides (CDNs) as adjuvants in the development of vaccines against infectious diseases and cancers.
Table 1. Selected application of cyclic dinucleotides (CDNs) as adjuvants in the development of vaccines against infectious diseases and cancers.
CDNsOthersSpeciesImmunization RouteAntigensImmunogenicityEfficacyReferences
c-di-GMP MiceSCBeta-galSerum IgG, Th1/Th2 [91]
c-di-GMP MiceINBeta-gal, OvaSerum IgG, mucosal IgA, Th1, CTL [92]
c-di-GMP MiceIN, IMInfluenza H5N1 (A/Anhui/1/05)Mucosal IgA, serum IgG, balanced Th1/Th2, high frequencies of multifunctional Th1 CD4+ cells. IN > IM [105]
c-di-GMP MiceIN, sublingual, IMInfluenza H5N1 (A/Anhui/1/05)Mucosal IgA, serum IgG, balanced Th1/Th2, high frequencies of splenic H5N1-specific multifunctional (IL-2+TNF-α+) CD4+ T cells.
IN or SL > IM
[106]
c-di-GMPChitosanMiceINNIBRG-14 (H5N1) HASerum and local antibody responses, HI antibody, higher frequencies of virus-specific polyfunctional CD4+ T cells, more Th1 cytokines (IFN-γ, IL-2, TNF-α). 7.5 μg > 1.5 or 0.3 [107]
c-di-GMPSilica nanoparticles (SiO2)MiceIntratrachealPlant-produced H1N1 influenza HAHigh systemic antibody responses, local IgG and IgA responses, local T-cell response (IL-2 and IFN-γ) [108]
c-di-GMP Mice, ferretsINHA A/Indonesia/05/05 (H5N1)IgG and IgA antibodiesSurvived the viral challenge[109]
c-di-GMPmicroneedleMiceMicroneedle skin patchesInfluenza microneedle vaccineEnhanced IgG, IgG subtypes, and cellular immune responses100% survival rate and rapid weight recovery[110]
c-di-GMPB. pertussis LP1569 (a TLR2 agonist)MiceSC, IP, INAcellular pertussis vaccineIFN-β, IL-12 and IL-23 responses, maturation of dendritic cells; IN: Th1 and Th17 responses; Th17 response and IL-17-secreting TRM cellsIN: protection against nasal colonization and lung infection (sustained for at least 10 months)[111]
c-di-GMP MiceSCClfA or mSEC
(S. aureus)
High titers of IgG1, IgG2a, IgG2b and IgG3 compared to alum adjuvant
mSEC > ClfA
Higher survival rates at day 7 with S. aureus challenge[112]
c-di-GMP MiceIN, IPS. pneumoniae PdB or PspAIP induced higher antigen-specific antibodiesSignificant decrease in bacterial load in lungs and blood; IP: increased survival compared to that with alum adjuvant[62]
c-di-GMP MiceINS. pneumonia PsaAStrong IgG and IgASignificantly reduced nasopharyngeal colonization[61]
c-di-GMPDNA vaccineMiceIDHIV-1 reverse transcriptaseTransient increase in IFN-γ production [113]
c-di-GMP Mice,
cattle,
pigs
IMFoot and mouth disease vaccineEarly onset of high neutralizing antibody titers; long-lasting immune memory response [114]
c-di-GMP MiceINGhrelin-PspASpecific antibody responsesReduced body weight gain in diet-induced obesity mice[115]
c-di-GMP Rats, miceINAngiotensin II type 1 receptor (AT1R) peptide conjugated with PspASpecific antibody responsesPrevented the development of hypertension in spontaneously hypertensive rats; Sera protected mice against lethal pneumococcal infection[116]
c-di-GMP and c-di-AMP MiceIN, sublingualH5N1 virosomesLocal and systemic humoral and cellular immune responses; long-lasting immunity; dose-sparingEffective protection against influenza H5N1[117]
c-di-AMP MiceSCOva (soluble or DC targeted)Serum IgG, Th1, CTL, and IFNγ-producing CD8 memory T-cell response, better than PolyI:C/CpG [96]
c-di-AMP MiceINInfluenza nucleoproteinIgG and IgA responses, strong Th1 response (IFN-γ and IL-2)Protection against A/Puerto Rico/8/34 (H1N1) virus[118]
c-di-AMP MiceINESAT-6Innate and adaptive immune responses and regulated autophagy of macrophagesProtection against i.v. challenge similar to antigen alone group[119]
c-di-AMPLipopeptide-based nano carrier systemsMiceINT and B cell epitopes of S. pyogenes M proteinAntigen dose-sparing [120]
c-di-AMPArchaeosomesMiceIN-IN-IN,
IM-IN-IN, or
IM-IM-IM
HCV rE1E2Induced more robust polyfunctional CD4+ T-cell responses [121]
c-di-AMP MiceINBtaF trimeric autotransporterLocal and systemic antibody responses, central memory CD4+ T cells, and strong Th1 responsesProtection against intragastric, but not respiratory, challenge with B. suis[122]
