Abstract
Nucleic acid sensors, primarily TLR and RLR family members, as well as cGAS–STING signalling, play a critical role in the preservation of cellular and organismal homeostasis. Accordingly, deregulated nucleic acid sensing contributes to the origin of a diverse range of disorders, including infectious diseases, as well as cardiovascular, autoimmune and neoplastic conditions. Accumulating evidence indicates that normalizing aberrant nucleic acid sensing can mediate robust therapeutic effects. However, targeting nucleic acid sensors with pharmacological agents, such as STING agonists, presents multiple obstacles, including drug-, target-, disease- and host-related issues. Here, we discuss preclinical and clinical data supporting the potential of this therapeutic paradigm and highlight key limitations and possible strategies to overcome them.
Introduction
Protecting the intracellular microenvironment from infectious pathogens is an ancient strategy for the preservation of cellular homeostasis. Accordingly, both prokaryotic and eukaryotic cells are equipped with mechanisms for the detection of exogenous nucleic acids, based on either nucleotide sequence (for example, prokaryotic CRISPR–Cas systems) or sequence-independent features (for example, eukaryotic pattern recognition receptors (PRRs))1,2 (Box 1). With the acquisition of multicellularity, early eukaryotes faced the need for preservation of organismal (over cellular) homeostasis. Thus, most (if not all) mechanisms of adaptation to cellular stress, including pathogen invasion, became able to initiate and coordinate systemic responses aimed at the maintenance of organismal homeostasis3. In addition, the molecular machinery for nucleic acid sensing underwent considerable expansion and diversification, and acquired the ability to detect endogenous nucleic acids localizing in non-physiological subcellular compartments, folding with non-conventional secondary structures or bearing specific chemical alterations4. Thus, modern mammals encode various compartmentalized systems that respond to DNA and RNA molecules of potential pathogenic significance (that is, potentially associated with cellular or organismal dysfunction)2. Such PRRs act by initiating a set of highly overlapping signal transduction pathways aimed at the restoration of cellular functions, in the context of preserved systemic homeostasis. This integrated response involves alterations of the intracellular microenvironment coupled with the secretion of bioactive factors that generally convey a danger signal as they mediate immunomodulatory effects, such as cytokines and damage-associated molecular patterns5,6 (Fig. 1).
When sufficient amounts of nucleic acids exhibiting structural features potentially recognized as non-self are available for physical engagement with their sensors (Box 1), the latter initiate signalling pathways that generally culminate in the activation of an adaptative response aimed at the restoration of cellular homeostasis (step 1) and the emission of bioactive factors (step 2), including cytokines and damage-associated molecular patterns (DAMPs). Depending on their concentration, stability and potential for systemic distribution within the bloodstream, these molecules can elicit biological responses in neighbouring cells of different types (step 3), as well as in distant cells (step 4), ultimately favouring the preservation of microenvironmental and organismal homeostasis. CR, cytokine receptor; PRR, pattern recognition receptor.
The detection of potentially pathogenic nucleic acids in mammalian cells involves mainly (1) specific members of the Toll-like receptor (TLR) family, including the RNA sensors TLR3, TLR7 and TLR8, as well as the DNA sensor TLR9 (ref.7), (2) so-called RIG-I-like receptors (RLRs), a group of RNA-specific PRRs named after RIG-I (retinoic acid-inducible gene I protein; also known as DDX58)8, and (3) cyclic GMP–AMP synthase (cGAS), a sensor of cytosolic double-stranded DNA (dsDNA) that signals via stimulator of interferon genes (STING; also known as TMEM173)9. Of note, although mammalian TLRs were named after their homologue in Drosophila melanogaster (Toll, the first member of the family to be characterized)10, the latter does not act as a bona fide PRR but mediates immunological effects downstream of a proteolytic cascade driven by recognition of microbial products by circulating or transmembrane receptors11 (Box 2). Accumulating evidence indicates that defects in TLR, RLR and cGAS signalling contribute to multiple human disorders beyond infectious diseases, encompassing cardiovascular, autoimmune and neoplastic conditions12. Consistent with this notion, normalizing aberrant nucleic acid sensing or the consequences thereof (for example, deregulated cytokine secretion) mediates beneficial effects in numerous preclinical models of disease12.
Nucleic acid sensors (NASs) have been standing out as attractive targets for the development of novel drugs for decades13. Nonetheless, very few pharmacological agents targeting NASs are currently available for use in humans. Existing drugs include (1) imiquimod, a synthetic TLR7 agonist licensed by the US Food and Drug Administration (FDA) for the topical treatment of genital warts, superficial basal cell carcinoma and actinic keratosis14 and (2) an oligonucleotide rich in CpG regions (acting as a TLR9 agonist) currently approved as an adjuvant to the hepatitis B virus (HBV) vaccine HEPLISAV-B15. In addition, bacillus Calmette–Guérin, an inactivated variant of Mycobacterium bovis, is used as a vaccine against tuberculosis, as well as for the treatment of non-invasive bladder carcinoma16, and Cadi-05, a preparation from Mycobacterium indicus pranii, is approved as an adjuvant for the treatment of leprosy17. However, while these agents are generally considered as mixed TLR2, TLR4 and TLR9 agonists, the largest fraction of their biological effects originates from TLR2 and TLR4 signalling16 and will therefore not be discussed further in this Review.
Here, we discuss preclinical and clinical data demonstrating the vast therapeutic potential of NAS-targeting molecules, identify the obstacles preventing the complete realization of such potential and formulate strategies to foster the clinical development of this therapeutic paradigm. The promise of targeting signalling pathways elicited by free nucleotides (for example, ATP) or their products (for example, adenosine) for therapeutic purposes was recently discussed elsewhere18 and hence is not covered in this Review. Similarly, discussion of the pharmacological modulation of signal transducers acting downstream of NASs is beyond the scope of this Review.
TLR agonists
The signalling cascades elicited by nucleic acid-sensing TLRs (Box 2), primarily TLR3, TLR7, TLR8 and TLR9, play a major role in the control of infectious and neoplastic disorders. Accordingly, a large number of agonists for one or more such TLRs are commercially available, and multiple agents have recently entered clinical development (Table 1). Despite such a potential, the development of pharmacological agonists of nucleic acid-sensing TLRs faces several obstacles, as discussed later in this Review.
TLR3 agonists
TLR3 is an endosomal TLR primarily expressed by B cells, specific dendritic cell (DC) subsets and epithelial and endothelial cells19. Viral double-stranded RNA (dsRNA) molecules are the prototypic ligands for TLR3 (ref.20) (Box 1), which relies on Toll-like receptor adaptor molecule 2 (TICAM2) and TICAM1 (also known as TRIF) for signalling21. Depending on the experimental setting, the Tlr3−/− genotype has been associated with detrimental, neutral or beneficial effects on antiviral responses22. Despite these apparently contradictory findings, which mainly reflect the use of different experimental end points, and the potentially divergent effects of short-term versus long-term NAS signalling3, robust preclinical evidence suggests that TLR3 agonists can be harnessed as immunostimulants in multiple settings23. Moreover, autosomal dominant mutations in TLR3 have recently been associated with severe influenza pneumonitis in children24, further reinforcing the relevance of TLR3 for antiviral responses in humans.
Commonly used TLR3 agonists include the synthetic dsRNA analogue polyinosinic:polycytidylic acid (poly(I:C)) and two derivatives: (1) poly(I:C) stabilized with poly(l-lysine) and carboxymethylcellulose (poly-ICLC; Hiltonol) and (2) poly(I:C) containing uridylic acid in a 12:1 molar ratio in the poly(C) strand (poly(I:C12U), also known as rintatolimod and marketed as Ampligen). Poly(I:C) was demonstrated to drive robust interferon responses and to favour cross-priming in both mouse and human experimental systems25,26, largely reflecting the elevated levels of TLR3 in CD8α+ DCs27. Abundant preclinical data demonstrate that poly(I:C) administered locally is a potent adjuvant for viral prophylaxis and therapeutic anticancer vaccination28. However, poly(I:C) is unstable, and results from early clinical trials testing poly(I:C) in patients with leukaemia unveiled side effects including shock, renal failure and hypersensitivity reactions in the context of limited therapeutic efficacy29. Thus, the clinical development of poly(I:C) was terminated (source http://www.clinicaltrials.gov).
Poly-ICLC
Poly-ICLC was initially synthesized to circumvent the stability issues of poly(I:C)30, and entered clinical testing in the late 1970s31, initially for oncological indications, including brain tumours32. Since then, the number of oncological indications in which poly-ICLC has been clinically tested (most often in combination with chemotherapeutics, radiation therapy or cancer vaccines) has grown to include a variety of haematological33,34 and solid35,36,37 malignancies, although most efforts remained focused on brain tumours38,39. In most of these studies, poly-ICLC was well tolerated and mediated robust immunostimulatory effects that, at least in some patients, were associated with clinical benefits37. Today, official sources list more than 50 clinical trials testing poly-ICLC in combination with a variety of (immuno)therapeutics in patients with cancer (source http://www.clinicaltrials.gov) (Table 1). Of note, results from a randomized, placebo-controlled, double-blinded clinical trial (NCT02071095) suggest that poly-ICLC may also be useful for the induction of (at least transient) antiviral responses in HIV-1-positive individuals40. However, no clinical trial is currently open to pursue this therapeutic paradigm (source http://www.clinicaltrials.gov).
Poly(I:C12U)
The first studies evaluating poly(I:C12U) as an anticancer agent were performed in the mid-1980s41, but the development of poly(I:C12U) was soon redirected towards antiviral applications42, largely on the basis of preclinical data demonstrating that poly(I:C12U) was able to reduce the concentration of antiretroviral agents required for HIV-1 control in vitro43. Although additional data in support of the ability of poly(I:C12U) to prevent the decrease in CD4+ levels associated with HIV-1 infection were reported44,45, development was discontinued and no clinical trials testing this therapeutic paradigm are currently open (source http://www.clinicaltrials.gov). In addition, poly(I:C12U) attracted attention as a therapeutic option for chronic fatigue syndrome46. Promising results from early clinical studies fostered the initiation of a phase III trial based on intravenous poly(I:C12U) administration (NCT00215800). In this setting, poly(I:C12U) ameliorated physical symptoms but did not reduce the levels of biochemical markers of disease, and side effects were frequent47. One trial testing intravenous poly(I:C12U) administration in patients with chronic fatigue syndrome is currently open to expanded access (NCT00215813; source http://www.clinicaltrials.gov). In addition, a few clinical trials are currently testing intradermal poly(I:C12U) in combination with immunotherapy in patients with breast cancer (NCT01355393, NCT03599453), ovarian cancer (NCT01312389, NCT02432378, NCT03734692), colorectal cancer (NCT03403634) and prostate cancer (NCT03899987) (source http://www.clinicaltrials.gov) (Table 1), a renewed interest fostered by promising preclinical data on the ability of poly(I:C12U) to act as an adjuvant to cancer vaccines in mice48.
Other agonists
ARNAX is a novel synthetic DNA–dsRNA hybrid molecule that also promotes robust cross-priming by DCs by acting as a TLR3 agonist49. Use of ARNAX in combination with a cancer vaccine and a programmed cell death 1 ligand 1 (PD-L1)-targeting immune checkpoint blocker (ICB) has been proposed as a strategy to overcome resistance to agents targeting programmed cell death 1 (PD-1; also known as PDCD1), at least in mice49. To the best of our knowledge, ARNAX has not yet entered clinical development (source http://www.clinicaltrials.gov).
TLR7 and TLR8 agonists
TLR7 and TLR8 are expressed in the endosomal compartment of DCs, monocytes, macrophages and mast cells50. Of note, TLR7 is preferentially expressed by plasmacytoid DCs and B cells, while TLR8 is mostly found in monocytes, neutrophils, and monocyte-derived DCs51. TLR7 and TLR8 recognize single-stranded RNA (ssRNA) molecules containing GU-rich or poly(U) sequences52,53 (Box 1), resulting in the secretion of multiple cytokines, including type I interferon, downstream of MYD88 signalling54 (Box 3). On the basis of the key role of type I interferon in viral interference, TLR7 has long been considered (together with TLR3 and TLR9) as an ideal target for the development of novel antiviral agents55, which culminated in the development of synthetic imidazoquinolines56. Although most of these compounds failed to allow robust viral interference and mediate direct protective effects on non-infected cells, some imidazoquinolines demonstrated considerable efficacy as immunostimulants23.
Imidazoquinolines as antivirals
Imiquimod (also known as R-837) is a prototypic imidazoquinoline that originally attracted attention for its antiviral effects in animal models of cytomegalovirus and herpes simplex virus 2 (HSV-2) infection57, and is now approved for the topical treatment of genital warts, superficial basal cell carcinoma and actinic keratosis58. Intriguingly, the efficacy of Aldara (5% imiquimod cream) may reflect both TLR7-dependent and TLR7-independent pathways59, underlying the key importance of formulation and drug–excipient interactions in the clinical activity of TLR agonists (and most likely many other drugs). Resiquimod (also known as R-848) is a mixed TLR7 and TLR8 agonist that was also conceived for antiviral applications, reflecting its ability to favour IL-12 secretion and limit viral replication in monocytes isolated from HIV-1-infected individuals60. However, despite promising efficacy results in randomized phase II clinical trials enrolling patients with HSV-2 infection61 or hepatitis C virus (HCV) infection62, the safety profile of resiquimod was not superior to that of recombinant interferon-α (IFNα; the historical gold standard treatment for HCV infection)62. Thus, the clinical development of imiquimod and resiquimod as antiviral agents has been discontinued (source http://www.clinicaltrials.gov).
Imidazoquinolines as anticancer agents
As the development of imidazoquinolines as antivirals was abandoned, efforts have been refocused on anticancer applications, at least partially due to the enthusiasm raised by the approval of imiquimod for the treatment of some skin tumours58. The type I interferon-dependent anticancer effects of systemic imiquimod were first reported in 1992 (ref.63), and since then imiquimod (both topical and systemic) has been demonstrated to mediate therapeutic effects, alone or in combination with other drugs, in a variety of preclinical tumour models, including models of mammary, lung and prostate carcinoma64,65. The anticancer activity of imiquimod has been consistently linked to the CC-chemokine ligand 2 (CCL2)-dependent recruitment of plasmacytoid DCs to the tumour bed, culminating in abundant type I interferon secretion linked to activation of CD4+ T helper cells producing interferon-γ (IFNγ), tumour necrosis factor (TNF) and IL-17, and intradermal γδ T cells, at least in mice66,67. Despite these promising findings, multiple clinical trials testing imiquimod as an off-label intervention in cancer patients, generally in combination with standard chemotherapeutic regimens, failed to demonstrate long-term efficacy68,69, with some notable exceptions, such as the combination of imiquimod and monobenzone in patients with melanoma70. It is tempting to speculate that such a restricted efficacy spectrum reflects the ability of some tumours to establish a robustly immunosuppressive microenvironment71. Supporting this possibility, intratumoural imiquimod exerted superior effects against thymoma cells established in Ido1−/− mice, which lack one enzyme with major immunosuppressive effects72. Similarly, imiquimod has been favourably combined with an ICB targeting cytotoxic T lymphocyte-associated protein 4 (CTLA4), which reverses local immunosuppression, in a patient with nivolumab-resistant melanoma73. The ability of imiquimod to mediate anticancer effects as an off-label intervention is currently being investigated in no fewer than 45 clinical trials, most of which involve topical imiquimod application (source http://www.clinicaltrials.gov) (Table 1).
In a phase I clinical trial, topically applied resiquimod demonstrated clinical activity in patients with cutaneous T cell lymphoma, a skin malignancy for which limited therapeutic options are available74. Specifically, 75% of patients with cutaneous T cell lymphoma being treated with resiquimod experienced a reduction in the number or size of cutaneous lesions, with nearly 30% of them achieving complete eradication of the lesions74. Although data on long-term disease outcome are still missing, these findings point to the possibility that resiquimod may mediate stand-alone therapeutic effects in patients with cutaneous T cell lymphoma. That said, the current development of resiquimod as an anticancer agent is mostly focused on its ability to act as an immunological adjuvant. Indeed, accumulating evidence indicates that topically applied resiquimod can be conveniently combined with peptide-based vaccines, such as recombinant cancer/testis antigen 1B (CTAG1B; also known as NY-ESO-1), to reactivate clinically relevant CD8+ cytotoxic T lymphocyte (CTL) responses against melanoma75 or can be used to reverse immunosuppression in models of cervical carcinoma linked to human papilloma virus 16 infection76. Nonetheless, only two clinical trials testing resiquimod as an immunological adjuvant for cancer therapy are currently active (NCT00960752, NCT02126579) (Table 1).
Recently, motolimod (also known as VTX-2337), another imidazoquinoline specific for TLR8 (ref.77), has attracted some attention. In preclinical studies, motolimod administration to unfractionated white blood cells improved the ability of natural killer cells (NK cells) to mediate antibody-dependent cellular cytotoxicity (ADCC) towards cancer cell lines and freshly isolated head and neck cancer cells78. Moreover, motolimod promoted IL-12, TNF and IFNγ secretion by DCs, thus favouring the activation of tumour-specific CTL responses78. On the basis of these findings, subcutaneously administered motolimod is currently being tested in combination with ICB specific for PD-1 and/or cetuximab in patients with head and neck cancer (NCT02124850, NCT03906526), as well as together with durvalumab plus chemotherapy in women with platinum-resistant ovarian cancer (NCT02431559) (source http://www.clinicaltrials.gov) (Table 1).
Other agonists
Results from the first-in-concept human trial testing the safety, tolerability and immunogenicity of a novel mixed TLR7, TLR8 and RIG-I agonist (CV8102) administered alone or with fractional doses of a licensed rabies vaccine as a model antigen (NCT02238756) have recently been released79. This study identified the safe and efficient dose range for CV8102 (ref.79), initiating further clinical testing in oncological indications. CV8102 is currently being tested in combination with a therapeutic vaccine in patients with hepatocellular carcinoma (NCT03203005), and together with a PD-1 blocker in individuals with various types of cancer, including melanoma (source http://www.clinicaltrials.gov) (Table 1).
TLR7 agonists have also been investigated for their ability to limit asthmatic and allergic reactions. The rationale of this approach resides in the fact that allergic responses involve the polarization of CD4+ T cells towards a type 2 T helper cell (TH2) cell profile, which (at least theoretically) can be counteracted by agents promoting TH1 cell polarization (including TLR7 agonists)80. In line with this model, the TLR7 agonist S-28463 reduced airway resistance, leukocyte infiltration and IgE levels in mouse models of allergic sensitization to ovalbumin80. Preliminary clinical data confirmed that the intranasal administration of the TLR7 ligand GSK2245035 is well tolerated and reduces reactivity in patients with allergic rhinitis81. However, after the completion of a series of clinical trials with largely unreported results (NCT01480271, NCT01607372, NCT01788813, NCT02446613, NCT02833974), the sponsor cancelled the last ongoing study (NCT03707678) (source http://www.clinicaltrials.gov). Additional preclinical investigation is required to further understand the potential of TLR7 agonists for asthma treatment.
TLR9 agonists
TLR9 is an endosomal TLR abundantly expressed by a variety of immune cells, including monocytes, plasmacytoid DCs, B cells and neutrophils as well as CD4+, CD8+ and γδ T cells (in both humans and mice)82. Moreover, TLR9 can be found in the pulmonary and intestinal epithelium as well as in keratinocytes82. The prototypical ligands for TLR9 are unmethylated CpG-rich oligodeoxynucleotides (ODNs), which are common in prokaryotic and viral genomes (but not in their mammalian counterparts, as CpG dinucleotides are often methylated therein)83 (Box 1). Immune cells from Tlr9−/− mice are hyporesponsive to CpG-rich ODNs as well as to cytokine stimulation, resulting in increased resistance to otherwise lethal CpG-rich ODN challenges84, but exacerbated sensitivity to pathogen infection85,86. Consistent with this, prophylactic treatment of mice with synthetic TLR9 ligands conveys transient protection against a wide range of viral, bacterial and parasitic pathogens, including Ebola virus, Bacillus anthracis, Listeria monocytogenes and Francisella tularensis87. Moreover, the supernatant of human peripheral blood mononuclear cellss exposed to CpG-rich ODNs allows robust viral interference against HCV, largely as a consequence of type I interferon signalling and downstream expression of 2′-5′-oligoadenylate synthetase 1 (OAS1; which activates cellular RNase L to cleave viral genomes)55. Altogether, these findings pointed to TLR9 as a potential target for the development of potent antiviral agents acting directly (via viral interference) as well as indirectly (on immunostimulation).
CpG-rich ODNs and IMOs as antivirals
Initial efforts focused on immunomodulatory oligonucleotides (IMOs), synthetic TLR9 agonists that incorporate cytosine or guanine analogues. As compared with CpG-rich ODNs, IMOs display increased stability, species-independent activity and a clear structure–activity relationship88. In line with this notion, IMOs were shown to promote IL-12 and IFNγ production in mouse, monkey and human cellular systems, as well as expression of activation markers such as CD86 and CD69 by plasmacytoid DCs and B cells, respectively88. Moreover, administration of IMOs to wild-type mice induced activation of Janus kinase–signal transducer and activator of transcription (JAK–STAT) signalling downstream of TLR9, type I interferon signalling and IFNγ signalling, as demonstrated in mice lacking TLR9 and mice lacking type I interferon receptors or IFNγ89. Such activity is relevant for chronic HCV infection, a setting in which patients often become resistant to treatment with recombinant IFNα90. Indeed, the liver of mice pretreated with IFNα remained responsive to transactivation of interferon-stimulated genes (ISGs) by IMOs, and the ISG-activatory activity of IMOs compared favourably with that of IFNα, correlating with increased hepatic infiltration by immune cells89. On the basis of these findings, one specific IMO (tilsotolimod, also known as IMO-2125) has been evaluated in clinical trials for toxicity in HCV-infected patients (NCT00728936, NCT00990938). Preliminary results from these studies suggest that the use of tilsotolimod as a stand-alone intervention is associated with a limited incidence of serious adverse events, which is greatly exacerbated in the presence of ribavirin (source http://www.clinicaltrials.gov). To the best of our knowledge, the clinical development of IMOs as antiviral agents has now been discontinued.
TLR9 agonists have also attracted attention as potential therapeutics for chronic HIV infection. Initial work with the mouse Friend retrovirus model demonstrated that CpG-rich ODNs are an effective post-exposure treatment, which is linked to considerable reductions in circulating and splenic viral load in the context of TH1 cell cytokine secretion and activation of virus-specific CTL responses91. Surprisingly, prophylactic administration of CpG-rich ODNs not only failed to prevent infection but aggravated viral load in this model91. Similar observations were made in rhesus monkeys treated intravaginally with CpG-rich ODNs and then exposed to simian immunodeficiency virus (SIV) via the same route92. Along similar lines, TLR9 activation in mouse splenic cells engineered to express the HIV-1 genome appears to favour (rather than inhibit) viral replication93, and a similar activation reportedly occurs in human latently infected mast cells94. Conversely, CpG-rich ODNs suppress the replication of both CC-chemokine receptor 5 (CCR5)-tropic and CXC-chemokine receptor 4 (CXCR4)-tropic HIV-1 in human lymphoid tissue ex vivo95. Although the precise molecular mechanisms explaining these apparently paradoxical observations remain to be elucidated, it is tempting to speculate that cellular context plays a major role in this setting. Results from early clinical studies investigating lefitolimod (a CpG-rich ODN with a covalently closed dumbbell shape, also known as MGN1703) in HIV-1-positive patients indicate that this approach is safe and induces robust virus-specific humoral and cellular immunity96, as well as signatures of type I interferon responses in the colonic epithelium97. However, long-term clinical benefits appear to be limited, as only one of 12 participants achieved prolonged control of viraemia96. Only one clinical study testing lefitolimod as an antiviral agent (in combination with HIV-1-specific antibodies) is currently open according to official sources (NCT03837756). Moreover, lefitolimod is being investigated in combination with chemotherapy or immunotherapy in cancer patients (NCT02077868, NCT02668770) (Table 1).
CpG-rich ODNs and IMOs as vaccine adjuvants
The development of TLR9 agonists as immunological adjuvants to prophylactic vaccines against viral infection culminated with the approval of one CpG-rich ODN, namely CpG-1018 (also known as 1018 ISS), for use in combination with recombinant hepatitis B surface antigen (HBsAg) as an HBV vaccine for humans in November 2017 (ref.15). Agatolimod (also known as CpG-7909 or PF-3512676) is another synthetic CpG-rich ODN tested as an adjuvant for prophylactic HBV vaccination. Despite promising preclinical and clinical findings, however, agatolimod failed clinical development for this indication98, for unclear reasons. In this setting, improved CpG-rich ODN formulations, including ODNs wrapped with non-agonistic ligands for DC receptors such as C-type lectin domain containing 7A (CLEC7A), have been shown to mediate superior immunostimulatory effects99. Recombinant HBsAg, hepatitis B core antigen (HBcAg) and CpG-rich ODNs have also been investigated as therapeutic vaccines against HBV, at least in preclinical models100. In this setting, vaccination promoted robust humoral responses against both HBsAg and HBcAg as well as potent CD8+ and CD4+ T cell responses that limited the circulating manifestation of infection in diseased mice, with no evident liver toxicity100. However, neither of these latter approaches has yet entered clinical development to our knowledge (source http://www.clinicaltrials.gov).
CpG ODNs and IMOs as anticancer agents
TLR9 is often expressed in the tumour microenvironment, not only by immune cells but also by (at least some) malignant cells and the tumour endothelium. Early efforts to use TLR9 agonists for cancer therapy were therefore focused on the activation of regulated cell death in malignant cells or the inhibition of neoangiogenesis101,102. However, it soon became clear that the greatest promise of TLR9 agonists for cancer therapy resided in their ability to promote type I interferon secretion, at least theoretically, setting the stage for the repolarization of the tumour microenvironment from an immunosuppressive state characterized by high levels of myeloid-derived suppressor cells and M2-like tumour-associated macrophages to an immunostimulatory one characterized by elevated levels of mature DCs and M1-like tumour-associated macrophages103. Abundant preclinical data suggest that TLR9 agonists are generally insufficient to mediate robust anticancer effects but can be conveniently combined with a variety of immunotherapeutics, especially when administered intratumourally104,105,106.
Results from clinical trials testing CpG-rich ODNs or IMOs in patients with haematological107,108 and solid tumours109,110,111, generally in combination with standard-of-care therapy, demonstrated that this therapeutic approach is associated with acceptable safety and tolerability, as well as preliminary evidence for activity. However, subsequent randomized studies generally restricted the initial enthusiasm, often owing to modest efficacy112 and accrued toxicity113,114. In particular, agatolimod failed to increase progression-free and overall survival in a phase III study on patients with lung cancer receiving standard-of-care chemotherapy113,114, despite promising results in a similar phase II setting115. IMOs were often administered subcutaneously in these studies, which may explain the relative lack of efficacy. Further supporting this possibility, CpG-rich ODNs delivered locally to patients with early-stage melanoma conferred durable disease control in two recent randomized phase II studies116. Of note, CpG-rich ODNs and IMOs often drive CTL responses characterized by elevated PD-1 expression117, suggesting that combinatorial regimens involving ICBs could be particularly efficient in this setting. Data in support of this possibility are emerging118. In particular, intratumoural administration of SD-101, a synthetic CpG-rich ODN, in the context of pembrolizumab therapy resulted in improved immune activation in patients with melanoma118. Similarly, SD-101 could be favourably combined with low-dose radiation therapy in patients with low-grade B cell lymphoma, resulting in a high rate of objective responses at non-irradiated tumour sites119, an immunological phenomenon commonly known as abscopal response120.
Several CpG-rich ODNs and IMOs are currently being tested in clinical trials for their ability to enhance the efficacy of chemotherapy, radiation therapy or ICB-based immunotherapy in patients with a variety of tumours. These oncological indications include (but are not limited to) head and neck cancer (NCT02521870), colorectal cancer (NCT02077868), melanoma (NCT02521870, NCT02644967, NCT03445533), lymphoma (NCT02927964, NCT03322384, NCT03410901), breast carcinoma (NCT01042379) and mixed solid tumours (NCT03007732, NCT03052205, NCT03831295) (Table 1). Of note, most of these trials involve SD-101, while the clinical development of agatolimod as an anticancer agent has been discontinued (source http://www.clinicaltrials.gov).
Other applications
Similarly to TLR7 agonists, TLR9 agonists have also attracted attention as potential agents for the management of allergic reactions121. Supporting the rationale of using TLR9 agonists to limit TH2 cell immune responses by boosting their TH1 cell counterparts, a CpG-rich ODN efficiently suppressed IL-33-driven airway hyperreactivity downstream of type I interferon-dependent activation of NK cells and consequent secretion of IFNγ (a potent TH1 cell polarizer) in mice122. However, a few studies testing this therapeutic paradigm in patients with moderate to severe allergic asthma (NCT01673672, NCT02087644) were prematurely withdrawn or terminated as they did not meet the primary end point (source http://www.clinicaltrials.gov).
RLR agonists
RLRs are a cytosolic family of DexD/H-box RNA helicases that respond to viral RNA molecules (Box 1) by activating type I interferon secretion downstream of mitochondrial antiviral signalling protein (MAVS) and TANK-binding kinase 1 (TBK1) activation8,123 (Box 5). RIG-I is arguably the best characterized member of the RLR family, and was first described only 15 years ago124. Thus, mammalian RLR research is a considerably younger field than mammalian TLR research. Nonetheless, RLRs are promising targets for the development of antiviral and anticancer agents.
RIG-I agonists
Antiviral agents
The vast majority of currently available RLR agonists targeting RIG-I were conceived for antiviral applications125, largely reflecting the key role of RLRs in natural antiviral defences. SB 9200 (also known as inarigivir soproxil or GS-9992) is an orally available prodrug of a dinucleotide agonist of RIG-I and nucleotide-binding oligomerization domain-containing protein 2 (NOD2), a cytosolic PRR involved in the elimination of invading pathogens126,127. SB 9200 not only favours RIG-I and NOD2 signalling, culminating with type I and type III interferon secretion126 and IL-1β release downstream of inflammasome activation128, respectively, but also locks RIG-I and NOD2 on pregenomic viral RNA, impeding the binding of the viral polymerase and synthesis of the DNA minus strand126. Consistent with this dual activity, SB 9200 mediated robust antiviral effects in woodchucks chronically infected with woodchuck hepatitis virus, a model of chronic HBV infection, both as a stand-alone agent and combined with entecavir126,129. In this setting, hepatic toxicity was limited, and the therapeutic efficacy of SB 9200 was comparable to that of antivirals currently used for the management of HBV infection (for example, tenofovir)126,129 SB 9200 has also been reported to be efficacious in models of chronic HCV infection130. Two clinical trials are currently testing the activity of SB 9200 (alone or combined with tenofovir) in patients with chronic HBV infection (NCT02751996, NCT03434353) (Table 1). To the best of our knowledge, the results from two additional trials that official sources list as completed have not yet been disseminated (source http://www.clinicaltrials.gov).
5′-triphosphate RNA (5′-pppRNA) is a natural agonist of RIG-I (ref.131) (Box 1). Human lung carcinoma A549 cells receiving 5′-pppRNA in vitro reportedly acquire resistance to infection not only by RNA viruses such as dengue virus and chikungunya virus132,133 but also by DNA viruses such as vesicular stomatitis virus and vaccinia virus133, reflecting the establishment of potent, type I interferon-dependent viral interference. Unfortunately, 5′-pppRNA is unstable and unable to cross the plasma membrane, and hence is not a good candidate for clinical development per se. In the attempt to resolve this issue, short stem–loop RNA (SLR) molecules that present a single duplex terminus and a triphosphorylated 5′ end (and hence retain strong RIG-I-binding capacity) have been developed134. SLRs activate RIG-I more potently than longer and unstructured 5′-pppRNA, both in vitro and in vivo, when delivered with a transfection reagent134. According to official sources, SLRs have not yet entered clinical development (source http://www.clinicaltrials.gov).
Anticancer agents
RIG-I has also attracted attention as a potential target for the development of novel anticancer agents135. One particularly interesting approach in this setting is the use of 5′-triphosphate small interfering RNAs, which simultaneously downregulate any protein of choice and drive RIG-I activation136. Recent data indicate that 5′-triphosphate small interfering RNAs specific for ATP-binding cassette subfamily B member 1 (ABCB1; also known as MDR1) mediate cytotoxic effects in chemoresistant leukaemia cells as they (1) increase sensitivity to doxorubicin and (2) drive potent type I interferon responses136. That said, only a few RIG-I agonists have entered clinical development for oncological indications, namely RTG100 (NCT03065023) and MK-4621 (NCT03739138) (Table 1). However, NCT03065023 was prematurely terminated, reflecting the sponsor’s decision to establish a new treatment protocol before continuation, and NCT03739138 has been recruiting for only a few months (source http://www.clinicaltrials.gov). Some cytotoxic chemotherapeutics favour the release of nuclear non-coding RNA molecules in the cytosol, de facto triggering therapeutically relevant RIG-I activation137. These latter findings suggest that commonly used therapies may activate NASs via poorly defined mechanisms that require urgent elucidation.
Other RLR agonists
Nucleic acid band 2 (NAB2) is a dsRNA molecule isolated from yeast that acts as a mixed TLR3 and melanoma differentiation associated protein 5 (MDA5) agonist138. Liposomally delivered NAB2 was superior to poly(I:C) in promoting type I interferon secretion by mouse bone marrow DCs, resulting in improved cross-priming of antigen-specific CTLs139. Similarly, liposomal delivery of NAB2 also stimulated higher type I interferon secretion in vivo as compared with poly(I:C)139, correlating with the ability to promote secretion of a variety of proinflammatory cytokines, including type I interferon and IL-12, by human monocyte-derived DCs138,139. Moreover, NAB2 complexed with a cationic agent effectively acted as an adjuvant to a prophylactic cancer vaccine based on vaccinia virus Ankara encoding a tumour-associated antigen138. BO-112 is a nanoplexed formulation of poly(I:C) complexed with polyethylenimine that on intratumoural administration to mouse melanomas drives a particularly immunogenic variant of cell death downstream of MDA5 signalling140,141, resulting in the activation of systemic immunity against non-treated, distant lesions142. One clinical trial has just been started to test the activity of BO-112 in adults with aggressive solid tumours (NCT02828098; source http://www.clinicaltrials.gov) (Table 1).
Despite there being only two MDA5 agonists under development, MDA5 stands out as a promising target for the creation of novel drugs with antiviral and anticancer applications. Indeed, a homozygous missense mutation in human IFIH1 (encoding MDA5) increased susceptibility to infection by common viruses, including seasonal rhinoviruses143. Moreover, MDA5 stands at the core of a signalling pathway that is particularly relevant for cancer therapy144. Thus, the inappropriate activation of MDA5 and protein kinase R (PKR; another RNA sensor, also known as EIF2AK2)145 by endogenous RNA molecules is normally prevented by A-to-I editing, the major catalytic function of adenosine deaminase RNA specific (ADAR)146. Notably, ADAR deficiency has been associated with decreased cancer cell proliferation, increased release of type I interferon and sensitization to ICB-based immunotherapy in mouse models of ICB-resistant cancers144, and some subsets of cancer cells exhibit bona fide non-oncogene addition to ADAR147. These results indicate that both ADAR inhibitors and MDA5 agonists may constitute optimal combinatorial partners for ICB-based immunotherapy for the management of ICB-resistant tumours. MDA5 also senses highly unstable native mitochondrial dsRNA molecules, which are generally degraded by SUV3-like RNA helicase (SUPV3L1) and polyribonucleotide nucleotidyltransferase 1 (PNPT1)148. Thus, mitochondrial dsRNA molecules stand out as interesting templates for the development of novel MDA5 agonists.
In 2017, a high-throughput screening based on ISG bioluminescent reporters identified the benzobisthiazole compound KIN1000 as a potent RLR inducer, driving the design and synthesis of hundreds of analogues that were tested for potency on the basis of interferon regulatory factor 3 (IRF3) nuclear translocation149. This approach led to the selection of KIN1148, which was further developed as an immunological adjuvant. Consistent with its ability to drive type I interferon responses, KIN1148 allowed prophylactic vaccination of mice against a species-adapted variant of a pandemic human influenza virus149. The actual RLR underlying the adjuvant-like activity of KIN1148, however, remains to be elucidated. Consistent with this lack of mechanistic knowledge, KIN1148 has not yet entered clinical development (source http://www.clinicaltrials.gov).
cGAS–STING agonists
cGAS was first characterized in 2013 as a sensor for cytosolic dsDNA (Box 1) that signals via STING150,151 (Box 5). In the same year, multiple mechanistic details of cGAS–STING signalling were revealed152,153,154, and the cGAS–STING pathway was also attributed key roles in antiviral defences155,156. One year later, type I interferon downstream of STING signalling was shown to underlie the ability of DCs to optimally cross-prime tumour-specific CTLs157,158, identifying the cGAS–STING pathway as a potential target for both antiviral and anticancer interventions. Consistent with this notion, Tmem173−/− mice are unable to mount therapeutic responses to ICB-based immunotherapy157, and STING-deficient cancer cells implanted in immunocompetent animals are impaired in their ability to elicit abscopal responses on irradiation159. Considerable efforts are currently being dedicated to the development of STING (rather than cGAS) agonists (see later), at least in part reflecting the notion that cGAS naturally recognizes dsDNA molecules that require a delivery platform to penetrate the cell membrane (Box 1), while natural STING agonists are small cyclic dinucleotides (CDNs) that can be easily mimicked by small molecules151. Importantly, accumulating evidence suggests that STING agonists lose efficacy when administered locally at high doses, especially in oncological settings160, at least in part reflecting the cytotoxic effects of robust STING activation in T lymphocytes161,162. This calls for the development of sustainable strategies for the delivery of STING agonists, potentially based on the systemic administration of prodrugs163.
DMXAA
5,6-Dimethylxanthone-4-acetic acid (DMXAA; also known as vadimezan or ASA404) is a potent STING agonist164 originally developed as a vasculature disrupting agent165. Consistent with its ability to promote the secretion of type I interferon and TNF, which was recognized long before the identification of STING166, DMXAA has been shown to mediate robust antiviral activity in models of HBV and HSV infection167,168 and stand-alone therapeutic efficacy in preclinical models of acute myeloid leukaemia and mammary carcinoma169,170. Results from multiple phase I and phase II clinical trials testing DXMAA in patients with a variety of tumours in the 2000s demonstrated that DXMAA can be safely administered in biologically active doses in most settings, including combinatorial regimens with cytotoxic chemotherapy171,172. However, none of these and subsequent clinical studies, including a randomized phase III trial testing DXMAA in combination with standard-of-care chemotherapy in patients with advanced non-small-cell lung cancer173, could demonstrate robust therapeutic benefits, and the clinical development of DXMAA was discontinued (source http://www.clinicaltrials.gov). The reasons underlying the dismal efficacy of DXMAA in patients with cancer remain to be elucidated. However, structural studies demonstrated that DXMAA efficiently binds mouse, but not human, STING174,175. Thus, the lack of STING-dependent immunostimulation may be one reason underlying the clinical failure of this agent.
Cyclic dinucleotides
Cyclic GMP–AMP (cGAMP) and other CDNs of bacterial origins are natural STING agonists151,176. Of note, some of these CDNs, such as cyclic diguanylate monophosphate have been successfully used in preclinical mouse tumour models to mediate stand-alone therapeutic activity177 or combinatorial therapeutic activity178. Similarly, cGAMP and cyclic diadenylate monophosphate reactivated latently infected cells and enhanced SIV-specific responses ex vivo in cynomolgus macaques with naturally controlled SIV infection179. However, natural CDNs, including cGAMP, are highly sensitive to hydrolysis by ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1)180, resulting in suboptimal therapeutic activity. Moreover, human (but not mouse) STING exists in different polymorphic variants (that is, wild type, REF, HAQ, R232H, AQ, R293Q, R71H, G230A, and HQ) with differential sensitivity to canonical CDNs181, as well as in multiple splicing isoforms with altered stability182.
Second-generation STING agonists
The considerations noted earlier drove the development of second-generation STING agonists with increased potency, largely based on rational design, including (1) 2′-3′-cGSASMP, a hydrolysis-resistant biphosphothioate analogue of cGAMP180 and (2) ADU-S100 (also known as MIW815 or ML RR-S2 CDA), a hydrolysis-resistant molecule that efficiently activates mouse as well as human STING (in all its isoforms)183. Intratumoural ADU-S100 administration mediated robust therapeutic effects in mouse models of melanoma, mammary carcinoma and colorectal cancer, coupled with the establishment of long-term immunological memory183. ADU-S100 has also been successfully combined with a variety of immunotherapeutic agents in preclinical tumour models, including ICBs and recombinant cytokines184. Along similar lines, a variant of ADU-S100 lacking the mixed linkage (an internal stabilizing bond) was successfully used to boost the efficacy of a cancer cell vaccine producing colony stimulating factor 2 (CSF2; also known as GM-CSF) against experimental mouse melanomas, which was further boosted by PD-1 neutralization185. These findings provided sufficient rationale to initiate the clinical development of ADU-S100, which is currently being tested in combination with CTLA4- or PD-1-blocking antibodies in patients with advanced lymphoma or solid tumours (NCT02675439, NCT03172936, NCT03937141) (Table 1). Importantly, accumulating evidence indicates that the intratumoural administration of STING agonists can mediate cytostatic or cytotoxic effects on CD8+ CTLs161,162 and B cells186. As these findings reduced (at least to some degree) initial expectations for STING agonists as anticancer agents, results from the aforementioned clinical trials are urgently awaited.
Amidobenzimidazoles
The lack of STING agonists that are compatible with systemic delivery prompted the initiation of a high-throughput screening aimed at the identification of cGAMP competitors for STING binding187. In this setting, two amidobenzimidazole compounds were selected and used for crystallographic studies, which revealed that each STING monomer binds one amidobenzimidazole molecule at (or very near to) the cGAMP-binding pocket. On the basis of these findings, a dimeric amidobenzimidazole molecule was developed and optimized for increased stability, leading to the generation of compound 3 (ref.187). Importantly, subcutaneous administration of compound 3 caused production of type I interferon, TNF and IL-6 in wild-type (but not Tmem173−/−) mice, and intravenous delivery mediated robust anticancer effects in a mouse model of colorectal cancer, which were abolished by the depletion of CD8+ CTLs187. These findings identify a promising lead for the development of non-nucleotide STING agonists amenable to intravenous administration.
Antagonizing NASs
Dysregulated cytokine signalling plays a major role in several autoimmune and inflammatory disorders, including (but not limited to) systemic lupus erythematosus (SLE), rheumatoid arthritis, multiple sclerosis, autoimmune encephalitis and psoriasis. Consistent with this notion, these conditions are generally managed with broad-spectrum anti-inflammatory agents such as glucocorticoids or with drugs that specifically target pathogenic cytokines, such as the TNF blocker etanercept or the IL-17A blocker secukinumab188. Accumulating preclinical data (discussed later) indicate that the overproduction of some cytokines involved in autoimmune and inflammatory disorders originates from deregulated nucleic acid sensing, suggesting that antagonists of NASs may be valuable therapeutic tools for these indications (Table 2). In addition, deregulated cytokine signalling downstream of NASs is involved in the origin of diseases previously thought to have no immunological causative component, such as some neurodegenerative and cardiovascular conditions189, further expanding the therapeutic potential of NAS antagonists.
TLR antagonists
Autoimmune disorders
The involvement of TLR3, TLR7, TLR8 and TLR9 in autoimmune and inflammatory conditions was first hypothesized on the basis of genome-wide association studies190. Consistent with this notion, MRL/lpr mice (an experimental model of SLE) exhibit nephritic kidney degeneration coupled with extensive infiltration by TLR3+ cells, and it was suggested that disease progression can be accelerated by IL-6, IL-12 and type I interferon production downstream of systemic poly(I:C) administration191. However, these findings could not be reproduced by independent investigators192. Moreover, when experimental SLE is established in a Tlr3−/− genetic background, the course of the disease remains unchanged193. Along similar lines, the Tlr8−/− and Tlr9−/− genotypes both fail to ameliorate (and rather aggravate) disease progression in MRL/lpr mice193,194,195. This contradicts the suggestion that TLR8 underlies deregulated type I interferon secretion in experimental models of SLE196 and the finding that TLR9 is required for B cells to produce autoantibodies involved in the pathogenesis of SLE, at least in mice193,195,197. Moreover, the levels of TLR9-expressing B cells, plasma cells and monocytes are increased in patients with SLE, correlating with the overproduction of pathogenic autoantibodies198,199. Although the precise reasons underlying these apparently controversial observations remain to be elucidated, it is tempting to speculate that the elevated redundancy of TLR signalling (Box 3) may play a considerable role. In support of this hypothesis, a chemical inhibitor of the common TLR effector TBK1 (that is, compound II)200, as well as a mutation in unc-93 homologue B1 (UNC93B1), a TLR signalling regulator that compromises endosomal TLR signalling201, mediates protective effects in experimental models of SLE.
Notably, TLR7 seems to play a non-redundant role in the pathogenesis of SLE, at least in mice. MPL/lpr mice backcrossed into Tlr7−/− mice have been reported to develop SLE with significantly reduced renal disease and pathogenic autoantibodies197,202. Moreover, male BXSB mice with a Y-linked autoimmune accelerator locus (Yaa) develop spontaneous SLE-like disease due to duplication of a four-megabase gene segment containing Tlr7 transposed to the Y chromosome203. The pathogenic role of TLR7 in this latter model was demonstrated by gene dosage experiments showing that Tlr7 copy number directly correlates with disease severity in Yaa male mice204. Consistent with this, class R inhibitory ODNs mediated therapeutic effects in MRL/lpr mice downstream of mixed TLR3 and TLR9 inhibition205. Altogether, these observations suggest that TLR7 antagonists may be valuable therapeutic tools for the management of SLE. Alternating 2′-O-ribose methylation in the sense strand has been proposed as a robust chemical approach for the generation of TLR7 antagonists206. However, to the best of our knowledge, the clinical development of TLR7 antagonists for the management of SLE stands at an impasse (source http://www.clinicaltrials.gov).
Conversely, this approach has been pursued in clinical development for other autoimmune disorders. Experimental evidence based on knockout mice indicate indeed that TLR7 contributes to the cause of rheumatoid arthritis207,208. At odds with the SLE setting, however, TLR3 and TLR8 also appear to mediate non-redundant pathological functions in rheumatoid arthritis. In line with this notion, Tlr3−/− and Ticam1−/− mice are protected from experimental rheumatoid arthritis as compared to their wild-type counterparts208 Moreover, topical resiquimod or imiquimod administration has been harnessed to provoke a systemic autoimmune disorder with rheumatoid arthritis features or a psoriasis-like condition in mice, respectively67,209. Finally, the synovial lining and sublining of patients with rheumatoid arthritis express high levels of several TLRs with pathogenic potential210,211.
Consistent with a pathological role for TLRs in various autoimmune conditions, TLR inhibitors, including ODN-1411, have been shown to limit deregulated cytokine secretion in human models of rheumatoid arthritis, in vitro212, and to decelerate disease progression in mouse models of psoriasis213. Moreover, experimental autoimmune encephalomyelitis (a model of multiple sclerosis) progresses more slowly in Tlr9−/− mice than in wild-type mice214. Along similar lines, silencing regulator of G-protein signalling 1 (RGS1) suppressed endosomal TLR signalling in a rat model of collagen-induced arthritis, culminating in reduced disease progression215. These observations spurred considerable interest in the possibility to antagonize nucleic acid-sensing TLRs for therapeutic purposes, culminating in the initiation of multiple clinical trials in patients with psoriasis (source http://www.clinicaltrials.gov). In particular, IMO-8400 (a mixed TLR7, TLR8 and TLR9 antagonist) demonstrated clinical activity in a phase IIa, randomized, placebo-controlled trial in patients with moderate-to-severe plaque psoriasis216 (NCT01899729). Additional IMO-8400 testing has been done in patients with dermatomyositis (NCT02612857), as well as in individuals with non-Hodgkin lymphoma expressing a hyperactive variant of MYD88 (NCT02092909, NCT02252146, NCT02612857). Although official sources list all these latter studies as completed, only the results of NCT02252146 have been disseminated, demonstrating the safety of IMO-8400 in patients with non-Hodgkin lymphoma. IMO-3100 (a mixed TLR7 and TLR8 antagonist) was also recently tested in patients with moderate-to-severe plaque psoriasis (NCT01622348), and subcutaneous administration resulted in a mean reduction in the thickness of index lesion in the absence of major toxicity. Another mixed antagonist of TLR7 and TLR8 is currently under clinical development for autoimmune disorders (source https://www.bms.com/researchers-and-partners/in-the-pipeline.html) but no further information is available on the molecule and precise indication.
Cardiovascular disorders
Deregulated TLR signalling may also be involved in the development and progression of atherosclerosis, a possibility that began to be investigated following the correlation between a variety of pathogens (for example, Chlamydia pneumoniae, Helicobacter pylori, HCV and HIV-1) and increased risk of cardiovascular disorders217,218,219. Apoe−/− mice (a genetic model of atherosclerosis) experience a reduction in atherosclerotic lesions coupled with decreased macrophage infiltration when backcrossed on Myd88−/− mice220,221. Although such a protective effect was initially attributed to the inhibition of TLR2 and TLR4 signalling (which is not initiated by nucleic acids)222,223, more recent genetic data suggest that TLR9 plays a similar detrimental role224. Consistent with this, the mixed TLR7 and TLR9 antagonist ODN-2088 ameliorated neointimal thickening and decreased macrophage infiltration in a mouse model of postinterventional remodelling225, an effect that appears to depend on TLR9 inhibition226. Indeed Tlr7−/−Apoe−/− mice exhibit accelerated disease progression as compared with their Apoe−/− counterparts226, and Tlr9−/− mice are protected against experimental cardiomyopathy driven by mitochondrial DNA spillage227. Besides identifying TLR9 as a target for the development of antagonists with cardioprotective effects, these observations raise the interesting possibility that TLR7 agonists, rather than antagonists, may be useful for the management of atherosclerosis. To the best of our knowledge, no antagonist of nucleic acid-sensing TLRs is currently under clinical development for the treatment of cardiovascular conditions (source http://www.clinicaltrials.gov).
Neurodegenerative disorders
Suggesting the involvement of TLRs in the pathogenesis of neurodegenerative disorders, Myd88−/− mice exhibit decreased amyloid accumulation in the central nervous system as compared with their wild-type littermates in a model of Alzheimer disease228, and deregulated TLR7 signalling has been shown to cause an Alzheimer disease-like neurodegenerative disorder in mice229. Similarly, neuronal degeneration is aggravated by CpG-rich ODNs and limited by deletion of Tlr9 in a toxin-induced model of Parkinson disease 230. In addition, TLR9 levels are elevated in the striatum of post-mortem parkinsonian brains (as compared with age-matched post-mortem brains)231 Conversely, TLR9 activation by CpG-rich ODNs mediated beneficial effects on amyloid accumulation and cognitive decline in a transgenic model of Alzheimer disease232. The reasons underlying this apparent paradox remain unknown but may be related to an intrinsic difference in the inflammatory component of Parkinson disease and Alzheimer disease. Irrespective of this conundrum, the TLR9 antagonist COV08-0064 (also known as MP-3964) limited neurodegeneration in mice exposed to the Parkinson disease-inducing toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine230, suggesting that TLR9 may be a target for the development of neuroprotective drugs. This therapeutic paradigm has not yet entered clinical development (source http://www.clinicaltrials.gov).
Taken together, these observations suggest that despite a large body of preclinical evidence in support of an aetiological role for multiple TLRs in several human diseases, the development of TLR antagonists proceeds at a slow pace.
RLR and cGAS–STING antagonists
Recently, pharmaceutical companies have begun to explore therapeutic RLR and cGAS–STING antagonists233, reflecting the aetiological contribution of deregulated NAS signalling to multiple disorders. For instance, ADAR — the main endogenous inhibitor of MDA5 signalling — is required for embryonic development and adult life owing to its ability to prevent systemic autoimmune reactions with lethal consequences and to favour the establishment of thymic tolerance234,235. Consistent with this notion, ADAR mutations have been associated with a variety of autoimmune conditions potentially leading to immunodeficiency, including SLE236, Evans syndrome237 and Aicardi–Goutières syndrome238, three different settings in which pharmacological inhibitors of MDA5 may mediate therapeutic effects.
Along similar lines, mutations in the three-prime repair exonuclease 1 gene (TREX1), which codes for the main endogenous inhibitor of cGAS–STING signalling159,239, have been associated with multiple autoimmune disorders, including some SLE variants240, and Aicardi–Goutières syndrome239. Moreover, deregulated cGAS–STING signalling has been involved in the pathogenesis of a variety of inflammatory conditions in preclinical models, including age-related macular degeneration241, myocardial infarction242, sepsis243, non-alcoholic steatohepatitis244,245, inflammation-driven carcinogenesis246 and the Sjögren syndrome247. Altogether, these observations identify multiple diseases that would benefit from the development of cGAS or STING inhibitors. Only a few such compounds have been synthesized so far, including (1) a series of small molecules (including C-176 and C-178) that display robust covalent binding to STING and therapeutic activity in Trex1−/− mice248, (2) astin C, a cyclopeptide isolated from the medicinal plant Aster tataricus that exhibits anti-inflammatory activity in both mouse Trex1−/− and human TREX1−/− models and sensitizes mice to HSV-1 infection249 and (3) X6, a small cGAS inhibitor that was superior to the antimalarial drug hydroxychloroquine in the attenuation of disease symptoms in Trex1−/− mice250. Of note, recent data suggest that aspirin also acts as a bona fide cGAS inhibitor251. This latter observation not only adds another entry to the ever-increasing list of aspirin targets but also raises the generally underappreciated possibility that common drugs may modulate NASs via unclear mechanisms, and that such pharmacological activities deserve attentive scrutiny.
In summary, although the development of RLR and cGAS–STING antagonists has just started, the signal transduction cascades emanating from these NASs stand out as particularly promising targets for the development of novel anti-inflammatory agents.
Persisting obstacles
Despite intensive efforts from numerous pharmaceutical companies, very few pharmacological NAS modulators are available for use in humans, and the development of many others was discontinued at preclinical or clinical stages, as discussed earlier. A variety of drug-related, NAS-related, disease-related and host-related obstacles can be invoked to explain the failure of several NAS modulators to enter the clinic. All these factors should be carefully taken into consideration for the successful development of novel NAS modulators for clinical use (Fig. 2).
To develop novel pharmacological modulators of nucleic acid sensors (NASs) for clinical use, it will be critical to identify in advance (1) specific therapeutic paradigms in which alterations of NAS signalling provide an aetiological contribution to disease, (2) patient subsets among whom such defects have a major impact on disease progression, (3) cell populations in which deregulated NAS signalling drives pathogenesis (diseased cells, which do not necessarily overlap with cell populations normally associated with the clinical condition) and (4) the precise nature of NAS signalling alterations and how they impact on the biology of diseased cells as well as on the biology of neighbouring cells not manifesting NAS defects (bystander cells). This may allow the identification (ID) of a precise target for pharmacological interventions, which should be interrogated for (1) single-nucleotide polymorphisms (SNPs) of biological relevance (in both the target itself and in downstream signal transducers or effectors), (2) crosstalk/redundancy with other NASs, (3) sensitivity to standard-of-care (SOC) therapy and (4) influence of sex on biological outcome. This integrated analysis may allow the identification of a drug candidate, which will have to be developed while (1) co-developing a delivery platform as specific for diseased cells as possible, (2) considering potential biological effects from the drug vehicle, (3) identifying a suitable delivery route and (4) analysing drug–drug interactions, especially if SOC therapies influence the biological functions of the drug target, its signal transducers or downstream effectors. Finally, the ability of the candidate drug to normalize NAS signalling in diseased cells, and hence restore normal biological functions in diseased or bystander cells, will have to be assessed. In the absence of efficacy, the target, candidate drug or delivery platforms will have to be reconsidered. Conversely, a clinical response potentially linked to prolonged patient survival may become evident. In the absence of either or both, the complete therapeutic paradigm or patient selection should be entirely re-evaluated. PD, pharmacodynamics; PK, pharmacokinetics.
Drug-related obstacles
Some NAS agonists are second messengers or short-lived nucleic acid species, such as cGAMP (the natural STING ligand, which is rapidly degraded by ENPP1)151 and 5′-pppRNA (the natural RIG-I ligand, which is particularly unstable)134. As a result, the use of natural NAS ligands or very similar synthetic compounds is associated with suboptimal pharmacological efficacy owing to their limited stability in vivo134,180. However, this issue can be resolved by the development of stabilized molecules, such as ENPP1-insensitive cGAMP derivatives180, SLR molecules134 and compound 3 (ref.187).
Similarly, the natural ligands of cytosolic NASs (that is, RLRs and the cGAS–STING system) are unable to cross the plasma membrane owing to their electrical charge134, which further complicates their use. Although multiple transfection agents are available to circumvent this issue in vitro, only a few of these are compatible with in vivo applications252, and even fewer have been tested in the clinic, including the linear polyethylenimine derivative JetPEI (source http://www.clinicaltrials.gov). These issues call for the development of small RLR and cGAS–STING modulators that efficiently penetrate the plasma membrane. Recent data demonstrating that aspirin can act as a cGAS inhibitor251 suggest that acetylation may be harnessed for the development of effective cGAS inhibitors. Furthermore, these data support the potential of drug repurposing approaches to uncover unsuspected modulators of cytosolic NASs with desirable physicochemical properties. As an alternative, efforts should be dedicated towards the development of efficient transfection reagents that are fully compatible with clinical applications. This latter approach may also be helpful for targeting NAS modulators to specific disease sites or cell populations. Indeed, multiple NAS agonists can be delivered only locally (for example, intratumourally or intradermally), reflecting a high potential for on-target toxicity67 (see later). In support of this concept, phospholipid micelles loaded with magnetic nanoparticles have been successfully used to promote the accumulation of a cancer vaccine with poly(I:C) and imiquimod as adjuvants in experimental melanomas and tumour-draining lymph nodes in mice, resulting in therapeutic effects that could be further enhanced by PD-1 blockage253.
Finally, it will be important to carefully consider the immunomodulatory effects of excipients, which (at least theoretically) may aggravate or compromise the effects of NAS modulators. As a notable example, 5% imiquimod cream (in one of the FDA-approved formulations) has been shown to mediate both TLR7-dependent and TLR7-independent immunostimulatory effects, part of which originated from the vehicle59. This latter observation implies that higher doses of imiquimod may have been required to achieve therapeutic effects in clinical settings if another excipient had been used in the drug formulation, potentially increasing TLR7-dependent toxic effects.
NAS-related obstacles
NASs are generally expressed by a wide variety of cell types, including immune cells, epithelial cells and endothelial cells, as well as (at least some) cancer cells, reflecting their key role in the control of pathogenic infections23. Such an expression pattern has been harnessed for the development of therapeutic paradigms based on NAS modulators, such as the use of TLR3 agonists for the induction of cancer cell apoptosis254. However, the same pattern poses specificity issues linked to on-target toxicity at non-diseased sites, including some cases of moderate cytokine release syndrome observed on intranasal administration of an experimental TLR7 agonist255. Moreover, the detection of potentially pathogenic nucleic acids is such a central biological process that several of the underlying molecular cascades exhibit a considerable degree of functional redundancy and crosstalk3 (Fig. 3). In line with this notion, although correlative data point to the implication of specific NASs in the pathogenesis of some inflammatory conditions, pharmacological or genetic inhibition of the NAS in question fails to mediate robust therapeutic effects, such as in the case of TLR3 and SLE191,193 (see above). This issue has both experimental and therapeutic implications. On the one hand, the elevated redundancy of the system (that is, the ability of multiple NASs to detect similar nucleic acids) implies that conclusively excluding the implication of nucleic acid sensing from the pathogenesis of a specific condition may require the simultaneous inactivation of several NASs, which may not always be feasible experimentally. On the other hand, the intimate crosstalk between signalling cascades elicited by multiple NASs implies that efforts to develop NAS antagonists may have to be refocused on targets that are shared by multiple NAS-driven pathways, such as MYD88, TBK1 or IRF3, or their shared biological effectors (for example, type I interferon, IL-6). Consistent with this notion, numerous agents that neutralize NAS-related cytokines, such as the IL-6 blocker tocilizumab and the TNF blockers etanercept and infliximab, are currently available for clinical use188. Conversely, a phase III trial testing anifrolumab, a monoclonal antibody that inhibits type I interferon signalling, in patients with SLE (NCT02446912) failed to meet the primary end points256, hence stalling the development of interferon blockers for this specific indication. The precise reasons underlying these unexpected and disappointing results remain to be elucidated. That said, it is tempting to link them to the elevated number of potentially pathogenic cytokines secreted downstream of NAS signalling.
Mammalian nucleic acid sensors (NASs) activate signalling pathways that exhibit considerable degrees of redundancy (that is, multiple NASs can be activated by the same molecular species) and crosstalk, not only as they involve shared signal transducers such as mitogen-activated protein kinase (MAPK), IκB kinase (IKK), NF-κB-inducing kinase (NIK) and TANK-binding kinase 1 (TBK1) but also as they activate transcriptional programmes that (at least in some settings) culminate in the transactivation of shared gene sets (not shown). This implies that currently available pharmacological NAS agonists (in red) and antagonists (in blue) may exhibit poor biological specificity (agonists),or limited biological efficacy (antagonists) (Boxes 2,3,5). 5′-pppRNA, 5′-triphosphate RNA; ABZI, amidobenzimidazole; AP-1, activator protein 1; CDN, cyclic dinucleotide; cGAS, cyclic GMP–AMP synthase; CMA, 10-carboxymethyl-9-acridanone; CREB1, cyclic AMP-responsive element-binding protein 1; DMXAA, 5,6-dimethylxanthone-4-acetic acid; IMO, immunomodulatory oligonucleotide; INH-ODN, inhibitory oligodeoxynucleotide; IRF, interferon regulatory factor; MDA5, melanoma differentiation associated protein 5; NAB2, nucleic acid band 2; NF-κB, nuclear factor-κB; RIG-I, retinoic acid-inducible gene I protein; SLR, short stem–loop RNA; STING, stimulator of interferon genes; TLR, Toll-like receptor.
The existence of NAS-coding gene polymorphisms also complicates the development of pharmacological NAS modulators. At least for some NASs, not all polymorphic variants are equally sensitive to a given drug or equally proficient at activating downstream signalling cascades. Thus, the five variants of human STING exhibit differential sensitivity to canonical CDNs181, and a large number of polymorphisms affecting nucleic acid-sensing TLRs or their signal transducers have been associated with differential functionality257,258. Further complicating the situation, various polymorphisms have also been identified in the promoter regions of NAS-coding genes259. Altogether, these latter observations point to a great degree of interindividual variability in NAS responsiveness to natural and pharmacological modulators. Developing drugs that have comparable activity on the most common NAS variants may be feasible, at least in some settings183. Conversely, genetically testing patients for polymorphic variants in NASs, their transducers and effectors may be challenging, especially considering that the functional impact of multiple polymorphisms remains to be elucidated. An alternative approach may consist of testing expected biological outcome (for example, cytokine secretion) on patient samples (for example, circulating monocytes, freshly isolated cancer biopsy specimens). However, standardizing this approach to generate a predictive biomarker for response also seems difficult.
Finally, we believe that the development of NAS modulators has been hampered by the lack of precise knowledge of the kinetics of NAS signalling, as well as by intrinsic biological differences between human NASs and their murine counterparts. Thus, in some settings, such as the tumour microenvironment, acute and robust secretion of NAS-related cytokines (for example, type I interferon) is beneficial and supports the generation of antitumour immunity, while the establishment of a mild, chronic response mediates detrimental effects3. Moreover, multiple agents that drive robust TLR signalling in human cells fail to do so in mouse systems260. Additional investigation is required to clarify the ideal kinetics of NAS modulation in each disease and to determine the suitability of murine platforms for preclinical development in this setting. Of note, recently uncovered biological aspects of NAS signalling (for example, the cGAS-inhibitory activity of acetylation)251 offer a promising approach for the development of improved screening platforms for the identification of novel NAS modulators. To the best of our knowledge, however, the actual potential for discovery of such screening strategies remains to be demonstrated.
Disease-related obstacles
The development of clinical NAS modulators has often been complicated by disease-related obstacles, including the underappreciated and underinvestigated ability of standard-of-care treatments to modulate NAS signalling. These treatments include (but most likely are not limited to) aspirin, which directly inhibits cGAS251; radiation therapy, which triggers both cGAS and RIG-I signalling137,159; and various chemotherapeutics (for example, doxorubicin, cisplatin, etoposide), which initiate RIG-I and/or TLR3 signalling137,261. As NAS modulators are generally administered in combination with other drugs, the development of safe and effective therapeutic regimens calls for the careful evaluation of the effects of such drugs on NAS signalling. An additional level of complexity is provided by the fact that, at least in some disease settings, including infectious and malignant disorders, the aetiological agent (that is, pathogens, cancer cells) is naturally provided with or acquires mechanisms for evading NAS signalling141,262. This implies that some NAS modulators may be unable to mediate the expected therapeutic effects, depending on the specific molecular alteration(s) involved in such an escape mechanism. For instance, a fraction of patients with breast cancer exhibit reduced IRF7 levels (linked to poor disease outcome)263, suggesting that these tumours may be poorly sensitive to activation of NASs that culminates in IRF7-dependent transcription (Boxes 2,3,5). Finally, while dysregulated NAS signalling has long been associated with infectious, autoimmune and malignant conditions, the notion that NASs may be implicated in the pathogenesis of other conditions has just begun to emerge. Thus, excessive type I interferon production downstream of cGAS–STING signalling play a key role in the cardiac maladaptation to myocardial infarction, suggesting that cGAS or STING antagonists may mediate therapeutic effects in this setting242. This possibility, however, remains to be experimentally validated. Similar considerations apply to RIG-I and cerebral stroke264 and (we suspect) many other disorders with a hitherto poorly characterized inflammatory component. Elucidating the precise contribution of NAS signalling to human disease may foster the development of clinically useful NAS modulators.
Host-related obstacles
An underappreciation of multiple host factors may have been an obstacle in the development of NAS modulators, as has been the case for other classes of compounds, such as autophagy modulators265. One such factor is represented by variants of genes that cannot be directly linked to the molecular machinery for nucleic acid sensing. For instance, specific splicing variants of WDFY family member 4 (WDFY4), a protein mostly known for its role in antigen cross-presentation266, have been reported to boost signalling by multiple NASs, including TLR3, TLR9 and MDA5, potentially explaining the impact of WDFY4 polymorphisms on susceptibility to SLE267 and dermatomyositis268. The existence of polymorphic variants in (at least theoretically) NAS-unrelated genes that influence NAS signalling adds yet another layer of complexity to the development of predictive biomarkers based on genetic profile. Sex has also been suggested to influence NAS signalling, via poorly characterized mechanisms. For instance, in a rat model of spontaneous hypertension, the difference in disease severity between males and females was attributed to differential levels of circulating mitochondrial DNA, resulting in exacerbated TLR9 signalling in the former269. Similarly, sex hormones have been proposed to affect the physiopathology of non-alcoholic steatohepatitis by altering MYD88 expression levels270. Although increasing attention is being given to the elucidation of sex-related differences in multiple biological processes, additional work is required to precisely characterize the impact of sex on NAS signalling.
Concluding remarks
Despite a large body of preclinical and clinical evidence demonstrating that NASs are promising targets for the development of novel pharmacological agents (Table 1), this therapeutic paradigm remains largely unrealized, at least in part due to the elevated degree of redundancy and molecular crosstalk that characterize not only NASs and their signal transduction cascades but also their ultimate biological effectors (that is, cytokines), their receptors and the molecular pathways they activate. Thus, numerous obstacles need to be considered for the development of clinically useful NAS modulators (Fig. 2). Initial efforts were focused on agonistic (over antagonistic) molecules, largely reflecting early findings on the beneficial role of NAS signalling in the host response to viral infection. Conversely, NAS antagonists have attracted attention from pharmaceutical companies only recently233, following the realization that the pathogenesis of multiple human disorders besides autoimmune and inflammatory conditions involves deregulated NAS signalling242. These observations explain, at least in part, the imbalance in the number of NAS agonists and antagonists under development. Over the past two decades, the development of agonists has been considerably refocused from antiviral to anticancer applications. We surmise that such a shift originated not only from the discovery of highly efficacious therapies that allow the lifelong control of previously deadly viruses (for example, HIV-1)271 but also from the concomitant explosion of immunotherapy as a new approach to the clinical management of cancer71. In this setting (that is, cancer immunotherapy), current efforts focus on using NAS agonists as immunological adjuvants (as opposed to stand-alone therapeutic agents), as a large body of preclinical and clinical literature demonstrate that the robust immunosuppressive circuitries established by progressing tumours most often cannot be circumvented by single interventions71.
One avenue that must be pursued to foster the development of NAS modulators with clinical applications relates to a large number of proteins that de facto interact with potentially pathogenic nucleic acids but have been relatively poorly investigated as therapeutic targets, including (but not limited to) (1) nucleases such as TREX1 (ref.159), RNase H272 and RNase L273, (2) polymerases such as RNA polymerase III (ref.274), (3) components of the DNA repair machinery such as DNA-dependent protein kinase (DNA-PK)275, breast cancer type 1 susceptibility protein (BRCA1)276, double-strand break repair protein MRE11 (ref.277), and double-strand break repair protein RAD50 (ref.277), and (4) additional DNA-binding proteins involved in innate immunity, such as DEAD-box helicase 41 (DDX41)278, absent in melanoma 2 (AIM2)279, interferon-γ-inducible protein 16 (IFI16)280, PKR147 and polyglutamine binding protein 1 (PQBP1)281. In particular, it will be important to determine the pathophysiological role of each of these proteins in various disease settings and understand whether they are amenable to therapeutic targeting. Moreover, it will be interesting to investigate the possibility of targeting common NAS signal transducers, such as MYD88, TBK1 and the IκB kinase complex, as a potential means to circumvent the elevated degree of redundancy and crosstalk of the system. Of note, it has recently been demonstrated that supramolecular platforms commonly used to drive cytokine secretion in response to nucleic acid sensing, such as the myddosome (Box 3), can be engineered to mediate user-defined biological outcomes282. These findings suggest that nucleic acid sensing involves a large degree of modularity that may be harnessed for therapeutic applications. This possibility awaits urgent experimental verification.
In summary, multiple components of the molecular machinery for nucleic acid sensing continue to attract attention as potential drug targets, although the clinical development of multiple NAS modulators has been discontinued. We surmise that the identification of pharmacological NAS modulators with increased stability and hence that are amenable to systemic delivery, combined with the acquisition of precise mechanistic knowledge of the role of NASs in healthy and diseased tissues, will be instrumental for the successful clinical translation of this therapeutic paradigm. Optimized screening strategies amenable to high-throughput applications, such as the luminescence-based approach that was recently harnessed for the identification of novel cGAS inhibitors283, will be essential in this context, as they will provide various molecular scaffolds for targeted medicinal chemistry. Additional work is required to harness the full therapeutic potential of NAS modulators.
References
Jackson, S. A. et al. CRISPR-Cas: adapting to change. Science 356, eaal5056 (2017).
Cao, X. Self-regulation and cross-regulation of pattern-recognition receptor signalling in health and disease. Nat. Rev. Immunol. 16, 35–50 (2016).
Vanpouille-Box, C., Demaria, S., Formenti, S. C. & Galluzzi, L. Cytosolic DNA sensing in organismal tumor control. Cancer Cell 34, 361–378 (2018).
Schlee, M. & Hartmann, G. Discriminating self from non-self in nucleic acid sensing. Nat. Rev. Immunol. 16, 566–580 (2016).
Kieser, K. J. & Kagan, J. C. Multi-receptor detection of individual bacterial products by the innate immune system. Nat. Rev. Immunol. 17, 376–390 (2017).
Matzinger, P. Tolerance, danger, and the extended family. Annu. Rev. Immunol. 12, 991–1045 (1994).
Roers, A., Hiller, B. & Hornung, V. Recognition of endogenous nucleic acids by the innate immune system. Immunity 44, 739–754 (2016).
Chow, K. T., Gale, M., Jr. & Loo, Y. M. RIG-I and Other RNA sensors in antiviral immunity. Annu. Rev. Immunol. 36, 667–694 (2018).
Ablasser, A. & Chen, Z. J. cGAS in action: expanding roles in immunity and inflammation. Science 363, eaat8657 (2019).
Lemaitre, B., Nicolas, E., Michaut, L., Reichhart, J. M. & Hoffmann, J. A. The dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in drosophila adults. Cell 86, 973–983 (1996).
Hoffmann, J. A. The immune response of Drosophila. Nature 426, 33–38 (2003).
Junt, T. & Barchet, W. Translating nucleic acid-sensing pathways into therapies. Nat. Rev. Immunol. 15, 529–544 (2015).
Zuany-Amorim, C., Hastewell, J. & Walker, C. Toll-like receptors as potential therapeutic targets for multiple diseases. Nat. Rev. Drug Discov. 1, 797–807 (2002).
Smith, M. et al. Trial watch: Toll-like receptor agonists in cancer immunotherapy. Oncoimmunology 7, e1526250 (2018).
Campbell, J. D. Development of the CpG adjuvant 1018: a case study. Methods Mol. Biol. 1494, 15–27 (2017).
Redelman-Sidi, G., Glickman, M. S. & Bochner, B. H. The mechanism of action of BCG therapy for bladder cancer–a current perspective. Nat. Rev. Urol. 11, 153–162 (2014).
Hancock, R. E., Nijnik, A. & Philpott, D. J. Modulating immunity as a therapy for bacterial infections. Nat. Rev. Microbiol. 10, 243–254 (2012).
Vijayan, D., Young, A., Teng, M. W. L. & Smyth, M. J. Targeting immunosuppressive adenosine in cancer. Nat. Rev. Cancer 17, 709–724 (2017).
Savva, A. & Roger, T. Targeting toll-like receptors: promising therapeutic strategies for the management of sepsis-associated pathology and infectious diseases. Front. Immunol. 4, 387 (2013).
Yu, M. & Levine, S. J. Toll-like receptor, RIG-I-like receptors and the NLRP3 inflammasome: key modulators of innate immune responses to double-stranded RNA viruses. Cytokine Growth Factor Rev. 22, 63–72 (2011).
Yamamoto, M. et al. Cutting edge: a novel Toll/IL-1 receptor domain-containing adapter that preferentially activates the IFN-beta promoter in the Toll-like receptor signaling. J. Immunol. 169, 6668–6672 (2002).
Matsumoto, M., Oshiumi, H. & Seya, T. Antiviral responses induced by the TLR3 pathway. Rev. Med. Virol. 21, 67–77 (2011).
Hennessy, E. J., Parker, A. E. & O’Neill, L. A. Targeting Toll-like receptors: emerging therapeutics? Nat. Rev. Drug Discov. 9, 293–307 (2010).
Lim, H. K. et al. Severe influenza pneumonitis in children with inherited TLR3 deficiency. J. Exp. Med. 216, 2038–2056 (2019).
Field, A. K., Tytell, A. A., Lampson, G. P. & Hilleman, M. R. Inducers of interferon and host resistance. II. Multistranded synthetic polynucleotide complexes. Proc. Natl Acad. Sci. USA 58, 1004–1010 (1967).
Gresser, I., Maury, C., Bandu, M. T., Tovey, M. & Maunoury, M. T. Role of endogenous interferon in the anti-tumor effect of poly I-C and statolon as demonstrated by the use of anti-mouse interferon serum. Int. J. Cancer 21, 72–77 (1978).
Schulz, O. et al. Toll-like receptor 3 promotes cross-priming to virus-infected cells. Nature 433, 887–892 (2005).
Cheng, L. et al. Human innate responses and adjuvant activity of TLR ligands in vivo in mice reconstituted with a human immune system. Vaccine 35, 6143–6153 (2017).
Robinson, R. A. et al. A phase I-II trial of multiple-dose polyriboinosic-polyribocytidylic acid in patieonts with leukemia or solid tumors. J. Natl Cancer Inst. 57, 599–602 (1976).
Levy, H. B. et al. A modified polyriboinosinic-polyribocytidylic acid complex that induces interferon in primates. J. Infect. Dis. 132, 434–439 (1975).
Levine, A. S., Sivulich, M., Wiernik, P. H. & Levy, H. B. Initial clinical trials in cancer patients of polyriboinosinic-polyribocytidylic acid stabilized with poly-L-lysine, in carboxymethylcellulose [poly(ICLC)], a highly effective interferon inducer. Cancer Res. 39, 1645–1650 (1979).
Nakamura, O., Shitara, N., Matsutani, M., Takakura, K. & Machida, H. Phase I-II trials of poly(ICLC) in malignant brain tumor patients. J. Interferon Res. 2, 1–4 (1982).
Hammerich, L. et al. Systemic clinical tumor regressions and potentiation of PD1 blockade with in situ vaccination. Nat. Med. 25, 814–824 (2019).
Lampkin, B. C., Levine, A. S., Levy, H., Krivit, W. & Hammond, D. Phase II trial of a complex polyriboinosinic-polyribocytidylic acid with poly-L-lysine and carboxymethyl cellulose in the treatment of children with acute leukemia and neuroblastoma: a report from the children’s cancer study group. Cancer Res. 45, 5904–5909 (1985).
Dillon, P. M. et al. A pilot study of the immunogenicity of a 9-peptide breast cancer vaccine plus poly-ICLC in early stage breast cancer. J. Immunother. Cancer 5, 92 (2017).
Mehrotra, S. et al. Vaccination with poly(IC:LC) and peptide-pulsed autologous dendritic cells in patients with pancreatic cancer. J. Hematol. Oncol. 10, 82 (2017).
Rodriguez-Ruiz, M. E. et al. Combined immunotherapy encompassing intratumoral poly-ICLC, dendritic-cell vaccination and radiotherapy in advanced cancer patients. Ann. Oncol. 29, 1312–1319 (2018).
Okada, H. et al. Induction of robust type-I CD8+ T-cell responses in WHO grade 2 low-grade glioma patients receiving peptide-based vaccines in combination with poly-ICLC. Clin. Cancer Res. 21, 286–294 (2015).
Pollack, I. F. et al. Antigen-specific immune responses and clinical outcome after vaccination with glioma-associated antigen peptides and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in children with newly diagnosed malignant brainstem and nonbrainstem gliomas. J. Clin. Oncol. 32, 2050–2058 (2014).
Saxena, M. et al. Poly-ICLC, a TLR3 agonist, induces transient innate immune responses in patients with treated HIV-infection: a randomized double-blinded placebo controlled trial. Front. Immunol. 10, 725 (2019).
Hubbell, H. R., Kvalnes-Krick, K., Carter, W. A. & Strayer, D. R. Antiproliferative and immunomodulatory actions of beta-interferon and double-stranded RNA, individually and in combination, on human bladder tumor xenografts in nude mice. Cancer Res. 45, 2481–2486 (1985).
Carter, W. A. et al. Clinical, immunological, and virological effects of ampligen, a mismatched double-stranded RNA, in patients with AIDS or AIDS-related complex. Lancet 1, 1286–1292 (1987).
Mitchell, W. M., Montefiori, D. C., Robinson, W. E. Jr., Strayer, D. R. & Carter, W. A. Mismatched double-stranded RNA (Ampligen) reduces concentration of zidovudine (azidothymidine) required for in-vitro inhibition of human immunodeficiency virus. Lancet 1, 890–892 (1987).
Armstrong, J. A. et al. A phase I study of Ampligen in human immunodeficiency virus-infected subjects. J. Infect. Dis. 166, 717–722 (1992).
Thompson, K. A. et al. Results of a double-blind placebo-controlled study of the double-stranded RNA drug polyI:polyC12U in the treatment of HIV infection. Eur. J. Clin. Microbiol. Infect. Dis. 15, 580–587 (1996).
Strayer, D. R. et al. A controlled clinical trial with a specifically configured RNA drug, poly(I).poly(C12U), in chronic fatigue syndrome. Clin. Infect. Dis. 18, S88–S95 (1994).
Strayer, D. R. et al. A double-blind, placebo-controlled, randomized, clinical trial of the TLR-3 agonist rintatolimod in severe cases of chronic fatigue syndrome. PLOS ONE 7, e31334 (2012).
Navabi, H. et al. A clinical grade poly I:C-analogue (Ampligen) promotes optimal DC maturation and Th1-type T cell responses of healthy donors and cancer patients in vitro. Vaccine 27, 107–115 (2009).
Takeda, Y. et al. A TLR3-specific adjuvant relieves innate resistance to PD-L1 blockade without cytokine toxicity in tumor vaccine immunotherapy. Cell Rep. 19, 1874–1887 (2017).
Kawai, T. & Akira, S. TLR signaling. Cell Death Differ. 13, 816–825 (2006).
Ishii, N., Funami, K., Tatematsu, M., Seya, T. & Matsumoto, M. Endosomal localization of TLR8 confers distinctive proteolytic processing on human myeloid cells. J. Immunol. 193, 5118–5128 (2014).
Diebold, S. S., Kaisho, T., Hemmi, H., Akira, S. & Reis e Sousa, C. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science 303, 1529–1531 (2004).
Heil, F. et al. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science 303, 1526–1529 (2004).
Hemmi, H. et al. Small anti-viral compounds activate immune cells via the TLR7 MyD88-dependent signaling pathway. Nat. Immunol. 3, 196–200 (2002).
Thomas, A. et al. Investigating Toll-like receptor agonists for potential to treat hepatitis C virus infection. Antimicrob. Agents Chemother. 51, 2969–2978 (2007).
Gerster, J. F. et al. Synthesis and structure-activity-relationships of 1H-imidazo[4,5-c]quinolines that induce interferon production. J. Med. Chem. 48, 3481–3491 (2005).
Harrison, C. J., Jenski, L., Voychehovski, T. & Bernstein, D. I. Modification of immunological responses and clinical disease during topical R-837 treatment of genital HSV-2 infection. Antivir. Res. 10, 209–223 (1988).
Hanna, E., Abadi, R. & Abbas, O. Imiquimod in dermatology: an overview. Int. J. Dermatol. 55, 831–844 (2016).
Walter, A. et al. Aldara activates TLR7-independent immune defence. Nat. Commun. 4, 1560 (2013). This study reveals that the immunostimulatory effects of Aldara depend on both TLR7-dependent pathways (elicited by imiquimod) and TLR7-independent mechanisms (elicited by the cream vehicle).
Nian, H. et al. R-848 triggers the expression of TLR7/8 and suppresses HIV replication in monocytes. BMC Infect. Dis. 12, 5 (2012).
Mark, K. E. et al. Topical resiquimod 0.01% gel decreases herpes simplex virus type 2 genital shedding: a randomized, controlled trial. J. Infect. Dis. 195, 1324–1331 (2007).
Pockros, P. J. et al. Oral resiquimod in chronic HCV infection: safety and efficacy in 2 placebo-controlled, double-blind phase IIa studies. J. Hepatol. 47, 174–182 (2007).
Sidky, Y. A. et al. Inhibition of murine tumor growth by an interferon-inducing imidazoquinolinamine. Cancer Res. 52, 3528–3533 (1992).
Dewan, M. Z. et al. Synergy of topical toll-like receptor 7 agonist with radiation and low-dose cyclophosphamide in a mouse model of cutaneous breast cancer. Clin. Cancer Res. 18, 6668–6678 (2012).
Han, J. H. et al. TLR7 expression is decreased during tumour progression in transgenic adenocarcinoma of mouse prostate mice and its activation inhibits growth of prostate cancer cells. Am J Reprod. Immunol. 70, 317–326 (2013).
Cai, Y. et al. Pivotal role of dermal IL-17-producing gammadelta T cells in skin inflammation. Immunity 35, 596–610 (2011).
van der Fits, L. et al. Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis. J. Immunol. 182, 5836–5845 (2009).
Salazar, L. G. et al. Topical imiquimod plus nab-paclitaxel for breast cancer cutaneous metastases: a phase 2 clinical trial. JAMA Oncol. 3, 969–973 (2017).
Hurt, C. N. et al. Recurrence of vulval intraepithelial neoplasia following treatment with cidofovir or imiquimod: results from a multicentre, randomised, phase II trial (RT3VIN). BJOG 125, 1171–1177 (2018).
Teulings, H. E. et al. Anti-melanoma immunity and local regression of cutaneous metastases in melanoma patients treated with monobenzone and imiquimod; a phase 2 a trial. Oncoimmunology 7, e1419113 (2018).
Galluzzi, L., Chan, T. A., Kroemer, G., Wolchok, J. D. & Lopez-Soto, A. The hallmarks of successful anticancer immunotherapy. Sci. Transl Med. 10, eaat7807 (2018).
Ito, H., Ando, T., Arioka, Y., Saito, K. & Seishima, M. Inhibition of indoleamine 2,3-dioxygenase activity enhances the anti-tumour effects of a Toll-like receptor 7 agonist in an established cancer model. Immunology 144, 621–630 (2015).
Fujimura, T. et al. Successful treatment of nivolumab-resistant multiple in-transit melanomas with ipilimumab and topical imiquimod. Case Rep. Oncol. 11, 1–5 (2018).
Rook, A. H. et al. Topical resiquimod can induce disease regression and enhance T-cell effector functions in cutaneous T-cell lymphoma. Blood 126, 1452–1461 (2015).
Sabado, R. L. et al. Resiquimod as an immunologic adjuvant for NY-ESO-1 protein vaccination in patients with high-risk melanoma. Cancer Immunol. Res. 3, 278–287 (2015).
Fahey, L. M., Raff, A. B., Da Silva, D. M. & Kast, W. M. Reversal of human papillomavirus-specific T cell immune suppression through TLR agonist treatment of Langerhans cells exposed to human papillomavirus type 16. J. Immunol. 182, 2919–2928 (2009).
Lu, H. et al. VTX-2337 is a novel TLR8 agonist that activates NK cells and augments ADCC. Clin. Cancer Res. 18, 499–509 (2012).
Stephenson, R. M. et al. TLR8 stimulation enhances cetuximab-mediated natural killer cell lysis of head and neck cancer cells and dendritic cell cross-priming of EGFR-specific CD8+ T cells. Cancer Immunol. Immunother. 62, 1347–1357 (2013).
Doener, F. et al. RNA-based adjuvant CV8102 enhances the immunogenicity of a licensed rabies vaccine in a first-in-human trial. Vaccine 37, 1819–1826 (2019). This article reports safety and activity results from the first-in-concept human trial of the RNA-based agonist CV8102 (an agonist of TLR7, TLR8 and RIG-I) administered alone or in combination with fractional doses of a rabies vaccine.
Moisan, J. et al. TLR7 ligand prevents allergen-induced airway hyperresponsiveness and eosinophilia in allergic asthma by a MYD88-dependent and MK2-independent pathway. Am. J. Physiol. Lung Cell Mol. Physiol. 290, L987–L995 (2006).
Ellis, A. K., Tsitoura, D. C., Quint, D., Powley, W. & Lee, L. A. Safety and pharmacodynamics of intranasal GSK2245035, a TLR7 agonist for allergic rhinitis: a randomized trial. Clin. Exp. Allergy 47, 1193–1203 (2017).
Lee, C. C., Avalos, A. M. & Ploegh, H. L. Accessory molecules for Toll-like receptors and their function. Nat. Rev. Immunol. 12, 168–179 (2012).
Akira, S., Takeda, K. & Kaisho, T. Toll-like receptors: critical proteins linking innate and acquired immunity. Nat. Immunol. 2, 675–680 (2001).
Hemmi, H. et al. A Toll-like receptor recognizes bacterial DNA. Nature 408, 740–745 (2000).
Xu, R. H. et al. Sequential activation of two pathogen-sensing pathways required for type I interferon expression and resistance to an acute DNA virus infection. Immunity 43, 1148–1159 (2015).
Ives, A. et al. MyD88 and TLR9 dependent immune responses mediate resistance to Leishmania guyanensis infections, irrespective of Leishmania RNA virus burden. PLOS ONE 9, e96766 (2014).
Klinman, D. M., Verthelyi, D., Takeshita, F. & Ishii, K. J. Immune recognition of foreign DNA: a cure for bioterrorism? Immunity 11, 123–129 (1999).
Kandimalla, E. R. et al. Immunomodulatory oligonucleotides containing a cytosine-phosphate-2′-deoxy-7-deazaguanosine motif as potent toll-like receptor 9 agonists. Proc. Natl Acad. Sci. USA 102, 6925–6930 (2005).
Makowska, Z. et al. Sequential induction of type I and II interferons mediates a long-lasting gene induction in the liver in response to a novel toll-like receptor 9 agonist. J. Hepatol. 58, 743–749 (2013).
Sarasin-Filipowicz, M. et al. Interferon signaling and treatment outcome in chronic hepatitis C. Proc. Natl Acad. Sci. USA 105, 7034–7039 (2008).
Olbrich, A. R. et al. Effective postexposure treatment of retrovirus-induced disease with immunostimulatory DNA containing CpG motifs. J. Virol. 76, 11397–11404 (2002).
Wang, Y. et al. The Toll-like receptor 7 (TLR7) agonist, imiquimod, and the TLR9 agonist, CpG ODN, induce antiviral cytokines and chemokines but do not prevent vaginal transmission of simian immunodeficiency virus when applied intravaginally to rhesus macaques. J. Virol. 79, 14355–14370 (2005).
Equils, O. et al. Toll-like receptor 2 (TLR2) and TLR9 signaling results in HIV-long terminal repeat trans-activation and HIV replication in HIV-1 transgenic mouse spleen cells: implications of simultaneous activation of TLRs on HIV replication. J. Immunol. 170, 5159–5164 (2003).
Sundstrom, J. B., Little, D. M., Villinger, F., Ellis, J. E. & Ansari, A. A. Signaling through Toll-like receptors triggers HIV-1 replication in latently infected mast cells. J. Immunol. 172, 4391–4401 (2004).
Brichacek, B. et al. Contrasting roles for TLR ligands in HIV-1 pathogenesis. PLOS ONE 5, e12831 (2010).
Vibholm, L. K. et al. Effects of 24 week Toll-like receptor 9 agonist treatment in HIV-1+ individuals: a single-arm, phase 1B/2A trial. AIDS 33, 1315–1325 (2019).
Krarup, A. R. et al. The TLR9 agonist MGN1703 triggers a potent type I interferon response in the sigmoid colon. Mucosal Immunol. 11, 449–461 (2018).
Cooper, C. L., Angel, J. B., Seguin, I., Davis, H. L. & Cameron, D. W. CPG 7909 adjuvant plus hepatitis B virus vaccination in HIV-infected adults achieves long-term seroprotection for up to 5 years. Clin. Infect. Dis. 46, 1310–1314 (2008).
Ito, H. et al. Induction of humoral and cellular immune response to hepatitis B virus (HBV) vaccine can be upregulated by CpG oligonucleotides complexed with dectin-1 ligand. J. Viral Hepat. 24, 155–162 (2017).
Li, J. et al. Hepatitis B surface antigen (HBsAg) and core antigen (HBcAg) combine CpG oligodeoxynucletides as a novel therapeutic vaccine for chronic hepatitis B infection. Vaccine 33, 4247–4254 (2015).
Brignole, C. et al. Therapeutic targeting of TLR9 inhibits cell growth and induces apoptosis in neuroblastoma. Cancer Res. 70, 9816–9826 (2010).
Damiano, V. et al. TLR9 agonist acts by different mechanisms synergizing with bevacizumab in sensitive and cetuximab-resistant colon cancer xenografts. Proc. Natl Acad. Sci. USA 104, 12468–12473 (2007).
Zoglmeier, C. et al. CpG blocks immunosuppression by myeloid-derived suppressor cells in tumor-bearing mice. Clin. Cancer Res. 17, 1765–1775 (2011).
Heckelsmiller, K. et al. Peritumoral CpG DNA elicits a coordinated response of CD8 T cells and innate effectors to cure established tumors in a murine colon carcinoma model. J. Immunol. 169, 3892–3899 (2002).
Houot, R. & Levy, R. T-cell modulation combined with intratumoral CpG cures lymphoma in a mouse model without the need for chemotherapy. Blood 113, 3546–3552 (2009).
Lonsdorf, A. S. et al. Intratumor CpG-oligodeoxynucleotide injection induces protective antitumor T cell immunity. J. Immunol. 171, 3941–3946 (2003).
Leonard, J. P. et al. Phase I trial of toll-like receptor 9 agonist PF-3512676 with and following rituximab in patients with recurrent indolent and aggressive non Hodgkin’s lymphoma. Clin. Cancer Res. 13, 6168–6174 (2007).
Zent, C. S. et al. Phase I clinical trial of CpG oligonucleotide 7909 (PF-03512676) in patients with previously treated chronic lymphocytic leukemia. Leuk. Lymphoma 53, 211–217 (2012).
Carpentier, A. et al. Intracerebral administration of CpG oligonucleotide for patients with recurrent glioblastoma: a phase II study. Neuro. Oncol. 12, 401–408 (2010).
Pashenkov, M. et al. Phase II trial of a toll-like receptor 9-activating oligonucleotide in patients with metastatic melanoma. J. Clin. Oncol. 24, 5716–5724 (2006).
Thompson, J. A., Kuzel, T., Drucker, B. J., Urba, W. J. & Bukowski, R. M. Safety and efficacy of PF-3512676 for the treatment of stage IV renal cell carcinoma: an open-label, multicenter phase I/II study. Clin. Genitourin. Cancer 7, E58–E65 (2009).
Weber, J. S. et al. Randomized phase 2/3 trial of CpG oligodeoxynucleotide PF-3512676 alone or with dacarbazine for patients with unresectable stage III and IV melanoma. Cancer 115, 3944–3954 (2009).
Hirsh, V. et al. Randomized phase III trial of paclitaxel/carboplatin with or without PF-3512676 (Toll-like receptor 9 agonist) as first-line treatment for advanced non-small-cell lung cancer. J. Clin. Oncol. 29, 2667–2674 (2011).
Manegold, C. et al. A phase III randomized study of gemcitabine and cisplatin with or without PF-3512676 (TLR9 agonist) as first-line treatment of advanced non-small-cell lung cancer. Ann. Oncol. 23, 72–77 (2012).
Manegold, C. et al. Randomized phase II trial of a toll-like receptor 9 agonist oligodeoxynucleotide, PF-3512676, in combination with first-line taxane plus platinum chemotherapy for advanced-stage non-small-cell lung cancer. J. Clin. Oncol. 26, 3979–3986 (2008).
Koster, B. D. et al. Local adjuvant treatment with low-dose CpG-B offers durable protection against disease recurrence in clinical stage I-II melanoma: data from two randomized phase ii trials. Clin. Cancer Res. 23, 5679–5686 (2017).
[No authors listed] Warming “cold” melanoma with TLR9 agonists. Cancer Discov. 8, 670 (2018).
Ribas, A. et al. SD-101 in Combination with pembrolizumab in advanced melanoma: results of a phase Ib, multicenter study. Cancer Discov. 8, 1250–1257 (2018).
Frank, M. J. et al. In Situ vaccination with a TLR9 agonist and local low-dose radiation induces systemic responses in untreated indolent lymphoma. Cancer Discov. 8, 1258–1269 (2018).
Rodriguez-Ruiz, M. E., Vanpouille-Box, C., Melero, I., Formenti, S. C. & Demaria, S. Immunological mechanisms responsible for radiation-induced abscopal effect. Trends Immunol. 39, 644–655 (2018).
Kline, J. N. & Krieg, A. M. Toll-like receptor 9 activation with CpG oligodeoxynucleotides for asthma therapy. Drug News Perspect. 21, 434–439 (2008).
Li-Ping Thio, C., Chuan-Ying Lai, A., Chi, P. Y., Webster, G. & Chang, Y. J. Toll-like receptor 9-dependent interferon production prevents group 2 innate lymphoid cell-driven airway hyperreactivity. J. Allergy Clin. Immunol. 144, 682–697.e9 (2019).
Reikine, S., Nguyen, J. B. & Modis, Y. Pattern recognition and signaling mechanisms of RIG-I and MDA5. Front. Immunol. 5, 342 (2014).
Yoneyama, M. et al. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat. Immunol. 5, 730–737 (2004).
Blander, J. M. A long-awaited merger of the pathways mediating host defence and programmed cell death. Nat. Rev. Immunol. 14, 601–618 (2014).
Korolowicz, K. E. et al. Antiviral efficacy and host innate immunity associated with SB 9200 treatment in the woodchuck model of chronic hepatitis B. PLOS ONE 11, e0161313 (2016).
Travassos, L. H. et al. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Nat. Immunol. 11, 55–62 (2010).
Mariathasan, S. et al. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature 440, 228–232 (2006).
Suresh, M. et al. Antiviral efficacy and host immune response induction during sequential treatment with sb 9200 followed by entecavir in woodchucks. PLOS ONE 12, e0169631 (2017).
Jones, M. et al. SB 9200, a novel agonist of innate immunity, shows potent antiviral activity against resistant HCV variants. J. Med. Virol. 89, 1620–1628 (2017).
Hornung, V. et al. 5′-Triphosphate RNA is the ligand for RIG-I. Science 314, 994–997 (2006).
Olagnier, D. et al. Inhibition of dengue and chikungunya virus infections by RIG-I-mediated type I interferon-independent stimulation of the innate antiviral response. J. Virol. 88, 4180–4194 (2014).
Goulet, M. L. et al. Systems analysis of a RIG-I agonist inducing broad spectrum inhibition of virus infectivity. PLOS Pathog. 9, e1003298 (2013).
Linehan, M. M. et al. A minimal RNA ligand for potent RIG-I activation in living mice. Sci. Adv. 4, e1701854 (2018).
Poeck, H. et al. 5′-Triphosphate-siRNA: turning gene silencing and Rig-I activation against melanoma. Nat. Med. 14, 1256–1263 (2008).
Li, D. et al. 5′-Triphosphate siRNA targeting MDR1 reverses multi-drug resistance and activates RIG-I-induced immune-stimulatory and apoptotic effects against human myeloid leukaemia cells. Leuk. Res. 58, 23–30 (2017).
Ranoa, D. R. et al. Cancer therapies activate RIG-I-like receptor pathway through endogenous non-coding RNAs. Oncotarget 7, 26496–26515 (2016).
Claudepierre, M. C. et al. Yeast virus-derived stimulator of the innate immune system augments the efficacy of virus vector-based immunotherapy. J. Virol. 88, 5242–5255 (2014).
Oberson, A. et al. NAB2 is a novel immune stimulator of MDA-5 that promotes a strong type I interferon response. Oncotarget 9, 5641–5651 (2018).
Tormo, D. et al. Targeted activation of innate immunity for therapeutic induction of autophagy and apoptosis in melanoma cells. Cancer Cell 16, 103–114 (2009).
Galluzzi, L., Buque, A., Kepp, O., Zitvogel, L. & Kroemer, G. Immunogenic cell death in cancer and infectious disease. Nat. Rev. Immunol. 17, 97–111 (2017).
Aznar, M. A. et al. Immunotherapeutic effects of intratumoral nanoplexed poly I:C. J. Immunother. Cancer 7, 116 (2019).
Lamborn, I. T. et al. Recurrent rhinovirus infections in a child with inherited MDA5 deficiency. J. Exp. Med. 214, 1949–1972 (2017). This is the first report of a homozygous missense mutation in IFIH1 in humans, resulting in increased susceptibility to viral infection.
Ishizuka, J. J. et al. Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade. Nature 565, 43–48 (2019). This study identifies in ADAR a therapeutic target for circumventing the resistance of cancer patients to ICBs, reflecting the ability of ADAR to limit the availability of dsRNA for PKR and MDA5 activation.
Donnelly, N., Gorman, A. M., Gupta, S. & Samali, A. The eIF2alpha kinases: their structures and functions. Cell Mol. Life Sci. 70, 3493–3511 (2013).
Liddicoat, B. J. et al. RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself. Science 349, 1115–1120 (2015).
Gannon, H. S. et al. Identification of ADAR1 adenosine deaminase dependency in a subset of cancer cells. Nat. Commun. 9, 5450 (2018).
Dhir, A. et al. Mitochondrial double-stranded RNA triggers antiviral signalling in humans. Nature 560, 238–242 (2018).
Probst, P. et al. A small-molecule IRF3 agonist functions as an influenza vaccine adjuvant by modulating the antiviral immune response. Vaccine 35, 1964–1971 (2017).
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 339, 786–791 (2013).
Ablasser, A. et al. cGAS produces a 2′-5′-linked cyclic dinucleotide second messenger that activates STING. Nature 498, 380–384 (2013).
Ablasser, A. et al. Cell intrinsic immunity spreads to bystander cells via the intercellular transfer of cGAMP. Nature 503, 530–534 (2013).
Gao, P. et al. Cyclic [G(2′,5′)pA(3′,5′)p] is the metazoan second messenger produced by DNA-activated cyclic GMP-AMP synthase. Cell 153, 1094–1107 (2013).
Civril, F. et al. Structural mechanism of cytosolic DNA sensing by cGAS. Nature 498, 332–337 (2013).
Gao, D. et al. Cyclic GMP-AMP synthase is an innate immune sensor of HIV and other retroviruses. Science 341, 903–906 (2013).
Li, X. D. et al. Pivotal roles of cGAS-cGAMP signaling in antiviral defense and immune adjuvant effects. Science 341, 1390–1394 (2013).
Woo, S. R. et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity 41, 830–842 (2014).
Deng, L. et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity 41, 843–852 (2014).
Vanpouille-Box, C. et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 8, 15618 (2017). This article demonstrates that the cytosolic exonuclease TREX1 is upregulated by radiation doses greater than 12–14 Gy in most cancer cells, resulting in limited availability of dsDNA for cGAS activation and hence reduced secretion of type I interferon.
Sivick, K. E. et al. Magnitude of therapeutic sting activation determines CD8+ T cell-mediated anti-tumor immunity. Cell Rep. 25, 3074–3085 e3075 (2018).
Gulen, M. F. et al. Signalling strength determines proapoptotic functions of STING. Nat. Commun. 8, 427 (2017). This article and reference 162 demonstrate that robust STING agonism mediates cytostatic and cytotoxic effects in T cells, hence being detrimental to (rather than beneficial for) anticancer immune responses.
Larkin, B. et al. Cutting edge: activation of sting in T cells induces type I IFN responses and cell death. J. Immunol. 199, 397–402 (2017).
Wu, T. Y. Strategies for designing synthetic immune agonists. Immunology 148, 315–325 (2016).
Prantner, D. et al. 5,6-Dimethylxanthenone-4-acetic acid (DMXAA) activates stimulator of interferon gene (STING)-dependent innate immune pathways and is regulated by mitochondrial membrane potential. J. Biol. Chem. 287, 39776–39788 (2012).
Kanwar, J. R., Kanwar, R. K., Pandey, S., Ching, L. M. & Krissansen, G. W. Vascular attack by 5,6-dimethylxanthenone-4-acetic acid combined with B7.1 (CD80)-mediated immunotherapy overcomes immune resistance and leads to the eradication of large tumors and multiple tumor foci. Cancer Res. 61, 1948–1956 (2001).
Perera, P. Y., Barber, S. A., Ching, L. M. & Vogel, S. N. Activation of LPS-inducible genes by the antitumor agent 5,6-dimethylxanthenone-4-acetic acid in primary murine macrophages. Dissection of signaling pathways leading to gene induction and tyrosine phosphorylation. J. Immunol. 153, 4684–4693 (1994).
Guo, F. et al. STING agonists induce an innate antiviral immune response against hepatitis B virus. Antimicrob. Agents Chemother. 59, 1273–1281 (2015).
Ceron, S., North, B. J., Taylor, S. A. & Leib, D. A. The STING agonist 5,6-dimethylxanthenone-4-acetic acid (DMXAA) stimulates an antiviral state and protects mice against herpes simplex virus-induced neurological disease. Virology 529, 23–28 (2019).
Curran, E. et al. STING Pathway activation stimulates potent immunity against acute myeloid leukemia. Cell Rep. 15, 2357–2366 (2016).
Weiss, J. M. et al. The STING agonist DMXAA triggers a cooperation between T lymphocytes and myeloid cells that leads to tumor regression. Oncoimmunology 6, e1346765 (2017).
McKeage, M. J. et al. 5,6-Dimethylxanthenone-4-acetic acid in the treatment of refractory tumors: a phase I safety study of a vascular disrupting agent. Clin. Cancer Res. 12, 1776–1784 (2006).
Pili, R. et al. Phase II study on the addition of ASA404 (vadimezan; 5,6-dimethylxanthenone-4-acetic acid) to docetaxel in CRMPC. Clin. Cancer Res. 16, 2906–2914 (2010).
Lara, P. N., Jr. et al. Randomized phase III placebo-controlled trial of carboplatin and paclitaxel with or without the vascular disrupting agent vadimezan (ASA404) in advanced non-small-cell lung cancer. J. Clin. Oncol. 29, 2965–2971 (2011).
Gao, P. et al. Structure-function analysis of STING activation by c[G(2′,5′)pA(3′,5′)p] and targeting by antiviral DMXAA. Cell 154, 748–762 (2013).
Conlon, J. et al. Mouse, but not human STING, binds and signals in response to the vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid. J. Immunol. 190, 5216–5225 (2013).
Burdette, D. L. et al. STING is a direct innate immune sensor of cyclic di-GMP. Nature 478, 515–518 (2011).
Ohkuri, T. et al. STING contributes to antiglioma immunity via triggering type I IFN signals in the tumor microenvironment. Cancer Immunol. Res. 2, 1199–1208 (2014).
Chandra, D. et al. STING ligand c-di-GMP improves cancer vaccination against metastatic breast cancer. Cancer Immunol. Res. 2, 901–910 (2014).
Yamamoto, T. et al. STING agonists activate latently infected cells and enhance SIV-specific responses ex vivo in naturally SIV controlled cynomolgus macaques. Sci. Rep. 9, 5917 (2019).
Li, L. et al. Hydrolysis of 2′3′-cGAMP by ENPP1 and design of nonhydrolyzable analogs. Nat. Chem. Biol. 10, 1043–1048 (2014).
Yi, G. et al. Single nucleotide polymorphisms of human STING can affect innate immune response to cyclic dinucleotides. PLOS ONE 8, e77846 (2013).
Rodriguez-Garcia, E. et al. TMEM173 alternative spliced isoforms modulate viral replication through the STING pathway. Immunohorizons 2, 363–376 (2018).
Corrales, L. et al. Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity. Cell Rep. 11, 1018–1030 (2015).
Ager, C. R. et al. Intratumoral STING activation with T-cell checkpoint modulation generates systemic antitumor immunity. Cancer Immunol. Res. 5, 676–684 (2017).
Fu, J. et al. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade. Sci. Transl Med. 7, 283ra252 (2015).
Tang, C. H. et al. Agonist-mediated activation of sting induces apoptosis in malignant b cells. Cancer Res. 76, 2137–2152 (2016).
Ramanjulu, J. M. et al. Design of amidobenzimidazole STING receptor agonists with systemic activity. Nature 564, 439–443 (2018). This is the first report of a STING agonist that is not a CDN, is amenable to systemic delivery and mediates robust antineoplastic effects as stand-alone therapeutic intervention.
Croft, M. & Siegel, R. M. Beyond TNF: TNF superfamily cytokines as targets for the treatment of rheumatic diseases. Nat. Rev. Rheumatol. 13, 217–233 (2017).
Galluzzi, L., Yamazaki, T. & Kroemer, G. Linking cellular stress responses to systemic homeostasis. Nat. Rev. Mol. Cell Biol. 19, 731–745 (2018).
Wang, C. M. et al. Genetic variations in Toll-like receptors (TLRs 3/7/8) are associated with systemic lupus erythematosus in a Taiwanese population. Sci. Rep. 4, 3792 (2014).
Patole, P. S. et al. Viral double-stranded RNA aggravates lupus nephritis through Toll-like receptor 3 on glomerular mesangial cells and antigen-presenting cells. J. Am. Soc. Nephrol. 16, 1326–1338 (2005).
Annable, T. et al. Using poly I:C as an adjuvant does not induce or exacerbate models of systemic lupus erythematosus. Autoimmunity 48, 29–39 (2015).
Christensen, S. R. et al. Toll-like receptor 9 controls anti-DNA autoantibody production in murine lupus. J. Exp. Med. 202, 321–331 (2005).
Tran, N. L., Manzin-Lorenzi, C. & Santiago-Raber, M. L. Toll-like receptor 8 deletion accelerates autoimmunity in a mouse model of lupus through a Toll-like receptor 7-dependent mechanism. Immunology 145, 60–70 (2015).
Nickerson, K. M. et al. TLR9 regulates TLR7- and MyD88-dependent autoantibody production and disease in a murine model of lupus. J. Immunol. 184, 1840–1848 (2010).
Vollmer, J. et al. Immune stimulation mediated by autoantigen binding sites within small nuclear RNAs involves Toll-like receptors 7 and 8. J. Exp. Med. 202, 1575–1585 (2005).
Christensen, S. R. et al. Toll-like receptor 7 and TLR9 dictate autoantibody specificity and have opposing inflammatory and regulatory roles in a murine model of lupus. Immunity 25, 417–428 (2006).
Papadimitraki, E. D. et al. Expansion of toll-like receptor 9-expressing B cells in active systemic lupus erythematosus: implications for the induction and maintenance of the autoimmune process. Arthritis Rheum. 54, 3601–3611 (2006).
Migita, K. et al. Toll-like receptor expression in lupus peripheral blood mononuclear cells. J. Rheumatol. 34, 493–500 (2007).
Hasan, M. et al. Cutting edge: inhibiting TBK1 by compound ii ameliorates autoimmune disease in mice. J. Immunol. 195, 4573–4577 (2015).
Kono, D. H. et al. Endosomal TLR signaling is required for anti-nucleic acid and rheumatoid factor autoantibodies in lupus. Proc. Natl Acad. Sci. USA 106, 12061–12066 (2009).
Berland, R. et al. Toll-like receptor 7-dependent loss of B cell tolerance in pathogenic autoantibody knockin mice. Immunity 25, 429–440 (2006).
Subramanian, S. et al. A Tlr7 translocation accelerates systemic autoimmunity in murine lupus. Proc. Natl Acad. Sci. USA 103, 9970–9975 (2006).
Deane, J. A. et al. Control of toll-like receptor 7 expression is essential to restrict autoimmunity and dendritic cell proliferation. Immunity 27, 801–810 (2007).
Lenert, P. et al. DNA-like class R inhibitory oligonucleotides (INH-ODNs) preferentially block autoantigen-induced B-cell and dendritic cell activation in vitro and autoantibody production in lupus-prone MRL-Faslpr/lpr mice in vivo. Arthritis Res. Ther. 11, R79 (2009).
Hamm, S. et al. Alternating 2′-O-ribose methylation is a universal approach for generating non-stimulatory siRNA by acting as TLR7 antagonist. Immunobiology 215, 559–569 (2010).
Alzabin, S. et al. Investigation of the role of endosomal Toll-like receptors in murine collagen-induced arthritis reveals a potential role for TLR7 in disease maintenance. Arthritis Res. Ther. 14, R142 (2012).
Duffau, P. et al. Promotion of inflammatory arthritis by interferon regulatory factor 5 in a mouse model. Arthritis Rheumatol. 67, 3146–3157 (2015).
Hasham, M. G. et al. Systemic autoimmunity induced by the TLR7/8 agonist resiquimod causes myocarditis and dilated cardiomyopathy in a new mouse model of autoimmune heart disease. Dis. Model. Mech. 10, 259–270 (2017).
Roelofs, M. F. et al. The expression of toll-like receptors 3 and 7 in rheumatoid arthritis synovium is increased and costimulation of toll-like receptors 3, 4, and 7/8 results in synergistic cytokine production by dendritic cells. Arthritis Rheum. 52, 2313–2322 (2005).
Ospelt, C. et al. Overexpression of toll-like receptors 3 and 4 in synovial tissue from patients with early rheumatoid arthritis: toll-like receptor expression in early and longstanding arthritis. Arthritis Rheum. 58, 3684–3692 (2008).
Sacre, S. et al. Oligodeoxynucleotide inhibition of Toll-like receptors 3, 7, 8, and 9 suppresses cytokine production in a human rheumatoid arthritis model. Eur. J. Immunol. 46, 772–781 (2016).
Lai, C. Y., Su, Y. W., Lin, K. I., Hsu, L. C. & Chuang, T. H. Natural modulators of endosomal toll-like receptor-mediated psoriatic skin inflammation. J. Immunol. Res. 2017, 7807313 (2017).
Prinz, M. et al. Innate immunity mediated by TLR9 modulates pathogenicity in an animal model of multiple sclerosis. J. Clin. Invest. 116, 456–464 (2006).
Hu, X. et al. RGS1 silencing inhibits the inflammatory response and angiogenesis in rheumatoid arthritis rats through the inactivation of Toll-like receptor signaling pathway. J. Cell. Physiol. 234, 20432–20442 (2019).
Balak, D. M. et al. IMO-8400, a toll-like receptor 7, 8, and 9 antagonist, demonstrates clinical activity in a phase 2a, randomized, placebo-controlled trial in patients with moderate-to-severe plaque psoriasis. Clin. Immunol. 174, 63–72 (2017).
Kearns, A., Gordon, J., Burdo, T. H. & Qin, X. HIV-1-Associated atherosclerosis: unraveling the missing link. J. Am. Coll. Cardiol. 69, 3084–3098 (2017).
Bassendine, M. F. et al. Hepatitis c virus and atherosclerosis: a legacy after virologic cure? Clin. Res. Hepatol. Gastroenterol. 41, 25–30 (2017).
Sessa, R. et al. Chlamydia pneumoniae and atherosclerosis: current state and future prospectives. Int. J. Immunopathol. Pharmacol. 22, 9–14 (2009).
Bjorkbacka, H. et al. Reduced atherosclerosis in MyD88-null mice links elevated serum cholesterol levels to activation of innate immunity signaling pathways. Nat. Med. 10, 416–421 (2004).
Michelsen, K. S. et al. Lack of Toll-like receptor 4 or myeloid differentiation factor 88 reduces atherosclerosis and alters plaque phenotype in mice deficient in apolipoprotein E. Proc. Natl Acad. Sci. USA 101, 10679–10684 (2004).
Schoneveld, A. H. et al. Toll-like receptor 2 stimulation induces intimal hyperplasia and atherosclerotic lesion development. Cardiovasc. Res. 66, 162–169 (2005).
Vink, A. et al. In vivo evidence for a role of toll-like receptor 4 in the development of intimal lesions. Circulation 106, 1985–1990 (2002).
Ma, C. et al. Toll-like receptor 9 inactivation alleviated atherosclerotic progression and inhibited macrophage polarized to M1 phenotype in ApoE−/− mice. Dis. Markers 2015, 909572 (2015).
Karper, J. C. et al. Blocking toll-like receptors 7 and 9 reduces postinterventional remodeling via reduced macrophage activation, foam cell formation, and migration. Arterioscler. Thromb. Vasc. Biol. 32, e72–e80 (2012).
Salagianni, M. et al. Toll-like receptor 7 protects from atherosclerosis by constraining “inflammatory” macrophage activation. Circulation 126, 952–962 (2012).
Oka, T. et al. Mitochondrial DNA that escapes from autophagy causes inflammation and heart failure. Nature 485, 251–255 (2012). This is the first demonstration that autophagy prevents dsDNA released by damaged mitochondria from initiating a TLR9-dependent pathway with cardiotoxic effects.
Lim, J. E. et al. MyD88 deficiency ameliorates beta-amyloidosis in an animal model of Alzheimer’s disease. Am. J. Pathol. 179, 1095–1103 (2011).
Lehmann, S. M. et al. An unconventional role for miRNA: let-7 activates Toll-like receptor 7 and causes neurodegeneration. Nat. Neurosci. 15, 827–835 (2012).
Maatouk, L. et al. TLR9 activation via microglial glucocorticoid receptors contributes to degeneration of midbrain dopamine neurons. Nat. Commun. 9, 2450 (2018).
Ros-Bernal, F. et al. Microglial glucocorticoid receptors play a pivotal role in regulating dopaminergic neurodegeneration in parkinsonism. Proc. Natl Acad. Sci. USA 108, 6632–6637 (2011).
Scholtzova, H. et al. Innate immunity stimulation via Toll-like receptor 9 ameliorates vascular amyloid pathology in Tg-SwDI mice with associated cognitive benefits. J. Neurosci. 37, 936–959 (2017).
Sheridan, C. Drug developers switch gears to inhibit STING. Nat. Biotechnol. 37, 199–201 (2019).
Pestal, K. et al. Isoforms of RNA-editing enzyme ADAR1 independently control nucleic acid sensor MDA5-driven autoimmunity and multi-organ development. Immunity 43, 933–944 (2015).
Nakahama, T. et al. ADAR1-mediated RNA editing is required for thymic self-tolerance and inhibition of autoimmunity. EMBO Rep. 19 (2018).
Laxminarayana, D., Khan, I. U. & Kammer, G. Transcript mutations of the alpha regulatory subunit of protein kinase A and up-regulation of the RNA-editing gene transcript in lupus T lymphocytes. Lancet 360, 842–849 (2002).
Hadjadj, J. et al. Pediatric Evans syndrome is associated with a high frequency of potentially damaging variants in immune genes. Blood 134, 9–21 (2019).
Kono, M. et al. Dyschromatosis symmetrica hereditaria and Aicardi-Goutieres syndrome 6 are phenotypic variants caused by ADAR1 mutations. J. Invest. Dermatol. 136, 875–878 (2016).
Crow, Y. J. et al. Mutations in the gene encoding the 3′-5′ DNA exonuclease TREX1 cause Aicardi-Goutieres syndrome at the AGS1 locus. Nat. Genet. 38, 917–920 (2006).
Lee-Kirsch, M. A. et al. Mutations in the gene encoding the 3′-5′ DNA exonuclease TREX1 are associated with systemic lupus erythematosus. Nat. Genet. 39, 1065–1067 (2007).
Kerur, N. et al. cGAS drives noncanonical-inflammasome activation in age-related macular degeneration. Nat. Med. 24, 50–61 (2018).
King, K. R. et al. IRF3 and type I interferons fuel a fatal response to myocardial infarction. Nat. Med. 23, 1481–1487 (2017). This is the first report proving that abnormal secretion of type I interferon downstream of deregulated CGAS–STING signalling contributes to the long-term detrimental consequences of myocardial infarction.
Hu, Q. et al. STING-mediated intestinal barrier dysfunction contributes to lethal sepsis. EBioMedicine 41, 497–508 (2019).
Luo, X. et al. Expression of sting is increased in liver tissues from patients with NAFLD and promotes macrophage-mediated hepatic inflammation and fibrosis in mice. Gastroenterology 155, 1971–1984.e1974 (2018).
Yu, Y. et al. STING-mediated inflammation in Kupffer cells contributes to progression of nonalcoholic steatohepatitis. J. Clin. Invest. 129, 546–555 (2019).
Ahn, J. et al. Inflammation-driven carcinogenesis is mediated through STING. Nat. Commun. 5, 5166 (2014).
Papinska, J. et al. Activation of stimulator of interferon genes (STING) and Sjogren syndrome. J. Dent. Res. 97, 893–900 (2018).
Haag, S. M. et al. Targeting STING with covalent small-molecule inhibitors. Nature 559, 269–273 (2018). This article and reference 249 independently report the identification of small molecules and a cyclopeptide that potently inhibit STING.
Li, S. et al. The cyclopeptide astin C specifically inhibits the innate immune CDN sensor sting. Cell Rep. 25, 3405–3421.e3407 (2018).
An, J. et al. Inhibition of cyclic GMP-AMP synthase using a novel antimalarial drug derivative in Trex1-deficient mice. Arthritis Rheumatol. 70, 1807–1819 (2018).
Dai, J. et al. Acetylation blocks cGA activity and inhibits self-DNA-induced autoimmunity. Cell 176, 1447–1460.e1414 (2019). This report reveals that aspirin promotes the non-enzymatic inhibitory acetylation of CGAS, unveiling a novel mechanism whereby aspirin mediates anti-inflammatory effects.
Wang, T., Larcher, L. M., Ma, L. & Veedu, R. N. Systematic screening of commonly used commercial transfection reagents towards efficient transfection of single-stranded oligonucleotides. Molecules 23, 2564 (2018).
Bocanegra Gondan, A. I. et al. Effective cancer immunotherapy in mice by polyIC-imiquimod complexes and engineered magnetic nanoparticles. Biomaterials 170, 95–115 (2018).
Salaun, B., Coste, I., Rissoan, M. C., Lebecque, S. J. & Renno, T. TLR3 can directly trigger apoptosis in human cancer cells. J. Immunol. 176, 4894–4901 (2006).
Tsitoura, D. et al. Early clinical evaluation of the intranasal TLR7 agonist GSK2245035: use of translational biomarkers to guide dosing and confirm target engagement. Clin. Pharmacol. Ther. 98, 369–380 (2015).
Felten, R., Scher, F., Sagez, F., Chasset, F. & Arnaud, L. Spotlight on anifrolumab and its potential for the treatment of moderate-to-severe systemic lupus erythematosus: evidence to date. Drug Des. Devel. Ther. 13, 1535–1543 (2019).
Lai, Y. F. et al. Functional polymorphisms of the TLR7 and TLR8 genes contribute to Mycobacterium tuberculosis infection. Tuberculosis 98, 125–131 (2016).
Mollaki, V. et al. Polymorphisms and haplotypes in TLR9 and MYD88 are associated with the development of Hodgkin’s lymphoma: a candidate-gene association study. J. Hum. Genet. 54, 655–659 (2009).
Bharti, D. et al. The role of TLR9 polymorphism in susceptibility to pulmonary tuberculosis. Immunogenetics 66, 675–681 (2014).
Forsbach, A. et al. Identification of RNA sequence motifs stimulating sequence-specific TLR8-dependent immune responses. J. Immunol. 180, 3729–3738 (2008).
Sistigu, A. et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat. Med. 20, 1301–1309 (2014).
Chan, Y. K. & Gack, M. U. Viral evasion of intracellular DNA and RNA sensing. Nat. Rev. Microbiol. 14, 360–373 (2016).
Bidwell, B. N. et al. Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape. Nat. Med. 18, 1224–1231 (2012).
Brand, F. J., 3rd, de Rivero Vaccari, J. C., Mejias, N. H., Alonso, O. F. & de Rivero Vaccari, J. P. RIG-I contributes to the innate immune response after cerebral ischemia. J. Inflamm. 12, 52 (2015).
Galluzzi, L., Bravo-San Pedro, J. M., Levine, B., Green, D. R. & Kroemer, G. Pharmacological modulation of autophagy: therapeutic potential and persisting obstacles. Nat. Rev. Drug Discov. 16, 487–511 (2017).
Theisen, D. J. et al. WDFY4 is required for cross-presentation in response to viral and tumor antigens. Science 362, 694–699 (2018).
Yang, W. et al. Genome-wide association study in Asian populations identifies variants in ETS1 and WDFY4 associated with systemic lupus erythematosus. PLOS Genet. 6, e1000841 (2010).
Kochi, Y. et al. Splicing variant of WDFY4 augments MDA5 signalling and the risk of clinically amyopathic dermatomyositis. Ann. Rheum. Dis. 77, 602–611 (2018).
Echem, C. et al. Mitochondrial DNA: a new driver for sex differences in spontaneous hypertension. Pharmacol. Res. 144, 142–150 (2019).
Xin, G. et al. Sex hormone affects the severity of non-alcoholic steatohepatitis through the MyD88-dependent IL-6 signaling pathway. Exp. Biol. Med. 240, 1279–1286 (2015).
Cillo, A. R. & Mellors, J. W. Which therapeutic strategy will achieve a cure for HIV-1? Curr. Opin. Virol. 18, 14–19 (2016).
Bartsch, K. et al. Absence of RNase H2 triggers generation of immunogenic micronuclei removed by autophagy. Hum. Mol. Genet. 26, 3960–3972 (2017).
Banerjee, S. et al. OAS-RNase L innate immune pathway mediates the cytotoxicity of a DNA-demethylating drug. Proc. Natl Acad. Sci. USA 116, 5071–5076 (2019).
Chiu, Y. H., Macmillan, J. B. & Chen, Z. J. RNA polymerase III detects cytosolic DNA and induces type I interferons through the RIG-I pathway. Cell 138, 576–591 (2009).
Ferguson, B. J., Mansur, D. S., Peters, N. E., Ren, H. & Smith, G. L. DNA-PK is a DNA sensor for IRF-3-dependent innate immunity. eLife 1, e00047 (2012).
Dutta, D. et al. BRCA1 Regulates IFI16 mediated nuclear innate sensing of herpes viral DNA and subsequent induction of the innate inflammasome and interferon-beta responses. PLOS Pathog. 11, e1005030 (2015).
Kondo, T. et al. DNA damage sensor MRE11 recognizes cytosolic double-stranded DNA and induces type I interferon by regulating STING trafficking. Proc. Natl Acad. Sci. USA 110, 2969–2974 (2013).
Parvatiyar, K. 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. 13, 1155–1161 (2012).
Hornung, V. et al. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature 458, 514–518 (2009).
Almine, J. F. et al. IFI16 and cGAS cooperate in the activation of STING during DNA sensing in human keratinocytes. Nat. Commun. 8, 14392 (2017).
Shannon, J. L. et al. Polyglutamine binding protein 1 (PQBP1) inhibits innate immune responses to cytosolic DNA. Mol. Immunol. 99, 182–190 (2018).
Tan, Y. & Kagan, J. C. Innate immune signaling organelles display natural and programmable signaling flexibility. Cell 177, 384–398.e311 (2019). This study demonstrates that supramolecular platforms commonly used to drive cytokine secretion in response to nucleic acid sensing, such as the myddosome, can be engineered to mediate user-defined biological outcomes.
Lama, L. et al. Development of human cGAS-specific small-molecule inhibitors for repression of dsDNA-triggered interferon expression. Nat. Commun. 10, 2261 (2019).
Chung, H. et al. Human ADAR1 prevents endogenous RNA from triggering translational shutdown. Cell 172, 811–824.e14 (2018).
Isaacs, A., Lindenmann, J. & Valentine, R. C. Virus interference. II. Some properties of interferon. Proc. R. Soc. Lond. B Biol. Sci. 147, 268–273 (1957).
Isaacs, A. & Lindenmann, J. Virus interference. I. The interferon. Proc. R. Soc. Lond. B Biol. Sci. 147, 258–267 (1957).
Jensen, K. E., Neal, A. L., Owens, R. E. & Warren, J. Interferon responses of chick embryo fibroblasts to nucleic acids and related compounds. Nature 200, 433–434 (1963).
Rotem, Z., Cox, R. A. & Isaacs, A. Inhibition of virus multiplication by foreign nucleic acid. Nature 197, 564–566 (1963).
Medzhitov, R., Preston-Hurlburt, P. & Janeway, C. A. Jr. A human homologue of the drosophila toll protein signals activation of adaptive immunity. Nature 388, 394–397 (1997).
Ferrandon, D., Imler, J. L., Hetru, C. & Hoffmann, J. A. The Drosophila systemic immune response: sensing and signalling during bacterial and fungal infections. Nat. Rev. Immunol. 7, 862–874 (2007).
Gay, N. J., Symmons, M. F., Gangloff, M. & Bryant, C. E. Assembly and localization of Toll-like receptor signalling complexes. Nat. Rev. Immunol. 14, 546–558 (2014).
Galluzzi, L., Vanpouille-Box, C., Bakhoum, S. F. & Demaria, S. SnapShot: CGAS-STING signaling. Cell 173, 276–276 e271 (2018).
Acknowledgements
The authors apologize to the authors of multiple preclinical and clinical studies on pharmacological modulators of the nucleic acid sensing machinery that could not be cited owing to space limitations. C.V.B. is supported by a start-up grant from the Department of Radiation Oncology at Weill Cornell Medicine (New York, NY, USA) and a Career Development Award from the Radiation Research Foundation (Kansas City, MI, USA). L.G. is supported by a Breakthrough Level 2 grant from the US Department of Defense, Breast Cancer Research Program (BC180476P1), by a start-up grant from the Department of Radiation Oncology at Weill Cornell Medicine (New York, NY, USA), by industrial collaborations with Lytix (Oslo, Norway) and Phosplatin (New York, NY, USA) and by donations from Phosplatin (New York, NY, USA), the Luke Heller TECPR2 Foundation (Boston, MA, USA) and Sotio (Prague, Czech Republic).
Author information
Authors and Affiliations
Contributions
C.V.B. and L.G. conceived this Review and wrote the first version of the manuscript, with constructive input from J.A.H. C.V.B. and L.G. prepared display items. All authors approved the final version of the manuscript.
Corresponding author
Ethics declarations
Competing interests
C.V.B. and J.H. have no relevant conflicts of interest. L.G. provides remunerated consulting to OmniSEQ (Buffalo, NY, USA), Astra Zeneca (Gaithersburg, MD, USA), VL47 (New York, NY, USA) and the Luke Heller TECPR2 Foundation (Boston, MA, USA), and is a member of the Scientific Advisory Committee of OmniSEQ (Buffalo, NY, USA).
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Related links
Bristol-Myers Squibb (In the Pipeline): https://www.bms.com/researchers-and-partners/in-the-pipeline.html
ClinicalTrials.gov: http://www.clinicaltrials.gov
Glossary
- Pattern recognition receptors
-
(PRRs). Members of an evolutionarily ancient group of receptors that initiate innate immunity on detection of conserved microbial products or endogenous damage-associated molecular patterns.
- Damage-associated molecular patterns
-
Endogenous molecules with immunomodulatory activity that are exposed or secreted upon cellular damage or death, culminating in the activation of pattern recognition receptors on neighbouring cells.
- CpG regions
-
Portions of a nucleic acid rich in unmethylated cytosine–guanine dimers.
- Dendritic cell
-
(DC). Haematopoietic cell of myeloid derivation specialized in the uptake of antigens from the microenvironment and their presentation to T cells.
- Cross-priming
-
Process whereby specific dendritic cell subsets initiate CD8+ (rather than CD4+) cell responses against antigens taken up from the microenvironment.
- Immune checkpoint blocker
-
(ICB). Member of a class of drugs that mediate anticancer effects by inhibiting the molecular systems involved in the natural extinction of immune responses.
- Plasmacytoid DCs
-
Particular variant of dendritic cells (DCs) that morphologically resemble plasma cells and secrete high levels of type I interferon in response to activatory signals.
- Monocyte-derived DCs
-
Specific variant of dendritic cells (DCs) that originate from circulating monocytes on recruitment to sites of inflammation.
- Viral interference
-
Process whereby virally infected cells establish resistance to infection locally via paracrine circuitries depending on type I interferon.
- Imidazoquinolines
-
Organic molecules characterized by a tricyclic structure that has been frequently used as a lead for the development of TLR7 agonists.
- CD4+ T helper cells
-
Lymphocytes that support and direct the activity of other immune cells, including CD8+ T cells, by releasing cytokines.
- γδ T cells
-
Relatively rare variant of T cells that express a γδ (rather than the common αβ) T cell receptor, and hence are endowed with major histocompatibility complex-independent activity.
- Monobenzone
-
Monobenzyl ether of hydroquinone, often used for medical depigmentation.
- Nivolumab
-
Clinically used monoclonal antibody that acts by inhibiting programmed cell death 1 (PD-1), hence unleashing the effector function of T cells.
- CD8+ cytotoxic T lymphocyte
-
(CTL). Lymphoid cell capable of mediating cytotoxic functions in response to a specific antigenic epitope presented on major histocompatibility complex class I molecules.
- Natural killer cells
-
(NK cells). Lymphoid cells capable of mediating cytotoxic functions in an antigen-independent manner.
- Cetuximab
-
Monoclonal antibody specific for epidermal growth factor receptor, commonly used in the clinic for the treatment of colorectal cancer and some forms of lung cancer.
- Durvalumab
-
Clinically used monoclonal antibody that acts by inhibiting PD-L1, hence unleashing the effector function of T cells.
- Oligodeoxynucleotides
-
Short nucleic acids based on a deoxyribose backbone.
- Interferon-stimulated genes
-
(ISGs). Members of a large gene set that is upregulated in response to type I interferon signalling, such as MX1 or CXCL10.
- Ribavirin
-
Commonly used antiviral medication that acts as a guanosine analogue, hence inhibiting viral replication and mRNA processing
- TH1 cell cytokine
-
Cytokine that favours the polarization of the immune response towards a type 1 T helper cell (TH1 cell) profile, which is generally associated with robust antiviral and anticancer functions (for example, interferon-γ, tumour necrosis factor).
- Regulated cell death
-
Variant of cell death that is mediated by a genetically encoded machinery, and hence can be modulated by pharmacological or genetic interventions.
- Neoangiogenesis
-
Process whereby cancer cells initiate the vascularization of developing tumours, which is instrumental for disease progression and metastatic dissemination.
- Myeloid-derived suppressor cells
-
Immature myeloid cells that mediate robust immunosuppressive effects, hence favouring cancer progression and resistance to therapy.
- M2-like tumour-associated macrophages
-
Tumour-infiltrating macrophages that have immunosuppressive and proangiogenic function.
- M1-like tumour-associated macrophages
-
Tumour-infiltrating macrophages that mediate immunostimulatory and inflammatory effects.
- Pembrolizumab
-
Clinically used monoclonal antibody that acts by inhibiting programmed cell death 1 (PD-1), hence unleashing the effector function of T cells.
- Abscopal response
-
In radiation oncology, the relatively rare phenomenon whereby irradiation of one malignant lesion results in objective responses at a non-irradiated disease site.
- Inflammasome
-
Supramolecular platform that initiates the proteolytic maturation of proinflammatory cytokines such as IL-1β and IL-18.
- Entecavir
-
Orally available antiviral medication that acts as a guanosine analogue, commonly used for the treatment of hepatitis B virus infection.
- Tenofovir
-
Antiviral medication that acts as a reverse transcriptase inhibitor, commonly used for the treatment of hepatitis B virus infection and pre-exposure and post-exposure HIV-1 prophylaxis.
- Small interfering RNAs
-
Short RNA duplexes extensively used for temporary gene silencing on transfection.
- A-to-I editing
-
Post-transcriptional modification of RNA molecules that involves the enzymatic deamination of adenosine to inosine.
- Non-oncogene addition
-
Concept identifying the dependency of some cancer cells on molecular functions that are not involved in oncogenesis.
- Cyclic dinucleotides
-
(CDNs). Single-phosphate nucleotides with a cyclic bond arrangement between the sugar and phosphate groups, largely used by both prokaryotes and eukaryotes as a second messenger.
- Systemic lupus erythematosus
-
(SLE). Systemic autoimmune disease in which the immune system mistakenly attacks healthy tissues, including the skin, joints, kidneys, brain and other organs.
- Rheumatoid arthritis
-
Long-term autoimmune disorder that primarily affects joints, resulting in warmness, swelling and pain and ultimately leading to permanent joint destruction.
- Synovial lining
-
The lining of the joints, normally only one or two cell layers thick, that is responsible for the production of the joint fluid.
- Dermatomyositis
-
Acquired muscle disease that is characterized by chronic muscle inflammation accompanied by weakness.
- Evans syndrome
-
Very rare autoimmune disorder in which the immune system destroys red blood cells, white blood cells and/or platelets.
- Aicardi–Goutières syndrome
-
Inherited encephalopathy that affects newborns and is characterized by dysregulated type I interferon production.
- Sjögren syndrome
-
Autoimmune disorder affecting salivary and tear glands that generally manifests itself with dry mouth and dry eyes.
- Tumour-draining lymph nodes
-
Lymph nodes that collect extracellular fluids from the anatomical district where a malignant lesion is located.
- Cytokine release syndrome
-
Potentially lethal side effect of some immunotherapies (including some nucleic acid sensor agonists) characterized by the widespread activation of immune cells and consequent release of large amounts of cytokines in the bloodstream.
- Autophagy
-
Evolutionarily old homeostatic process culminating in the lysosomal degradation of superfluous, ectopic or potentially dangerous cytosolic entities.
Rights and permissions
About this article
Cite this article
Vanpouille-Box, C., Hoffmann, J.A. & Galluzzi, L. Pharmacological modulation of nucleic acid sensors — therapeutic potential and persisting obstacles. Nat Rev Drug Discov 18, 845–867 (2019). https://doi.org/10.1038/s41573-019-0043-2
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41573-019-0043-2