c-di-AMPConjugated at N-terminal of target antigenMiceINT. cruzi recombinant Tc52Predominantly Th17 and Th1 immune responsesBetter protection against infection with lower parasitemia and weight loss, and higher survival rates[123]
c-di-AMP MiceINTraspain (trivalent T. cruzi antigen)Primed Th1/Th17 immune responseReduced parasite load and chronic inflammation[124,125]
c-di-AMPPoly(lactic-co-glycolic acid (PLGA) microparticlesPigletsIMInactivated M. hyopneumoniae field isolate BA 2940-99Strong innate immune responses and robust Th1 or Th17 responses [126]
c-di-AMP or c-di-IMP MiceINBeta-gal, OvaSerum IgG, mucosal IgA, Th1/Th2/Th17, CTL [49,93]
cGAMP MiceIN, single doseInactivated whole virus H7N9 vaccine Enhanced humoral, cellular, and mucosal immune responses. significantly higher nucleoprotein-specific CD4+ and CD8+ T-cell responsesProtection against a high lethal dose, effective cross-protection against H1N1, H3N2, and H9N2 influenza virus[127]
cGAMP MiceINHA vaccineEnhanced IgA, IgG, and T cell responses, including in nasal-associated lymphoid tissue [128]
cGAMP PigsIDH5N1 and 2009 H1N1 pandemic influenza vaccinesVigorous immune responses elicited with no overt skin irritation [129]
cGAMPBiomimetic liposomesMice,
ferrets
INH1N1 vaccineVigorously augmented humoral and CD8+ T-cell responses, importance of alveolar epithelial cells in heterosubtypic immunityStrong cross-protection against distant H1N1 and heterosubtypic H3N2, H5N1, and H7N9 viruses[130]
cGAMPMicroneedle patches and
saponin (Quil-A)
Aged miceMicroneedle patchesH1N1 vaccineIncreased IgG and IgG2aComplete protection[131]
cGAMP MiceIN, SC, IMH. pylori urease A, urease B, and neutrophil-activating proteinSerum IgG and mucosal IgA, Th1, and particularly Th17 responses (IN, SC), IL-17 responses (IN)Significantly reduced gastric mucosal H. pylori colonization (IN, SC)[132]
cGAMP MiceIMM. tuberculosis Rv3852 (H-NS)High serum levels of IL-2, IL-12p40, and TNF-α, specific CD4 and CD8 T-cell responsesReduced bacterial counts in the spleen but not in the lung after H37Rv challenge[133]
cGAMPCpG-C ODNMiceSCHPV 16 E7 (E7GRG)High IgG level and cell proliferation; IFN-γ and granzyme B levelsSuppressed TC-1 tumour growth[134]
cGAMP MiceNot specifiedHBsAgSignificantly enhanced humoral and cellular immune responses [135]
cGAMPAlum (Alhydrogel)Mice
(newborns)
IMrHA influenza vaccine (Flublok)Enhanced HA-specific IgG2a/c titers, IFNγ production by CD4+ T cells, increased T follicular helper cell and GL-7+CD138+ germinal centre B cell responses in newborns [136]
3′3′ cGAMPAcetalated dextran (Ace-DEX) microparticulesMiceIMInfluenza matrix protein 2 (M2e)Humoral and cellular responses, broadly protective immunitySubstantial cross-protection against distant H1N1 and heterosubtypic H3N2, H5N1, and H7N9 viruses[137]
3′3′ cGAMP MiceSLBacillus anthracis protective antigen (PA)Higher serum IgG and saliva IgA than PA + cholera toxin; Th1, Th2, and Th17 responses;
rapid IFN-β and IL-10 responses in sublingual tissues and cervical lymph
[138]
RR-CDG MiceIN, SCAntigen-85B, ESAT-6, Rv1733c, Rv2626c, and RpfD fusion proteinIN: significantly boosted BCG-induced immunity; elicited both Th1 and Th17 immune responsesIN: superior protection than BCG[139]
RR-CDG NanoparticlesMiceSCY. pestis F1-VInduced rapid and long-lived protective immunity100% protected from Y. pestis lethal challenge within 14 days post-immunization, and 75% protected at 182 days post-immunization[140]
rBCG-disA-OEEndogenous c-di-AMPGuinea pigsIDBCGSignificantly stronger TNF-α, IL-6, IL-1β, IRF3, and IFN-β levels than BCG in murine macrophages cultureSignificantly reduced lung weights, pathology scores, and bacterial counts in lungs vs. BCG[104]
rBCG-DisAEndogenous c-di-AMPMiceSCBCGProduced more IFN-γ, IL-2, and IL-10,
stronger expression of H3K4me3 vs. BCG
No additional protection against M. tuberculosis infection[103]
rBCG (BCGΔBCG1419c) MiceSCBCGBetter activation of specific T-cell population vs. BCGNo difference in protection vs. BCG[102]
rBCG-Rv1357cc-di-GMP phosphodiesterase gene Rv1357cMiceIDBCGMore phagocytosed vs. BCGSimilar protection against M. tuberculosis challenge vs. BCG[101]
SC, Subcutaneous; Beta-gal, Beta-galactosidase; Th1/Th2, Type 1/type 2 T helper cells; IN, Intranasal; OVA, Ovalbumin; CTL, Cytotoxic T lymphocytes; IM, Intramuscular; IL, Interleukin; TNF-α, Tumor necrosis factor type; HI, Hemagglutination inhibition; IFN, Interferon; TLR, Toll-like receptors; IP, Intraperitoneal; Th17, T helper cells type 17; TRM cells, Tissue-resident memory T cells; ClfA, Clumping factor A; mSEC, mutant Staphylococcal enterotoxin C; PdB, genetic toxoid derivative of Pneumolysin; PspA, Pneumococcal surface protein A; PsaA, Pneumococcal surface adhesin A; ID, Intradermal; AT1R, Angiotensin II type 1 receptor; ESAT-6, 6 kDa Mycobacterium tuberculosis early secretory antigenic target; i.v, Intravenous; HCV, Hepatitis C virus; HPV, Human papillomaviruses; HBsAg, Hepatitis B surface antigen; BCG, Bacille Calmette-Guérin.
Table 2. Selected examples of cyclic dinucleotides (CDNs) delivery systems.
Table 2. Selected examples of cyclic dinucleotides (CDNs) delivery systems.
CDNsDeliver SystemsFunctionReference
CDNBiodegradable, poly(beta-amino ester) nanoparticlesCytosol delivery and 10-fold improved treatment potency in established B16 melanoma tumours with anti-PD-1 in mice compared to free CDN[80]
c-di-GMPLiposomes made of a synthetic, pH-sensitive lipid of high fusogenicity (YSK05)Deliver into the cytosol and induced melanoma regression[160,161]
c-di-GMPLiposome nanoparticlesTargeting lymphatics and draining lymph nodes after parental injection[162]
c-di-GMP and 3′3′-cGAMPCell-penetrating peptideEnhanced both cellular delivery and biological activity of the c-di-GMP in murine splenocytes[97]
c-di-GMPCytotoxic cationic silica nanoparticlesLocal retention and potent melanoma growth inhibition[79]
c-di-GMPMesoporous silica nanoparticle (immuno-MSN)Brain delivery for treatment of glioblastoma multiforme[77]
c-di-GMPRhodamine B isothiocyanate fluorescent mesoporous silica nanoparticles, synthesized and modified with poly(ethylene glycol) and an ammonium-based cationic molecule“In situ vaccination” strategy; dramatic inhibition of 4T1 breast tumour growth in mice[78]
c-di-GMP and c-di-AMPMicroneedles (MN)Skin delivery better than SC and MN-based alum[163]
c-di-AMPSurfactant-based inverse micellar sugar glass nanoparticles (NPs)Needle-free transfollicular antigen delivery from intact skin; better than polylactic-co-glycolic acid (PLGA) and chitosan-PLGA NPs[164]
c-di-AMPCationic liposomesEnhanced M. hyopneumoniae-specific antibody and T-cell responses[126]
cGAMPA polymersome delivery platformImproved half-life of cGAMP, enhanced immune activation, and tumour growth suppression[74]
cGAMPLinear polyethyleneimine/hyaluronic acid hydrogelsDelivered into phagocytic macrophage cells[165]
cGAMPCationic liposomes with varying surface polyethylene glycol levelsCytosolic delivery and delivery to metastatic melanoma tumour sites and APCs[166]
cGAMPMicroneedle skin patches (MNPs)Increased IgG and IgG2a responses to influenza vaccines in aged mice (21-month-old), and full protection against challenge[131]
cGAMPLipidoid nanoparticle (93-O17S-F)Cytosolic delivery[167]
PD-1, programmed cell death protein 1; SC, subcutaneous; APCs, antigen-presenting cells.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yan, H.; Chen, W. The Promise and Challenges of Cyclic Dinucleotides as Molecular Adjuvants for Vaccine Development. Vaccines 2021, 9, 917. https://doi.org/10.3390/vaccines9080917

AMA Style

Yan H, Chen W. The Promise and Challenges of Cyclic Dinucleotides as Molecular Adjuvants for Vaccine Development. Vaccines. 2021; 9(8):917. https://doi.org/10.3390/vaccines9080917

Chicago/Turabian Style

Yan, Hongbin, and Wangxue Chen. 2021. "The Promise and Challenges of Cyclic Dinucleotides as Molecular Adjuvants for Vaccine Development" Vaccines 9, no. 8: 917. https://doi.org/10.3390/vaccines9080917

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop