Chromosomal instability (CIN) is a hallmark of cancer that drives tumour evolution. It is now recognised that CIN in cancer leads to the constitutive production of misplaced DNA in the form of micronuclei and chromatin bridges. These structures are detected by the nucleic acid sensor cGAS, leading to the production of the second messenger 2′3′-cGAMP and activation of the critical hub of innate immune signalling STING. Activation of this immune pathway should instigate the influx and activation of immune cells, resulting in the eradication of cancer cells. That this does not universally occur in the context of CIN remains an unanswered paradox in cancer. Instead, CIN-high cancers are notably adept at immune evasion and are highly metastatic with typically poor outcomes. In this review, we discuss the diverse facets of the cGAS–STING signalling pathway, including emerging roles in homeostatic processes and their intersection with genome stability regulation, its role as a driver of chronic pro-tumour inflammation, and crosstalk with the tumour microenvironment, which may collectively underlie its apparent maintenance in cancers. A better understanding of the mechanisms whereby this immune surveillance pathway is commandeered by chromosomally unstable cancers is critical to the identification of new vulnerabilities for therapeutic exploitation.

Chromosomal instability, characterised by persistent errors in chromosome segregation, is a hallmark of cancer that drives tumour evolution [1]. Chromosome segregation errors frequently lead to the formation of structures known as micronuclei (MN) — which arise from whole chromosomes or fragments that lag behind during anaphase — and chromatin bridges [2]. Upon mitotic exit, mis-segregated chromosomes recruit their own nuclear envelope, which is often structurally unstable, leading to MN envelope collapse and exposure of micronuclear contents to the cytosol [3] (Figure 1).

CIN as a driver of cGAS–STING signalling.

Figure 1.
CIN as a driver of cGAS–STING signalling.

Chromosome segregation errors in chromosomally unstable cells can lead to the generation of immunostimulatory genomic dsDNA in the form of micronuclear DNA that becomes exposed to the cytosol upon MN rupture or stretched chromatin in chromatin bridges. Production of 2′3′-cGAMP by cGAS activates STING, which, in turn, activates TBK1. TBK1 phosphorylates the transcription factor IRF3, driving the expression of several type I IFNs. Besides TBK1/IRF3/IFN signalling, STING also instigates NF-κB signalling programs. Canonical NF-κB signalling is triggered through the phosphorylation and suppression of NF-κB inhibitor, alpha (IκBα), releasing the RelA/p50 complex into the nucleus. STING also elicits non-canonical NF-κB signalling through the p100/RelB complex through an as yet poorly understood mechanism. CIN, chromosomal instability; dsDNA, double-stranded DNA; IFN, interferon; MN, micronucleus.

Figure 1.
CIN as a driver of cGAS–STING signalling.

Chromosome segregation errors in chromosomally unstable cells can lead to the generation of immunostimulatory genomic dsDNA in the form of micronuclear DNA that becomes exposed to the cytosol upon MN rupture or stretched chromatin in chromatin bridges. Production of 2′3′-cGAMP by cGAS activates STING, which, in turn, activates TBK1. TBK1 phosphorylates the transcription factor IRF3, driving the expression of several type I IFNs. Besides TBK1/IRF3/IFN signalling, STING also instigates NF-κB signalling programs. Canonical NF-κB signalling is triggered through the phosphorylation and suppression of NF-κB inhibitor, alpha (IκBα), releasing the RelA/p50 complex into the nucleus. STING also elicits non-canonical NF-κB signalling through the p100/RelB complex through an as yet poorly understood mechanism. CIN, chromosomal instability; dsDNA, double-stranded DNA; IFN, interferon; MN, micronucleus.

Close modal

The cyclic GMP-AMP synthase (cGAS) is the principal sensor of pathogen-derived cytosolic double-stranded DNA (dsDNA) of infected host cells in mammals [4–6]. However, owing to its mechanism of activation, which relies on the detection of ‘out-of-context’ cytosolic dsDNA rather than pathogen-specific DNA sequences and features [7], misplaced ‘self-DNA’ arising from ruptured MN [8–10] and chromatin bridges [11] is also recognised by cGAS, linking CIN to innate immunity [8–10, 12, 13]. Binding of cGAS to dsDNA activates its catalytic activity and triggers the production of the cyclic dinucleotide 2′3′-cGAMP, which activates the immune signalling hub, stimulator of interferon genes (STING) [4–6]. Upon activation, STING relocalises from the endoplasmic reticulum to the perinuclear Golgi apparatus, where it forms an oligomeric platform on which TANK-binding kinase 1 (TBK1) phosphorylates the transcription factor interferon regulatory factor 3 (IRF3), prompting its nuclear entry and the expression of genes encoding type I interferons (type I IFNs) [14]. STING relocation also activates the canonical and non-canonical nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathways [15], which drive the expression of pro-inflammatory genes encoding a swathe of pleiotropic cytokines and chemokines [16].

Importantly, whilst STING-driven IRF3- and NF-κB-dependent signalling cascades have overlapping functions with regards to mounting cell-intrinsic innate immune responses, their differential regulation downstream of STING, as well as the chronicity and intensity of signalling are major determinants of cGAS–STING pathway output as either anti- or pro-tumorigenic [17]. While it is evident that stimulation, particularly in the acute setting, of STING in antigen-presenting cells is tumour suppressive [18–20], the role of cGAS–STING signalling as a result of constitutive CIN-driven stimulation in the tumour environment is not as clearly defined. Indeed, recent evidence instead favours a role for cGAS–STING in the progression of chromosomally unstable cancers [21].

The near-ubiquitous presence of varying degrees of CIN among cancers [1] suggests that tumour cells experience selective pressures that should compel them to overcome cGAS–STING activation and immune surveillance. Indeed, that cGAS–STING activity can represent a barrier to tumorigenesis has been demonstrated experimentally in vivo, including in murine models of colitis-associated colon cancer [22–24], and inferred from observations that cGAS and STING are the subject of loss-of-function mutations and/or epigenetic silencing in a variety of cancers [25], including subsets of glioma [26], colorectal [27], melanoma [28, 29], gastric [30], lung [31] and ovarian [32] cancers. Nevertheless, mutations are rare — with a mutational frequency of just 0.6% and 0.5% for cGAS and STING, respectively, among tumours of The Cancer Genome Atlas (TCGA) PanCancer database [1] — and the extent of epigenetic silencing demonstrates a large variation in cGAS and STING promoter methylation in tumour samples compared with corresponding normal tissues. Indeed, in multiple tumour types, including upper gastrointestinal, colorectal, bladder and thyroid cancers, average cGAS–STING promoter methylation is decreased, with a concomitant increase in expression, in tumour over normal tissue [25, 33], suggesting that direct silencing of cGAS and STING is not a pervasive mechanism for immune evasion in cancers.

To the contrary, emerging evidence from multiple tumour settings, including carcinogen-induced skin cancer [34], Lewis lung carcinoma [35], chromosomally unstable triple-negative breast cancer (TNBC) [36] and metastatic lung and breast cancer models [13, 37] has now begun to reveal a role for cGAS–STING activity as a driver of tumour progression.

The cGAS–STING pathway has mainly been examined through the lens of its tumour cell-extrinsic function: namely, the activation of cell-mediated immunity through type I IFN signalling [20]. However, emerging evidence suggest that there are myriad cellular processes beyond cytosolic immunity in which cGAS and STING are active participants, which may explain why pathway activity is maintained in some tumour contexts. Moreover, the convergence of multiple of these processes, including DNA damage responses, cell cycle control, senescence, autophagy and cell death, on genome stability regulation underscores the relevance of cell-intrinsic cGAS and STING functions to CIN tumours in particular (Figure 2).

Cell-intrinsic functions of cGAS and STING in genome stability regulation.

Figure 2.
Cell-intrinsic functions of cGAS and STING in genome stability regulation.

cGAS and STING (both inter- and independently) exert influence over several signalling programs for the regulation of genome stability and cell survival outcomes downstream of CIN. Regulatory functions of cGAS–STING can be beneficial to cell survival and chromosomal stability in the context of lower levels of genomic stress, supporting orderly cell division, DNA repair responses and the autophagic clearance of cytosolic DNA. However, as genotoxic stress increases, cGAS–STING activity skews increasingly towards genome-destabilising, as well as anti-proliferative and pro-apoptotic signalling programs, such as DDR inhibition, senescence and autophagic and mitotic death, pushing cells past the tolerable CIN threshold and restricting the propagation of cells with excessively unstable genomes. DDR, DNA damage response; dsDNA, double-stranded DNA; MN, micronucleus; SASP, senescence-associated secretory phenotype.

Figure 2.
Cell-intrinsic functions of cGAS and STING in genome stability regulation.

cGAS and STING (both inter- and independently) exert influence over several signalling programs for the regulation of genome stability and cell survival outcomes downstream of CIN. Regulatory functions of cGAS–STING can be beneficial to cell survival and chromosomal stability in the context of lower levels of genomic stress, supporting orderly cell division, DNA repair responses and the autophagic clearance of cytosolic DNA. However, as genotoxic stress increases, cGAS–STING activity skews increasingly towards genome-destabilising, as well as anti-proliferative and pro-apoptotic signalling programs, such as DDR inhibition, senescence and autophagic and mitotic death, pushing cells past the tolerable CIN threshold and restricting the propagation of cells with excessively unstable genomes. DDR, DNA damage response; dsDNA, double-stranded DNA; MN, micronucleus; SASP, senescence-associated secretory phenotype.

Close modal

DNA damage responses

Immune-dependent

The role of cGAS–STING-driven immune surveillance as a cell-extrinsic genome surveillance mechanism downstream of DNA damage-induced CIN has been well-established [8, 9, 12, 38]. However, components of the cGAS–STING pathway have been implicated in more direct modes of genome surveillance in DNA damage contexts.

For example, several studies have shown that the expression of a subset of interferon-stimulated genes (ISGs), known as the IFN-related DNA damage resistance signature (IRDS), which is associated with resistance to DNA-damaging chemo- and radiotherapies [39–41] is sustained by cGAS–STING-driven IFN activation following prolonged DNA damage [42–44]. Although the full extent of mechanisms underlying the protective nature of certain chronic IFN responses remains unknown, two studies linked persistent STING activity with the IFN-driven expression of several PARP-family genes, including PARP9 and PARP12, posited to promote tumour cell resistance to genotoxic stresses [43, 45] by enhancing DNA damage repair responses [46].

Whilst this supports that subsets of cGAS–STING-driven ISGs can exert genome-stabilising effects that mediate resistance to some genotoxic stresses, genome de-stabilising functions have also been reported. One such instance involves the ubiquitin-like modifier ISG15, an important downstream target of STING-driven IFN [47]. Up-regulation of ISG15 is reported to accelerate DNA replication fork progression by promoting the fork restart activity of the DNA helicase RECQ1 after fork stalling, resulting in extensive DNA damage and chromosomal aberrations, sensitising cancer cells to chemotherapeutic treatments [48]. However, a number of genome-stabilising functions have also been ascribed to ISG15 [49–52], in keeping with its described function as a transcriptional target of p53 [53], exemplifying the broader paradox of innate immune responses in the modulation of the DNA damage response (DDR) [54].

The role of innate immune signalling in DDR regulation extends beyond STING-driven IFN responses, with several reports of canonical and non-canonical NF-κB signalling pathway involvement in double-strand break (DSB) repair [55–59]. However, notwithstanding the well-established role of NF-κB pathways as downstream effector programs of the cGAS–STING axis [15], a direct participation of cGAS–STING was not investigated in these reports.

Despite the differential impact of cGAS–STING-dependent responses on genome stability, these studies collectively demonstrate the centrality of cGAS–STING signalling, not only as a consequence of CIN, but as an active regulator of DNA damage responses.

Immune-independent

Multiple emerging mechanisms of DDR modulation by cGAS–STING pathway components have also been described to occur in a manner that is immune signalling or interferon independent. Intriguingly, cGAMP-induced activation of the DDR has recently been described in cells from invertebrate species that lack IRF3 and type I interferons, suggesting that involvement of the cGAS–STING axis in DDR signalling predates the evolution of the type I interferon system in vertebrates [60].

One such mechanism, reported by Banerjee et al. [60], involves the cGAS/STING/TBK1 axis, but not its downstream canonical IFN signalling pathway, in mediating DDR signalling following treatment with genotoxic agents [60]. Mechanistically, cGAS–STING-activated TBK1 was shown to promote the autophosphorylation of the DDR kinase ATM, resulting in the engagement of the CHK2–p53–p21 transduction pathway and the consequent enforcement of a G1 cell cycle arrest.

Other immune-independent functions for cGAS–STING in DDR regulation have been ascribed to non-canonical functions of cGAS that pertain to its capacity to enter the nucleus and associate with chromatin [61]. For instance, two independent studies recently found that nuclear cGAS can act as a suppressor of homologous recombination (HR)-mediated DSB repair, albeit through distinct proposed mechanisms [62, 63]. In the model proposed by Liu et al. [62], cGAS is recruited to DSBs, where it interacts with PARP1 via poly(ADP-ribose) moieties, impeding the formation of the PARP1-Timeless complex, required for efficient HR [62]. Jiang et al. [63], on the other hand, proposed a more indirect mode of HR regulation, whereby cGAS oligomerisation compacts the dsDNA template into a higher-order state that is less accessible to RAD51-mediated strand invasion [63]. Although distinct in mechanistic terms, functionally both studies showed that cGAS exacerbates DNA damage and accelerates CIN, promoting tumorigenesis [62] or precipitating cell death in cells exposed to acute genomic stress [63].

Conversely, two recent studies ascribed genome-stabilising effects to nuclear cGAS with respect to its ability to mediate responses to DNA damage. In the first of these, recruitment of cGAS to replication forks was shown to decelerate fork progression to safeguard genome stability. More specifically, loss of cGAS was shown to result in excessive fork speed and premature fork restart after fork stalling, eliciting the activation of the DDR sensor ATR and sensitising cells to DNA-damaging treatments [64]. In a second report, cGAS was observed to occupy deprotected telomeres during mitosis, where it was proposed to repress the DNA damage repair signalling activity that gives rise to CIN-initiating chromosome end-to-end fusions [65].

Of note, the above-described functions of nuclear cGAS were shown to be independent of STING when investigated [63–65], suggesting that cGAS and STING integrate distinct functions for the regulation of DNA damage responses and genome stability. Indeed, STING itself has also been proposed to act in a genome stabilising-manner, independently of cGAS. For example, a study in breast cancer cells identified that part of the intracellular STING pool resides at the inner nuclear membrane, acting as a positive regulator of the DDR and shielding against excessive genomic instability separately from both cGAS and downstream interferon signalling [66].

Altogether, these studies further add to the notion that the increasingly diverse spread in reports surrounding the localisations of cGAS and STING and their proposed inter- and independent functions reflect an array of roles in various homeostatic processes that intersect with cell-intrinsic genome stability regulation.

Cell division & senescence

This notion is further bolstered by recent studies implicating cGAS and/or STING and their downstream transcriptional programs in cell division regulation [64, 67, 68]. In one such report, STING was found to drive the p53- and NF-κB-dependent accumulation of the cyclin-dependent kinase inhibitor p21, ensuring timely entry to S-phase and mitosis [67]. Loss of STING, but not cGAS, promoted precocious cell division, resulting in the acquisition of chromosomal aberrations and CIN, which was further enhanced by ionising radiation. A similar requirement for STING, as well as cGAS, in regulating p21 levels, was recently also found to impede premature mitotic entry and resultant chromosome segregation errors through the IRF3-driven transcriptional up-regulation of p53 [68], suggesting that multiple components downstream of STING, including the IRF3 and NF-κB signalling arms, converge on the regulation of cell division to maintain genome stability.

However, control of cGAS–STING over the cell cycle extends beyond the maintenance of chromosomal homeostasis during routine cell division and also plays a key role in establishing a state of permanent cell-cycle arrest, known as cellular senescence, in response to persistent sub-lethal genomic stress [69]. The cytosolic DNA species that are generated in such stress contexts are recognised by cGAS, instigating the STING-mediated production of multiple cytokines and chemokines [70–72], collectively referred to as the senescence-associated secretory phenotype (SASP), which reinforces and spreads the senescent growth arrest phenotype in autocrine and paracrine manners, respectively [73–76]. As such, cGAS and STING can co-operate to ensure segregation error-free cell division at steady state and prevent the expansion of cells with excessively unstable genomes.

Autophagy

The macroautophagic pathway (autophagy) is a strictly regulated pathway that oversees the degradation of excess or dysfunctional cytosolic components, and is therefore tightly interrelated with genome integrity [77]. Indeed, CIN and aneuploidy — a phenotypic outcome of CIN [78] — are associated with an increase in autophagic activity [79–81] and endow cells with a selective vulnerability to the suppression of autophagy [82, 83], suggesting that autophagic activity attenuates the deleterious effects of CIN-associated stresses.

A similarly mutual relationship has emerged between autophagy and the cGAS–STING pathway, with multiple reports highlighting the role of autophagy in tuning STING responses through the degradation of multiple components of the cGAS–STING axis, including cGAS and STING themselves, as well as its cytosolic dsDNA substrates [84–91]. Crucially, cGAS–STING was recently shown to play an evolutionarily conserved role in autophagy activation, independently of downstream interferon induction, suggesting that it constitutes a primordial effector function of the STING pathway [92].

Moreover, cGAS–STING-driven autophagy has been shown to mediate the clearance of free endogenous cytosolic dsDNA upon both chromosome gains [93] and DNA damage [92], analogous to its initially reported role in the clearance of pathogenic cytosolic DNA to prevent persistent immune signalling [87]. Together with other observations of autophagic clearance of cytosolic genomic dsDNA in genomic stress contexts [94–96], this suggests that cGAS–STING-driven clearance of cytosolic genomic DNA may represent a tolerogenic mechanism for genomic instability. In line with this, cGAS-dependent autophagy activation was recently reported to promote the growth and survival of highly chromosomally unstable BT-549 TNBC cells, but not of breast cancer lines with lower cytosolic genomic DNA burdens, by enabling the clearance of DNA from their cytosol [97].

Autophagic degradation has also been observed to mediate the removal of micronuclei [10, 98]. Since MN not only constitute a passive marker of CIN, but can actively fuel it through their reincorporation into primary nuclei [99, 100], their autophagic clearance may thus have a directly genome-stabilising effect, echoing early reports highlighting the genome-stabilising nature of autophagy [101, 102]. Intriguingly, a direct role between autophagic clearance of MN and cGAS was recently reported by Zhao et al. [103], who showed that micronuclear cGAS interacts with the autophagy mediator LC3B to help target MN to lysosomes for degradation [103]. Strikingly, the capacity of cGAS to act as a ‘MN-phagy’ receptor was found to be independent of both its cGAMP enzymatic activity and DNA-binding capacity, suggesting that it represents a function that is distinct from its role as a driver of STING-dependent autophagy upon cytosolic DNA sensing [87, 92, 104].

Cell death

Despite the cytoprotective role of autophagy in certain stress contexts, its hyperactivation can also lead to cell death [105]. Indeed, a recent study showed that in cells that bypass senescence (in the absence of p53 and Rb) and reach replicative crisis, extensive telomeric DNA damage leads to the generation of cytosolic DNA species that drive autophagic cell death in a cGAS–STING-dependent manner [106]. Interestingly, attenuation of autophagy or the cGAS–STING pathway enabled cells to bypass crisis despite the accumulation of gross karyotypic aberrations, suggesting that cGAS–STING-driven autophagic death is an essential safeguard against genomic instability driven by telomere crisis. The observation that cGAS–STING-driven autophagic death also occurs outside of telomere crisis in response to cell–cell fusion — a catastrophic event that precipitates aberrant mitosis and CIN — suggests it may constitute a conserved tumour-suppressive mechanism for the elimination of cells experiencing catastrophic levels of CIN [107], despite reports of a protective role for autophagy in less severe CIN contexts.

As such, cGAS–STING-driven autophagy may, to an extent, act on a cell-intrinsic level to ensure unstable genome homeostasis by enabling cell survival at low-level CIN and driving the elimination of tumour cells in circumstances of high or acutely-driven CIN. Since autophagy is also critical for mediating cell-extrinsic homeostatic effects through its involvement in multiple contrasting facets of immunity, including innate immune signalling and danger signal emission (i.e. adjuvanticity), as well as tumour cell antigenicity [108, 109], it is possible that its control through cGAS–STING represents another avenue through which tumours may interface with the local microenvironment (discussed below). However, given the contradictory nature of autophagy in immunity, it remains unclear how such tumour cell-extrinsic effects of STING-driven autophagy may influence tumour cell survival. Nonetheless, it is tempting to speculate that they may act in a manner akin to its cell-intrinsic effects: enabling survival and maintaining tumour growth by suppressing immune recognition at lower-level CIN and stimulating immune responses through the release of danger signals and antigens from dead and dying cells in high-CIN settings.

cGAS–STING activity appears to drive similarly paradoxical outcomes with respect to its function in cell division. For instance, while cGAS–STING pathway activity slows mitosis to avoid error-prone premature cell division under baseline conditions [67, 68], it can also instigate mitotic death in an IRF3 activity-dependent, yet transcription-independent, manner in cells undergoing a prolonged arrest in mitosis [110]. Thus, the strength and duration of activating signals — emerging determinants of how cGAS–STING-signalling outcomes are shaped in different immune cell contexts [17, 111, 112] — also appear to dictate signalling outcomes in CIN contexts.

Although the emerging link between genome stability regulation and the non-canonical functions of cGAS–STING may explain how some chromosomally unstable tumours may derive benefits from STING pathway maintenance, it does not explain how it enables tumours to overcome the high cost of immune surveillance that is described to oftentimes ensue CIN-driven inflammatory signalling [8, 9, 12, 38]. Strikingly, mounting evidence suggests that continuous activation of the cGAS–STING pathway as a result of CIN can fuel chronic inflammation which favours rather than limits the formation, progression and spread of tumours [113, 114], suggesting that tumours harbouring unstable genomes are able to de-emphasise overtly anti-tumour signalling outcomes downstream of STING in favour of more pro-tumour ones (Figure 3).

The dichotomous role of CIN-driven cell-intrinsic cGAS–STING inflammatory signalling in cancer.

Figure 3.
The dichotomous role of CIN-driven cell-intrinsic cGAS–STING inflammatory signalling in cancer.

Activation of cGAS–STING-dependent inflammatory signalling downstream of CIN can exert both tumour-suppressive, as well as tumour-promoting effects in a cell-intrinsic manner. Expression of type I IFNs and downstream engagement of IFNAR/STAT1 induces the expression of multiple effectors, including pro-apoptotic and anti-proliferative genes, that are detrimental to tumour cell survival. Conversely, cGAS–STING-dependent activation of NC-NF-κB signalling can drive IL6/STAT3 signalling and EMT programs, which promote tumour growth and metastasis, respectively. Induction of protective pathways associated with chronic cGAS–STING activity, such as the IRDS and NC-NF-κB signalling can also directly antagonise type I IFN activation, dampening the overtly tumour-suppressive effects of cGAS–STING. IFN, interferon; IFNAR, IFN receptor; IL-6R, IL-6 receptor; IRDS, IFN-related DNA damage resistance signature; MN, micronucleus; NC-NF-κB, non-canonical NF-κB.

Figure 3.
The dichotomous role of CIN-driven cell-intrinsic cGAS–STING inflammatory signalling in cancer.

Activation of cGAS–STING-dependent inflammatory signalling downstream of CIN can exert both tumour-suppressive, as well as tumour-promoting effects in a cell-intrinsic manner. Expression of type I IFNs and downstream engagement of IFNAR/STAT1 induces the expression of multiple effectors, including pro-apoptotic and anti-proliferative genes, that are detrimental to tumour cell survival. Conversely, cGAS–STING-dependent activation of NC-NF-κB signalling can drive IL6/STAT3 signalling and EMT programs, which promote tumour growth and metastasis, respectively. Induction of protective pathways associated with chronic cGAS–STING activity, such as the IRDS and NC-NF-κB signalling can also directly antagonise type I IFN activation, dampening the overtly tumour-suppressive effects of cGAS–STING. IFN, interferon; IFNAR, IFN receptor; IL-6R, IL-6 receptor; IRDS, IFN-related DNA damage resistance signature; MN, micronucleus; NC-NF-κB, non-canonical NF-κB.

Close modal

Using transplantable metastatic tumour models of TNBC, Bakhoum, Ngo et al. [13] recently demonstrated that CIN enriches tumour cells for epithelial–mesenchymal transition (EMT)-related gene expression, supporting metastatic dissemination, in a cGAS–STING-dependent manner [13]. Strikingly, rather than activating canonical NF-κB or type I IFN signalling, cGAS–STING activity in CIN-high TNBC cells was shown to preferentially activate the metastasis-promoting non-canonical NF-κB (NC-NF-κB) pathway [115, 116], suggesting that tumour cells can rewire their cGAS–STING signalling circuitry to co-opt its pro-tumour signalling arms. Although it is unclear how universal a metastatic escape route this represents across different tumour types, CIN has also recently been reported to fuel cGAS–STING-dependent pro-metastatic inflammation in subsets of uveal melanoma (UM) [117] and pancreatic ductal adenocarcinoma (PDAC) [118], besides aggressive breast cancer [13].

Collectively, these studies suggest that CIN-driven activation of cGAS–STING is an important driver of tumour cell invasiveness and metastasis. However, invasiveness is not an obligate outcome of CIN. For instance, Vasudevan et al. [119] recently showed that acute induction of CIN suppresses rather than promotes invasive behaviour across several cancer and non-cancer cell lines, despite inducing extensive micronucleation and NC-NF-κB and inflammatory signal activation [119]. How these discrepancies can be reconciled is unclear, but understanding in what contexts cGAS–STING produces pro-metastatic versus anti-tumour outcomes will be crucial to the appropriate administration of therapies that converge on STING pathway activation.

The tumour cell-intrinsic benefits of CIN-driven inflammation extend beyond its enhancement of metastatic behaviour in advanced stage tumours and have recently also been implicated in early tumour development. For instance, Hong et al. [36] recently showed that some CIN-high primary tumours rely on a pro-survival cGAS–STING-dependent inflammatory response to surmount CIN-imposed stresses [36]. In vitro work showed that loss of cGAS and STING sensitised TNBC cells to both acute (chemical-induced) and chronic (genetic) induction of chromosome mis-segregation and was rescued by the additional abrogation of STAT1, indicating a largely STAT1-driven sensitivity to CIN in their model system. As CIN induction was shown to trigger IL-6 expression and the engagement of the pro-survival IL-6/JAK/STAT3 signalling axis [120, 121] in a cGAS–STING-dependent manner, it was posited that its activation may constitute a requirement to overcome the deleterious effects of CIN-induced STAT1. Moreover, loss of cGAS and STING, as well as inhibition of the IL-6 receptor (IL-6R) were shown to specifically impair the outgrowth of chromosomally unstable TNBC tumours in vivo, suggesting that CIN imparts a tumour cell-intrinsic — if not an additional cell-extrinsic — dependency on cGAS–STING-driven IL-6/STAT3 signalling in primary TNBC cancers.

A similar reliance on cell-intrinsic cGAS–STING-driven inflammation for in vivo tumour growth was also recently reported for human and murine breast tumour cells exhibiting genomic instability due to the overexpression of MYO10, an unconventional myosin that is up-regulated in several aggressive cancers [122]. Interestingly, in this report, STING depletion was shown to suppress the acceleration in tumour growth seen to occur upon CIN induction to the growth rate observed in ‘genomically stable’ tumours, rather than curbing growth altogether, as reported above [36]. This implies that, whereas in some CIN contexts, cGAS–STING-driven inflammation may help cancer cells survive by counterbalancing the costs of genome instability, in other settings, it explicitly lets them thrive.

Altogether, these studies suggest that CIN-driven cGAS–STING inflammatory signalling may be tolerated and even co-opted by tumours that are able to shunt pathway activity towards pro-tumour signalling programs, such as the NC-NF-κB and STAT3 arms of the cGAS–STING pathway, rather than more outrightly anti-tumour arms, such as type I IFN receptor (IFNAR)-driven STAT1 signalling. Moreover, emerging evidence suggests that activation of pro-survival immune signalling arms downstream of cGAS–STING, such as NC-NF-κB signalling and the IRDS, may even directly antagonise the induction of its anti-tumour arms [44, 123, 124], implying that some tumour cells may become addicted to the protective effects of cGAS–STING responses in the face of persistent CIN.

The past few years have seen a shift in our understanding of cGAS–STING signalling in cancer. It is becoming increasingly clear that cGAS–STING pathway functions in tumours extend beyond the induction of tumour-suppressive inflammation. Indeed, multiple studies now point to cGAS–STING pathway involvement in multiple cell-intrinsic signalling programs (as outlined above) that can exert opposing outcomes on tumour cell fitness. Hence, whether cGAS–STING engagement results in an outcome that is beneficial or detrimental to tumour cell survival depends on the status of each of its downstream pathway components (Figure 4). As chromosomally unstable cancers are under especially high selective pressure to acquire adaptations that help avoid tumour-suppressive cGAS–STING-driven programs (e.g. senescence, type I IFN signalling and autophagic death upon crisis) [1, 125], they may disproportionately skew pathway activity towards pro-survival outcomes that further tumour progression.

Genetic context and degree of CIN dictate cGAS–STING signalling outcomes.

Figure 4.
Genetic context and degree of CIN dictate cGAS–STING signalling outcomes.

The cGAS–STING pathway triggers multiple signalling programs in a cell-autonomous manner, which can produce opposing effects on tumour cell fitness. Whether engagement of the cGAS–STING axis results in an outcome that is beneficial or detrimental to tumour cell survival depends on the cumulative status of its downstream pathway components. Highly chromosomally unstable tumours likely acquire adaptations that enable them to avoid activation of overtly anti-proliferative and/or pro-apoptotic signalling programs, such as the senescence program, telomere crisis and type I IFN signalling, and further enhance the beneficial effects of pro-tumour programs. In addition, the fitness costs imparted by CIN-associated stresses may increase the reliance of high CIN tumours on the genome-stabilising effects and pro-survival programs induced by cGAS–STING. As such, chromosomally unstable cancers may tip the balance between beneficial and detrimental cGAS–STING signalling outcomes downstream of CIN in their favour. CIN, chromosomal instability; DDR, DNA damage response; dsDNA, double-stranded DNA; IFN, interferon.

Figure 4.
Genetic context and degree of CIN dictate cGAS–STING signalling outcomes.

The cGAS–STING pathway triggers multiple signalling programs in a cell-autonomous manner, which can produce opposing effects on tumour cell fitness. Whether engagement of the cGAS–STING axis results in an outcome that is beneficial or detrimental to tumour cell survival depends on the cumulative status of its downstream pathway components. Highly chromosomally unstable tumours likely acquire adaptations that enable them to avoid activation of overtly anti-proliferative and/or pro-apoptotic signalling programs, such as the senescence program, telomere crisis and type I IFN signalling, and further enhance the beneficial effects of pro-tumour programs. In addition, the fitness costs imparted by CIN-associated stresses may increase the reliance of high CIN tumours on the genome-stabilising effects and pro-survival programs induced by cGAS–STING. As such, chromosomally unstable cancers may tip the balance between beneficial and detrimental cGAS–STING signalling outcomes downstream of CIN in their favour. CIN, chromosomal instability; DDR, DNA damage response; dsDNA, double-stranded DNA; IFN, interferon.

Close modal

The full scope of pro-tumorigenic cGAS–STING functions likely involves the concerted action of multiple components of the tumour microenvironment (TME), beyond cancer cells. Indeed, many studies have ascribed the influence of cGAS–STING on tumour development and metastasis to the activation of STING signalling in multiple cellular components of the TME other than tumour cells, including astrocytes [37], mesenchymal stromal cells [126] and phagocytes [34]. This propagation of STING signalling into the TME is enabled by a complex network of intercellular transport routes that comprises several cell type-specific channels [127–129], transporters [130–133], gap junctions [37, 134–137] and extracellular vesicles (EVs) [138], as well as endocytic routes, such as phagocytosis [139–141] and transcytosis [142] (Figure 5a).

The pro-tumour functions of cGAS–STING signal transmission in the TME.

Figure 5.
The pro-tumour functions of cGAS–STING signal transmission in the TME.

(a) Intercellular routes through which mediators of cGAS–STING signalling, predominantly in the form of cGAMP or dsDNA, can be propagated into neighbouring (via cell–cell contacts) or more distant cells (via the extracellular space) of the TME to shape the tumour landscape. (b) Reported functions of cGAS–STING signalling in the establishment of an immune-suppressive pro-tumour TME which may be at play in CIN tumour settings. CAF, cancer-associated fibroblast; CIN, chromosomal instability; dsDNA, double-stranded DNA; IFN, interferon; MDSC, myeloid-derived suppressor cell; SASP, senescence-associated secretory phenotype; TAM, tumour-associated macrophage; TME, tumour microenvironment; Treg, regulatory T cell.

Figure 5.
The pro-tumour functions of cGAS–STING signal transmission in the TME.

(a) Intercellular routes through which mediators of cGAS–STING signalling, predominantly in the form of cGAMP or dsDNA, can be propagated into neighbouring (via cell–cell contacts) or more distant cells (via the extracellular space) of the TME to shape the tumour landscape. (b) Reported functions of cGAS–STING signalling in the establishment of an immune-suppressive pro-tumour TME which may be at play in CIN tumour settings. CAF, cancer-associated fibroblast; CIN, chromosomal instability; dsDNA, double-stranded DNA; IFN, interferon; MDSC, myeloid-derived suppressor cell; SASP, senescence-associated secretory phenotype; TAM, tumour-associated macrophage; TME, tumour microenvironment; Treg, regulatory T cell.

Close modal

Collectively, these routes help transmit cGAS–STING signal mediators, such as cGAMP and dsDNA, from chromosomally unstable tumour cells to neighbouring bystander cells via direct cell–cell contacts or across the extracellular space to more distant cells [143]. As such, cumulative STING activity within tumours is not just determined by the intrinsic cytosolic DNA burden and the expression of cGAS–STING components within cancer cells, but also by the composition of the TME and the capacity of its distinct cellular components to import and respond to cGAS–STING ligands (Figure 5b). This not only permits cGAS–STING activation within the TME of CIN-high tumours with defective DNA sensing pathways, but also offers ample opportunity for cancer cells to intercept or fine-tune cGAS–STING signal transmissions to their benefit. A notable example of this involves the breakdown of extracellular cGAMP by the cGAMP hydrolase ENPP1 [144, 145], which can drive the accumulation of the immune-suppressive metabolite adenosine [146], resulting in decreased immune infiltration and increased metastasis in CIN tumours [147].

Whilst additional mechanisms, such as the digestion of chromatin by the secreted deoxyribonuclease (DNase) I-family nucleases DNase I and DNase I-like 3 (DNase IL3), have been proposed to limit extracellular cGAS substrate availability [148, 149], it is unclear whether and to what extent they restrain cGAS–STING signal transmission to the TME by CIN tumours.

Whether chronic STING activation in tumours necessarily fosters a pro-tumorigenic TME remains unclear. A pan-cancer correlative analysis of TCGA datasets looking at the association between STING mRNA expression and immune cell infiltration revealed that STING expression correlates positively with infiltration of most immune cell types across cancers, rather than any one subtype [150]. This reflects that rather than recruiting specific subsets of cytotoxic or immunosuppressive immune cells into the tumour, STING exerts a broadly chemo-attractive effect, which may underlie its paradoxical role in the shaping of tumour landscapes. Indeed, aside from its well-established involvement in promoting anti-tumour immune responses [20], increasing evidence indicates that STING activity also plays a role in establishing a tolerogenic tumour microenvironment.

STING activity has been reported to promote the recruitment and/or induction of immune-suppressive myeloid-derived suppressor cell (MDSC) [35, 151–153] and regulatory T cell (Treg) [33, 154] populations, as well as the polarisation of monocytes to immune-suppressive ‘M2-like’ tumour-associated macrophages [155, 156]. In addition, chronic or over-stimulation of STING has been shown to selectively initiate anti-proliferative and pro-apoptotic programs in T cells [111, 129, 157–161], which, combined with the STING/IFN-dependent up-regulation of immune checkpoints, such as IDO1 and PD-L1, in the TME [12, 35, 162–167] may limit the overall anti-tumour potency of the T cell compartment.

As such, whilst transient STING induction appears favourable to anti-tumour immunity, prolonged activation can elicit immunoregulatory mechanisms that help tumours avoid immune-mediated destruction. Indeed, several correlative pan-cancer studies have related the degree of aneuploidy and CIN to increased markers of immune evasion and a decreased cytotoxic immune infiltrate [168–170], suggesting a role for CIN, albeit not necessarily CIN-driven cGAS–STING, in driving immunosuppression. However, the functional interaction between chronic cGAS–STING activation as a consequence of CIN and the tumour landscape has not been closely examined.

Crucially, aside from immune cells, cells of the non-immune stromal compartment, such as cancer-associated fibroblasts (CAFs) and endothelial cells, also act as key mediators and effectors of cGAS–STING signalling in the TME [142, 171–174]. However, despite evidence that higher tumour aneuploidy correlates with increased non-immune stromal fractions across several solid tumours [169], potential interactions between CIN-driven STING signalling and such stromal components remain unexplored. Further investigation of this and other aspects of cGAS–STING signalling in distinct compartments of the tumour microenvironment is likely to provide valuable insight enabling tailored targeting of cGAS–STING-driven signalling.

It is becoming increasingly clear that the outcomes of cell-intrinsic cGAS–STING-driven programs in CIN contexts are paradoxical in nature. Whereas in homeostatic and low genomic stress conditions they appear to promote survival mechanisms and genome stability, in excessively high CIN settings they can amplify CIN to catastrophic levels and bring about cell death (Figure 2). As such, cGAS–STING-driven genome-stabilising functions (e.g. DDR promotion, cell division control, autophagy) and anti-proliferative programs (e.g. DDR inhibition, mitotic death, autophagic death, senescence) appear to operate in concert as a multi-tiered cell-intrinsic safeguard against genome instability. Whether this clear parallel with the role of STING-driven inflammatory signalling as an immune-dependent genome surveillance mechanism [8, 9, 12] alludes to a primordial function for non-immune cell-intrinsic effects in genome surveillance remains unclear [61]. Whether and how genome surveillance mechanisms, which are primarily described to restrict the propagation of neoplastic cells, may benefit chromosomally unstable cancers also remain outstanding questions.

Cancer cells are proposed to exist within a narrow optimal range of chromosome mis-segregation rates, above which CIN becomes tumour suppressive [175–181]: a notion that is supported by observations that CIN and aneuploidy render tumour cells more vulnerable to treatments that elevate chromosome mis-segregation rates [182–185]. Given the emerging links between cGAS–STING and CIN in tumours [21, 114], it is tempting to speculate that by emphasising the CIN- and CIN-associated stress-limiting properties of cGAS–STING activation, chromosomally unstable cancers help maintain a window of CIN that is optimal to their survival. However, ascertaining to what extent this notion holds true or not will require further investigation.

  • Chromosomal instability is a key mechanism by which cancers gain resistance to therapy and promote metastasis. A paradox is identified whereby chromosomal instability constitutively activates cGAS–STING signalling — an innate immune pathway critical to immune surveillance.

  • Emerging evidence supports context-dependent pro-tumorigenic effects of cGAS–STING signalling, both on a cell-intrinsic and tumour microenvironment level.

  • Leveraging our understanding of the mechanisms of cancer-mediated cGAS–STING pathway control and how cGAS–STING signalling can be beneficial to the tumour could identify novel therapeutic targets in chromosomally unstable cancer.

The authors declare that there are no competing interests associated with the manuscript.

E.E.P. is supported by a Wellcome Trust Clinical Research Career Development Fellowship (224623/Z/21/Z). B.B. is funded by a Cancer Research UK (CRUK) Studentship (C2195/A31281).

Open access for this article was enabled by the participation of University of Oxford in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with JISC.

CIN

chromosomal instability

DDR

DNA damage response

DSB

double-strand break

EMT

epithelial–mesenchymal transition

HR

homologous recombination

IRDS

IFN-related DNA damage resistance signature

IRF3

interferon regulatory factor 3

ISGs

interferon-stimulated genes

MDSC

myeloid-derived suppressor cell

MN

micronuclei

SASP

senescence-associated secretory phenotype

STING

stimulator of interferon genes

TBK1

TANK-binding kinase 1

TCGA

The Cancer Genome Atlas

TME

tumour microenvironment

TNBC

triple-negative breast cancer

1
Bakhoum
,
S.F.
and
Cantley
,
L.C.
(
2018
)
The multifaceted role of chromosomal instability in cancer and its microenvironment
.
Cell
174
,
1347
1360
2
Fenech
,
M.
(
2007
)
Cytokinesis-block micronucleus cytome assay
.
Nat. Protoc.
2
,
1084
1104
3
Hatch
,
E.M.
,
Fischer
,
A.H.
,
Deerinck
,
T.J.
and
Hetzer
,
M.W.
(
2013
)
Catastrophic nuclear envelope collapse in cancer cell micronuclei
.
Cell
154
,
47
60
4
Sun
,
L.
,
Wu
,
J.
,
Du
,
F.
,
Chen
,
X.
and
Chen
,
Z.J.
(
2013
)
Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway
.
Science
339
,
786
791
5
Wu
,
J.
,
Sun
,
L.
,
Chen
,
X.
,
Du
,
F.
,
Shi
,
H.
,
Chen
,
C.
et al (
2013
)
Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA
.
Science
339
,
826
830
6
Ablasser
,
A.
,
Goldeck
,
M.
,
Cavlar
,
T.
,
Deimling
,
T.
,
Witte
,
G.
,
Röhl
,
I.
et al (
2013
)
cGAS produces a 2′-5′-linked cyclic dinucleotide second messenger that activates STING
.
Nature
498
,
380
384
7
Civril
,
F.
,
Deimling
,
T.
,
de Oliveira Mann
,
C.C.
,
Ablasser
,
A.
,
Moldt
,
M.
,
Witte
,
G.
et al (
2013
)
Structural mechanism of cytosolic DNA sensing by cGAS
.
Nature
498
,
332
337
8
Mackenzie
,
K.J.
,
Carroll
,
P.
,
Martin
,
C.-A.
,
Murina
,
O.
,
Fluteau
,
A.
,
Simpson
,
D.J.
et al (
2017
)
cGAS surveillance of micronuclei links genome instability to innate immunity
.
Nature
548
,
461
465
9
Harding
,
S.M.
,
Benci
,
J.L.
,
Irianto
,
J.
,
Discher
,
D.E.
,
Minn
,
A.J.
and
Greenberg
,
R.A.
(
2017
)
Mitotic progression following DNA damage enables pattern recognition within micronuclei
.
Nature
548
,
466
470
10
Bartsch
,
K.
,
Knittler
,
K.
,
Borowski
,
C.
,
Rudnik
,
S.
,
Damme
,
M.
,
Aden
,
K.
et al (
2017
)
Absence of RNase H2 triggers generation of immunogenic micronuclei removed by autophagy
.
Hum. Mol. Genet.
26
,
3960
3972
11
Flynn
,
P.J.
,
Koch
,
P.D.
and
Mitchison
,
T.J.
(
2021
)
Chromatin bridges, not micronuclei, activate cGAS after drug-induced mitotic errors in human cells
.
Proc. Natl Acad. Sci. U.S.A.
118
,
e2103585118
12
Parkes
,
E.E.
,
Walker
,
S.M.
,
Taggart
,
L.E.
,
McCabe
,
N.
,
Knight
,
L.A.
,
Wilkinson
,
R.
et al (
2017
)
Activation of STING-dependent innate immune signaling by S-phase-specific DNA damage in breast cancer
.
JNCI J. Natl Cancer Inst.
109
,
djw199
13
Bakhoum
,
S.F.
,
Ngo
,
B.
,
Laughney
,
A.M.
,
Cavallo
,
J.-A.
,
Murphy
,
C.J.
,
Ly
,
P.
et al (
2018
)
Chromosomal instability drives metastasis through a cytosolic DNA response
.
Nature
553
,
467
472
14
Ishikawa
,
H.
and
Barber
,
G.N.
(
2008
)
STING Is an endoplasmic reticulum adaptor that facilitates innate immune signalling
.
Nature
455
,
674
678
15
Abe
,
T.
and
Barber
,
G.N.
(
2014
)
Cytosolic-DNA-mediated, STING-dependent proinflammatory gene induction necessitates canonical NF-κB activation through TBK1
.
J. Virol.
88
,
5328
5341
16
Taniguchi
,
K.
and
Karin
,
M.
(
2018
)
NF-κB, inflammation, immunity and cancer: coming of age
.
Nat. Rev. Immunol.
18
,
309
324
17
Decout
,
A.
,
Katz
,
J.D.
,
Venkatraman
,
S.
and
Ablasser
,
A.
(
2021
)
The cGAS–STING pathway as a therapeutic target in inflammatory diseases
.
Nat. Rev. Immunol.
21
,
548
569
18
Woo
,
S.-R.
,
Fuertes
,
M.B.
,
Corrales
,
L.
,
Spranger
,
S.
,
Furdyna
,
M.J.
,
Leung
,
M.Y.K.
et al (
2014
)
STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors
.
Immunity
41
,
830
842
19
Corrales
,
L.
,
Glickman
,
L.H.
,
McWhirter
,
S.M.
,
Kanne
,
D.B.
,
Sivick
,
K.E.
,
Katibah
,
G.E.
et al (
2015
)
Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity
.
Cell Rep.
11
,
1018
1030
20
Zhu
,
Y.
,
An
,
X.
,
Zhang
,
X.
,
Qiao
,
Y.
,
Zheng
,
T.
and
Li
,
X.
(
2019
)
STING: a master regulator in the cancer-immunity cycle
.
Mol. Cancer
18
,
152
21
Hong
,
C.
,
Tijhuis
,
A.E.
and
Foijer
,
F.
(
2019
)
The cGAS paradox: contrasting roles for cGAS-STING pathway in chromosomal instability
.
Cells
8
,
E1228
22
Hu
,
S.
,
Fang
,
Y.
,
Chen
,
X.
,
Cheng
,
T.
,
Zhao
,
M.
,
Du
,
M.
et al (
2021
)
cGAS restricts colon cancer development by protecting intestinal barrier integrity
.
Proc. Natl. Acad. Sci. U.S.A.
118
,
e2105747118
23
Ahn
,
J.
,
Konno
,
H.
and
Barber
,
G.N.
(
2015
)
Diverse roles of STING-dependent signaling on the development of cancer
.
Oncogene
34
,
5302
5308
24
Zhu
,
Q.
,
Man
,
S.M.
,
Gurung
,
P.
,
Liu
,
Z.
,
Vogel
,
P.
,
Lamkanfi
,
M.
et al (
2014
)
STING mediates protection against colorectal tumorigenesis by governing the magnitude of intestinal inflammation
.
J. Immunol.
193
,
4779
4782
25
Konno
,
H.
,
Yamauchi
,
S.
,
Berglund
,
A.
,
Putney
,
R.M.
,
Mulé
,
J.J.
and
Barber
,
G.N.
(
2018
)
Suppression of STING signaling through epigenetic silencing and missense mutation impedes DNA damage mediated cytokine production
.
Oncogene
37
,
2037
2051
26
Low
,
J.T.
,
Chandramohan
,
V.
,
Bowie
,
M.L.
,
Brown
,
M.C.
,
Waitkus
,
M.S.
,
Briley
,
A.
et al (
2022
)
Epigenetic STING silencing is developmentally conserved in gliomas and can be rescued by methyltransferase inhibition
.
Cancer Cell
40
,
439
440
27
Xia
,
T.
,
Konno
,
H.
,
Ahn
,
J.
and
Barber
,
G.N.
(
2016
)
Deregulation of STING signaling in colorectal carcinoma constrains DNA damage responses and correlates with tumorigenesis
.
Cell Rep.
14
,
282
297
28
Falahat
,
R.
,
Berglund
,
A.
,
Putney
,
R.M.
,
Perez-Villarroel
,
P.
,
Aoyama
,
S.
,
Pilon-Thomas
,
S.
et al (
2021
)
Epigenetic reprogramming of tumor cell–intrinsic STING function sculpts antigenicity and T cell recognition of melanoma
.
Proc. Natl. Acad. Sci. U.S.A.
118
,
e2013598118
29
Xia
,
T.
,
Konno
,
H.
and
Barber
,
G.N.
(
2016
)
Recurrent loss of STING signaling in melanoma correlates with susceptibility to viral oncolysis
.
Cancer Res.
76
,
6747
6759
30
Song
,
S.
,
Peng
,
P.
,
Tang
,
Z.
,
Zhao
,
J.
,
Wu
,
W.
,
Li
,
H.
et al (
2017
)
Decreased expression of STING predicts poor prognosis in patients with gastric cancer
.
Sci. Rep.
7
,
39858
31
Kitajima
,
S.
,
Ivanova
,
E.
,
Guo
,
S.
,
Yoshida
,
R.
,
Campisi
,
M.
,
Sundararaman
,
S.K.
et al (
2019
)
Suppression of STING associated with LKB1 loss in KRAS-driven lung cancer
.
Cancer Discov.
9
,
34
45
32
de Queiroz
,
N.M.G.P.
,
Xia
,
T.
,
Konno
,
H.
and
Barber
,
G.N.
(
2019
)
Ovarian cancer cells commonly exhibit defective STING signaling which affects sensitivity to viral oncolysis
.
Mol. Cancer Res.
17
,
974
986
33
Liang
,
D.
,
Xiao-Feng
,
H.
,
Guan-Jun
,
D.
,
Er-Ling
,
H.
,
Sheng
,
C.
,
Ting-Ting
,
W.
et al (
2015
)
Activated STING enhances tregs infiltration in the HPV-related carcinogenesis of tongue squamous cells via the c-jun/CCL22 signal
.
Biochim. Biophys. Acta Mol. Basis Dis.
1852
,
2494
2503
34
Ahn
,
J.
,
Xia
,
T.
,
Konno
,
H.
,
Konno
,
K.
,
Ruiz
,
P.
and
Barber
,
G.N.
(
2014
)
Inflammation-driven carcinogenesis is mediated through STING
.
Nat. Commun.
5
,
5166
35
Lemos
,
H.
,
Mohamed
,
E.
,
Huang
,
L.
,
Ou
,
R.
,
Pacholczyk
,
G.
,
Arbab
,
A.S.
et al (
2016
)
STING promotes the growth of tumors characterized by low antigenicity via IDO activation
.
Cancer Research
76
,
2076
2081
36
Hong
,
C.
,
Schubert
,
M.
,
Tijhuis
,
A.E.
,
Requesens
,
M.
,
Roorda
,
M.
,
van den Brink
,
A.
et al (
2022
)
cGAS–STING drives the IL-6-dependent survival of chromosomally instable cancers
.
Nature
607
,
366
373
37
Chen
,
Q.
,
Boire
,
A.
,
Jin
,
X.
,
Valiente
,
M.
,
Er
,
E.E.
,
Lopez-Soto
,
A.
et al (
2016
)
Carcinoma–astrocyte gap junctions promote brain metastasis by cGAMP transfer
.
Nature
533
,
493
498
38
Santaguida
,
S.
,
Richardson
,
A.
,
Iyer
,
D.R.
,
M'Saad
,
O.
,
Zasadil
,
L.
,
Knouse
,
K.A.
et al (
2017
)
Chromosome mis-segregation generates cell-cycle-arrested cells with complex karyotypes that are eliminated by the immune system
.
Dev. Cell
41
,
638
651.e5
39
Khodarev
,
N.N.
,
Beckett
,
M.
,
Labay
,
E.
,
Darga
,
T.
,
Roizman
,
B.
and
Weichselbaum
,
R.R.
(
2004
)
STAT1 is overexpressed in tumors selected for radioresistance and confers protection from radiation in transduced sensitive cells
.
Proc. Natl Acad. Sci. U.S.A.
101
,
1714
1719
40
Weichselbaum
,
R.R.
,
Ishwaran
,
H.
,
Yoon
,
T.
,
Nuyten
,
D.S.A.
,
Baker
,
S.W.
,
Khodarev
,
N.
et al (
2008
)
An interferon-related gene signature for DNA damage resistance is a predictive marker for chemotherapy and radiation for breast cancer
.
Proc. Natl Acad. Sci. U.S.A.
105
,
18490
18495
41
Cheon
,
H.
,
Wang
,
Y.
,
Wightman
,
S.M.
,
Jackson
,
M.W.
and
Stark
,
G.R.
(
2022
)
How cancer cells make and respond to interferon-I
.
Trends Cancer
9
,
83
92
42
Erdal
,
E.
,
Haider
,
S.
,
Rehwinkel
,
J.
,
Harris
,
A.L.
and
McHugh
,
P.J.
(
2017
)
A prosurvival DNA damage-induced cytoplasmic interferon response is mediated by end resection factors and is limited by Trex1
.
Genes Dev.
31
,
353
369
43
Gaston
,
J.
,
Cheradame
,
L.
,
Yvonnet
,
V.
,
Deas
,
O.
,
Poupon
,
M.-F.
,
Judde
,
J.-G.
et al (
2016
)
Intracellular STING inactivation sensitizes breast cancer cells to genotoxic agents
.
Oncotarget
7
,
77205
77224
44
Cheon
,
H.
,
Holvey-Bates
,
E.G.
McGrail
,
D.J.
and
Stark
,
G.R.
(
2021
)
PD-L1 sustains chronic, cancer cell–intrinsic responses to type I interferon, enhancing resistance to DNA damage
.
Proc. Natl Acad. Sci. U.S.A.
118
,
e2112258118
45
Gaston
,
J.
,
Cheradame
,
L.
,
Yvonnet
,
V.
,
Deas
,
O.
,
Poupon
,
M.-F.
,
Judde
,
J.-G.
et al (
2019
)
Correction: intracellular STING inactivation sensitizes breast cancer cells to genotoxic agents
.
Oncotarget
10
,
4249
4251
46
Wu
,
Z.
,
Oeck
,
S.
,
West
,
A.P.
,
Mangalhara
,
K.C.
,
Sainz
,
A.G.
,
Newman
,
L.E.
et al (
2019
)
Mitochondrial DNA stress signalling protects the nuclear genome
.
Nat. Metab.
1
,
1209
1218
47
Perng
,
Y.-C.
and
Lenschow
,
D.J.
(
2018
)
ISG15 in antiviral immunity and beyond
.
Nat. Rev. Microbiol.
16
,
423
439
48
Raso
,
M.C.
,
Djoric
,
N.
,
Walser
,
F.
,
Hess
,
S.
,
Schmid
,
F.M.
,
Burger
,
S.
et al (
2020
)
Interferon-stimulated gene 15 accelerates replication fork progression inducing chromosomal breakage
.
J. Cell Biol.
219
,
e202002175
49
Park
,
J.M.
,
Yang
,
S.W.
,
Yu
,
K.R.
,
Ka
,
S.H.
,
Lee
,
S.W.
,
Seol
,
J.H.
et al (
2014
)
Modification of PCNA by ISG15 plays a crucial role in termination of error-prone translesion DNA synthesis
.
Mol. Cell
54
,
626
638
50
Jeon
,
Y.J.
,
Jo
,
M.G.
,
Yoo
,
H.M.
,
Hong
,
S.-H.
,
Park
,
J.-M.
,
Ka
,
S.H.
et al (
2012
)
Chemosensitivity is controlled by p63 modification with ubiquitin-like protein ISG15
.
J. Clin. Invest.
122
,
2622
2636
51
Sandy
,
Z.
,
da Costa
,
I.C.
and
Schmidt
,
C.K.
(
2020
)
More than meets the ISG15: emerging roles in the DNA damage response and beyond
.
Biomolecules
10
,
1557
52
Wardlaw
,
C.P.
and
Petrini
,
J.H.J.
(
2022
)
ISG15 conjugation to proteins on nascent DNA mitigates DNA replication stress
.
Nat. Commun.
13
,
5971
53
Park
,
J.H.
,
Yang
,
S.W.
,
Park
,
J.M.
,
Ka
,
S.H.
,
Kim
,
J.-H.
,
Kong
,
Y.-Y.
et al (
2016
)
Positive feedback regulation of p53 transactivity by DNA damage-induced ISG15 modification
.
Nat. Commun.
7
,
12513
54
Hayman
,
T.J.
and
Glazer
,
P.M.
(
2021
)
Regulation of the cell-intrinsic DNA damage response by the innate immune machinery
.
Int. J. Mol. Sci.
22
,
12761
55
Budke
,
B.
,
Zhong
,
A.
,
Sullivan
,
K.
,
Park
,
C.
,
Gittin
,
D.I.
,
Kountz
,
T.S.
et al (
2022
)
Noncanonical NF-κB factor p100/p52 regulates homologous recombination and modulates sensitivity to DNA-damaging therapy
.
Nucleic Acids Res.
50
,
6251
6263
56
Volcic
,
M.
,
Karl
,
S.
,
Baumann
,
B.
,
Salles
,
D.
,
Daniel
,
P.
,
Fulda
,
S.
et al (
2012
)
NF-κB regulates DNA double-strand break repair in conjunction with BRCA1–CtIP complexes
.
Nucleic Acids Res.
40
,
181
195
57
Cron
,
K.R.
,
Zhu
,
K.
,
Kushwaha
,
D.S.
,
Hsieh
,
G.
,
Merzon
,
D.
,
Rameseder
,
J.
et al (
2013
)
Proteasome inhibitors block DNA repair and radiosensitize non-small cell lung cancer
.
PLoS ONE
8
,
e73710
58
Harte
,
M.T.
,
Gorski
,
J.J.
,
Savage
,
K.I.
,
Purcell
,
J.W.
,
Barros
,
E.M.
,
Burn
,
P.M.
et al (
2014
)
NF-κB Is a critical mediator of BRCA1-induced chemoresistance
.
Oncogene
33
,
713
723
59
Deraska
,
P.V.
,
O'Leary
,
C.
,
Reavis
,
H.D.
,
Labe
,
S.
,
Dinh
,
T.-K.
,
Lazaro
,
J.-B.
et al (
2018
)
NF-κB inhibition by dimethylaminoparthenolide radiosensitizes non-small-cell lung carcinoma by blocking DNA double-strand break repair
.
Cell Death Discov.
4
,
10
60
Banerjee
,
D.
,
Langberg
,
K.
,
Abbas
,
S.
,
Odermatt
,
E.
,
Yerramothu
,
P.
,
Volaric
,
M.
et al (
2021
)
A non-canonical, interferon-independent signaling activity of cGAMP triggers DNA damage response signaling
.
Nat. Commun.
12
,
6207
61
de Oliveira Mann
,
C.C.
and
Hopfner
,
K.-P.
(
2021
)
Nuclear cGAS: guard or prisoner?
EMBO J.
40
,
e108293
62
Liu
,
H.
,
Zhang
,
H.
,
Wu
,
X.
,
Ma
,
D.
,
Wu
,
J.
,
Wang
,
L.
et al (
2018
)
Nuclear cGAS suppresses DNA repair and promotes tumorigenesis
.
Nature
563
,
131
136
63
Jiang
,
H.
,
Xue
,
X.
,
Panda
,
S.
,
Kawale
,
A.
,
Hooy
,
R.M.
,
Liang
,
F.
et al (
2019
)
Chromatin-bound cGAS is an inhibitor of DNA repair and hence accelerates genome destabilization and cell death
.
EMBO J.
38
,
e102718
64
Chen
,
H.
,
Chen
,
H.
,
Zhang
,
J.
,
Wang
,
Y.
,
Simoneau
,
A.
,
Yang
,
H.
et al (
2020
)
cGAS suppresses genomic instability as a decelerator of replication forks
.
Sci Adv
6
,
eabb8941
65
Li
,
X.
,
Li
,
X.
,
Xie
,
C.
,
Cai
,
S.
,
Li
,
M.
,
Jin
,
H.
et al (
2022
)
cGAS guards against chromosome end-to-end fusions during mitosis and facilitates replicative senescence
.
Protein Cell
13
,
47
64
66
Cheradame
,
L.
,
Guerrera
,
I.C.
,
Gaston
,
J.
,
Schmitt
,
A.
,
Jung
,
V.
,
Goudin
,
N.
et al (
2021
)
STING protects breast cancer cells from intrinsic and genotoxic-induced DNA instability via a non-canonical, cell-autonomous pathway
.
Oncogene
40
,
6627
6640
67
Ranoa
,
D.R.E.
,
Widau
,
R.C.
,
Mallon
,
S.
,
Parekh
,
A.D.
,
Nicolae
,
C.M.
,
Huang
,
X.
et al (
2019
)
STING promotes homeostasis via regulation of cell proliferation and chromosomal stability
.
Cancer Res.
79
,
1465
1479
68
Basit
,
A.
,
Cho
,
M.-G.
,
Kim
,
E.-Y.
,
Kwon
,
D.
,
Kang
,
S.-J.
and
Lee
,
J.-H.
(
2020
)
The cGAS/STING/TBK1/IRF3 innate immunity pathway maintains chromosomal stability through regulation of p21 levels
.
Exp. Mol. Med.
52
,
643
657
69
Li
,
T.
and
Chen
,
Z.J.
(
2018
)
The cGAS–cGAMP–STING pathway connects DNA damage to inflammation, senescence, and cancer
.
J. Exp. Med.
215
,
1287
1299
70
Glück
,
S.
,
Guey
,
B.
,
Gulen
,
M.F.
,
Wolter
,
K.
,
Kang
,
T.-W.
,
Schmacke
,
N.A.
et al (
2017
)
Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence
.
Nat. Cell Biol.
19
,
1061
1070
71
Yang
,
H.
,
Wang
,
H.
,
Ren
,
J.
,
Chen
,
Q.
and
Chen
,
Z.J.
(
2017
)
cGAS is essential for cellular senescence
.
Proc. Natl Acad. Sci. U.S.A.
114
,
E4612
E4620
72
Dou
,
Z.
,
Ghosh
,
K.
,
Vizioli
,
M.G.
,
Zhu
,
J.
,
Sen
,
P.
,
Wangensteen
,
K.J.
et al (
2017
)
Cytoplasmic chromatin triggers inflammation in senescence and cancer
.
Nature
550
,
402
406
73
Wajapeyee
,
N.
,
Serra
,
R.W.
,
Zhu
,
X.
,
Mahalingam
,
M.
and
Green
,
M.R.
(
2008
)
Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7
.
Cell
132
,
363
374
74
Acosta
,
J.C.
,
Banito
,
A.
,
Wuestefeld
,
T.
,
Georgilis
,
A.
,
Janich
,
P.
,
Morton
,
J.P.
et al (
2013
)
A complex secretory program orchestrated by the inflammasome controls paracrine senescence
.
Nat. Cell Biol.
15
,
978
990
75
Acosta
,
J.C.
,
O'Loghlen
,
A.
,
Banito
,
A.
,
Guijarro
,
M.V.
,
Augert
,
A.
,
Raguz
,
S.
et al (
2008
)
Chemokine signaling via the CXCR2 receptor reinforces senescence
.
Cell
133
,
1006
1018
76
Kuilman
,
T.
,
Michaloglou
,
C.
,
Vredeveld
,
L.C.W.
,
Douma
,
S.
,
van Doorn
,
R.
,
Desmet
,
C.J.
et al (
2008
)
Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network
.
Cell
133
,
1019
1031
77
Vessoni
,
A.T.
,
Filippi-Chiela
,
E.C.
,
Menck
,
C.F.
and
Lenz
,
G.
(
2013
)
Autophagy and genomic integrity
.
Cell Death Differ.
20
,
1444
1454
78
Vasudevan
,
A.
,
Schukken
,
K.M.
,
Sausville
,
E.L.
,
Girish
,
V.
,
Adebambo
,
O.A.
and
Sheltzer
,
J.M.
(
2021
)
Aneuploidy as a promoter and suppressor of malignant growth
.
Nat. Rev. Cancer
21
,
89
103
79
Stingele
,
S.
,
Stoehr
,
G.
,
Peplowska
,
K.
,
Cox
,
J.
,
Mann
,
M.
and
Storchova
,
Z.
(
2012
)
Global analysis of genome, transcriptome and proteome reveals the response to aneuploidy in human cells
.
Mol. Syst. Biol.
8
,
608
80
Santaguida
,
S.
,
Vasile
,
E.
,
White
,
E.
and
Amon
,
A.
(
2015
)
Aneuploidy-induced cellular stresses limit autophagic degradation
.
Genes Dev.
29
,
2010
2021
81
Buccitelli
,
C.
,
Salgueiro
,
L.
,
Rowald
,
K.
,
Sotillo
,
R.
,
Mardin
,
B.R.
and
Korbel
,
J.O.
(
2017
)
Pan-cancer analysis distinguishes transcriptional changes of aneuploidy from proliferation
.
Genome Res.
27
,
501
511
82
Tang
,
Y.-C.
,
Williams
,
B.R.
,
Siegel
,
J.J.
and
Amon
,
A.
(
2011
)
Identification of aneuploidy-selective antiproliferation compounds
.
Cell
144
,
499
512
83
Liu
,
D.
,
Shaukat
,
Z.
,
Xu
,
T.
,
Denton
,
D.
,
Saint
,
R.
and
Gregory
,
S.
(
2016
)
Autophagy regulates the survival of cells with chromosomal instability
.
Oncotarget
7
,
63913
63923
84
Konno
,
H.
,
Konno
,
K.
and
Barber
,
G.N.
(
2013
)
Cyclic dinucleotides trigger ULK1 (ATG1) phosphorylation of STING to prevent sustained innate immune signaling
.
Cell
155
,
688
698
85
Prabakaran
,
T.
,
Bodda
,
C.
,
Krapp
,
C.
,
Zhang
,
B.
,
Christensen
,
M.H.
,
Sun
,
C.
et al (
2018
)
Attenuation of cGAS-STING signaling is mediated by a p62/SQSTM1-dependent autophagy pathway activated by TBK1
.
EMBO J.
37
,
e97858
86
Chen
,
M.
,
Meng
,
Q.
,
Qin
,
Y.
,
Liang
,
P.
,
Tan
,
P.
,
He
,
L.
et al (
2016
)
TRIM14 inhibits cGAS degradation mediated by selective autophagy receptor p62 to promote innate immune responses
.
Mol. Cell
64
,
105
119
87
Liang
,
Q.
,
Seo
,
G.J.
,
Choi
,
Y.J.
,
Kwak
,
M.-J.
,
Ge
,
J.
,
Rodgers
,
M.A.
et al (
2014
)
Crosstalk between the cGAS DNA sensor and beclin-1 autophagy protein shapes innate antimicrobial immune responses
.
Cell Host Microbe
15
,
228
238
88
Saitoh
,
T.
,
Fujita
,
N.
,
Hayashi
,
T.
,
Takahara
,
K.
,
Satoh
,
T.
,
Lee
,
H.
et al (
2009
)
Atg9a controls dsDNA-driven dynamic translocation of STING and the innate immune response
.
Proc. Natl Acad. Sci. U.S.A.
106
,
20842
20846
89
Zhang
,
K.
,
Wang
,
S.
,
Gou
,
H.
,
Zhang
,
J.
and
Li
,
C.
(
2021
)
Crosstalk between autophagy and the cGAS–STING signaling pathway in type I interferon production
.
Front. Cell Dev. Biol.
9
,
748485
90
Liu
,
D.
,
Wu
,
H.
,
Wang
,
C.
,
Li
,
Y.
,
Tian
,
H.
,
Siraj
,
S.
et al (
2019
)
STING directly activates autophagy to tune the innate immune response
.
Cell Death Differ.
26
,
1735
1749
91
Gonugunta
,
V.K.
,
Sakai
,
T.
,
Pokatayev
,
V.
,
Yang
,
K.
,
Wu
,
J.
,
Dobbs
,
N.
et al (
2017
)
Trafficking-mediated STING degradation requires sorting to acidified endolysosomes and can be targeted to enhance anti-tumor response
.
Cell Rep.
21
,
3234
3242
92
Gui
,
X.
,
Yang
,
H.
,
Li
,
T.
,
Tan
,
X.
,
Shi
,
P.
,
Li
,
M.
et al (
2019
)
Autophagy induction via STING trafficking is a primordial function of the cGAS pathway
.
Nature
567
,
262
266
93
Krivega
,
M.
,
Stiefel
,
C.M.
,
Karbassi
,
S.
,
Andersen
,
L.L.
,
Chunduri
,
N.K.
,
Donnelly
,
N.
et al (
2021
)
Genotoxic stress in constitutive trisomies induces autophagy and the innate immune response via the cGAS-STING pathway
.
Commun. Biol.
4
,
831
94
Lan
,
Y.Y.
,
Londoño
,
D.
,
Bouley
,
R.
,
Rooney
,
M.S.
and
Hacohen
,
N.
(
2014
)
Dnase2a deficiency uncovers lysosomal clearance of damaged nuclear DNA via autophagy
.
Cell Rep.
9
,
180
192
95
Ivanov
,
A.
,
Pawlikowski
,
J.
,
Manoharan
,
I.
,
van Tuyn
,
J.
,
Nelson
,
D.M.
,
Rai
,
T.S.
et al (
2013
)
Lysosome-mediated processing of chromatin in senescence
.
J. Cell Biol.
202
,
129
143
96
Guo
,
H.
,
Chitiprolu
,
M.
,
Gagnon
,
D.
,
Meng
,
L.
,
Perez-Iratxeta
,
C.
,
Lagace
,
D.
et al (
2014
)
Autophagy supports genomic stability by degrading retrotransposon RNA
.
Nat. Commun.
5
,
5276
97
Yao
,
M.
,
Wu
,
Y.
,
Cao
,
Y.
,
Liu
,
H.
,
Ma
,
N.
,
Chai
,
Y.
et al (
2021
)
Autophagy-mediated clearance of free genomic DNA in the cytoplasm protects the growth and survival of cancer cells
.
Front. Oncol.
11
,
1927
98
Rello-Varona
,
S.
,
Lissa
,
D.
,
Shen
,
S.
,
Niso-Santano
,
M.
,
Senovilla
,
L.
,
Mariño
,
G.
et al (
2012
)
Autophagic removal of micronuclei
.
Cell Cycle
11
,
170
176
99
Crasta
,
K.
,
Ganem
,
N.J.
,
Dagher
,
R.
,
Lantermann
,
A.B.
,
Ivanova
,
E.V.
,
Pan
,
Y.
et al (
2012
)
DNA breaks and chromosome pulverization from errors in mitosis
.
Nature
482
,
53
58
100
Zhang
,
C.-Z.
,
Spektor
,
A.
,
Cornils
,
H.
,
Francis
,
J.M.
,
Jackson
,
E.K.
,
Liu
,
S.
et al (
2015
)
Chromothripsis from DNA damage in micronuclei
.
Nature
522
,
179
184
101
Karantza-Wadsworth
,
V.
,
Patel
,
S.
,
Kravchuk
,
O.
,
Chen
,
G.
,
Mathew
,
R.
,
Jin
,
S.
et al (
2007
)
Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis
.
Genes Dev.
21
,
1621
1635
102
Mathew
,
R.
,
Kongara
,
S.
,
Beaudoin
,
B.
,
Karp
,
C.M.
,
Bray
,
K.
,
Degenhardt
,
K.
et al (
2007
)
Autophagy suppresses tumor progression by limiting chromosomal instability
.
Genes Dev.
21
,
1367
1381
103
Zhao
,
M.
,
Wang
,
F.
,
Wu
,
J.
,
Cheng
,
Y.
,
Cao
,
Y.
,
Wu
,
X.
et al (
2021
)
CGAS is a micronucleophagy receptor for the clearance of micronuclei
.
Autophagy
17
,
3976
3991
104
Watson
,
R.O.
,
Bell
,
S.L.
,
MacDuff
,
D.A.
,
Kimmey
,
J.M.
,
Diner
,
E.J.
,
Olivas
,
J.
et al (
2015
)
The cytosolic sensor cGAS detects Mycobacterium tuberculosis DNA to induce type I interferons and activate autophagy
.
Cell Host Microbe
17
,
811
819
105
Denton
,
D.
and
Kumar
,
S.
(
2019
)
Autophagy-dependent cell death
.
Cell Death Differ.
26
,
605
616
106
Nassour
,
J.
,
Radford
,
R.
,
Correia
,
A.
,
Fusté
,
J.M.
,
Schoell
,
B.
,
Jauch
,
A.
et al (
2019
)
Autophagic cell death restricts chromosomal instability during replicative crisis
.
Nature
565
,
659
663
107
Ku
,
J.W.K.
,
Chen
,
Y.
,
Lim
,
B.J.W.
,
Gasser
,
S.
,
Crasta
,
K.C.
and
Gan
,
Y.-H.
(
2020
)
Bacterial-induced cell fusion is a danger signal triggering cGAS–STING pathway via micronuclei formation
.
Proc. Natl Acad. Sci. U.S.A.
117
,
15923
15934
108
Cadwell
,
K.
(
2016
)
Crosstalk between autophagy and inflammatory signalling pathways: balancing defence and homeostasis
.
Nat. Rev. Immunol.
16
,
661
675
109
Klionsky
,
D.J.
,
Petroni
,
G.
,
Amaravadi
,
R.K.
,
Baehrecke
,
E.H.
,
Ballabio
,
A.
,
Boya
,
P.
et al (
2021
)
Autophagy in major human diseases
.
EMBO J.
40
,
e108863
110
Zierhut
,
C.
,
Yamaguchi
,
N.
,
Paredes
,
M.
,
Luo
,
J.-D.
,
Carroll
,
T.
and
Funabiki
,
H.
(
2019
)
The cytoplasmic DNA sensor cGAS promotes mitotic cell death
.
Cell
178
,
302
315.e23
111
Gulen
,
M.F.
,
Koch
,
U.
,
Haag
,
S.M.
,
Schuler
,
F.
,
Apetoh
,
L.
,
Villunger
,
A.
et al (
2017
)
Signalling strength determines proapoptotic functions of STING
.
Nat. Commun.
8
,
427
112
Kabelitz
,
D.
,
Zarobkiewicz
,
M.
,
Heib
,
M.
,
Serrano
,
R.
,
Kunz
,
M.
,
Chitadze
,
G.
et al (
2022
)
Signal strength of STING activation determines cytokine plasticity and cell death in human monocytes
.
Sci. Rep.
12
,
17827
113
Coussens
,
L.M.
and
Werb
,
Z.
(
2002
)
Inflammation and cancer
.
Nature
420
,
860
867
114
Tijhuis
,
A.E.
,
Johnson
,
S.C.
and
McClelland
,
S.E.
(
2019
)
The emerging links between chromosomal instability (CIN), metastasis, inflammation and tumour immunity
.
Mol. Cytogenet.
12
,
17
115
Wang
,
J.
,
Yi
,
S.
,
Zhou
,
J.
,
Zhang
,
Y.
and
Guo
,
F.
(
2016
)
The NF-κB subunit RelB regulates the migration and invasion abilities and the radio-sensitivity of prostate cancer cells
.
Int. J. Oncol.
49
,
381
392
116
Yang
,
C.
,
Davis
,
J.L.
,
Zeng
,
R.
,
Vora
,
P.
,
Su
,
X.
,
Collins
,
L.I.
et al (
2013
)
Antagonism of inhibitor of apoptosis proteins increases bone metastasis via unexpected osteoclast activation
.
Cancer Discov.
3
,
212
223
117
Bakhoum
,
M.F.
,
Francis
,
J.H.
,
Agustinus
,
A.
,
Earlie
,
E.M.
,
Di Bona
,
M.
,
Abramson
,
D.H.
et al (
2021
)
Loss of polycomb repressive complex 1 activity and chromosomal instability drive uveal melanoma progression
.
Nat. Commun.
12
,
5402
118
Wörmann
,
S.M.
,
Zhang
,
A.
,
Thege
,
F.I.
,
Cowan
,
R.W.
,
Rupani
,
D.N.
,
Wang
,
R.
et al (
2021
)
APOBEC3A drives deaminase domain-independent chromosomal instability to promote pancreatic cancer metastasis
.
Nat. Cancer
2
,
1338
1356
119
Vasudevan
,
A.
,
Baruah
,
P.S.
,
Smith
,
J.C.
,
Wang
,
Z.
,
Sayles
,
N.M.
,
Andrews
,
P.
et al (
2020
)
Single-chromosomal gains can function as metastasis suppressors and promoters in colon cancer
.
Dev. Cell
52
,
413
428.e6
120
Yu
,
H.
,
Pardoll
,
D.
and
Jove
,
R.
(
2009
)
STATs in cancer inflammation and immunity: a leading role for STAT3
.
Nat. Rev. Cancer
9
,
798
809
121
Johnson
,
D.E.
,
O'Keefe
,
R.A.
and
Grandis
,
J.R.
(
2018
)
Targeting the IL-6/JAK/STAT3 signalling axis in cancer
.
Nat. Rev. Clin. Oncol.
15
,
234
248
122
Mayca Pozo
,
F.
,
Geng
,
X.
,
Tamagno
,
I.
,
Jackson
,
M.W.
,
Heimsath
,
E.G.
Hammer
,
J.A.
et al (
2021
)
MYO10 drives genomic instability and inflammation in cancer
.
Sci. Adv.
7
,
eabg6908
123
Hou
,
Y.
,
Liang
,
H.
,
Rao
,
E.
,
Zheng
,
W.
,
Huang
,
X.
,
Deng
,
L.
et al (
2018
)
Non-canonical NF-κB antagonizes STING sensor-mediated DNA sensing in radiotherapy
.
Immunity
49
,
490
503.e4
124
Pei
,
J.
,
Zhang
,
Y.
,
Luo
,
Q.
,
Zheng
,
W.
,
Li
,
W.
,
Zeng
,
X.
et al (
2019
)
STAT3 inhibition enhances CDN-induced STING signaling and antitumor immunity
.
Cancer Lett.
450
,
110
122
125
van Vugt
,
M.A.T.M.
and
Parkes
,
E.E.
(
2022
)
When breaks get hot: inflammatory signaling in BRCA1/2-mutant cancers
.
Trends Cancer
8
,
174
189
126
Zheng
,
Z.
,
Jia
,
S.
,
Shao
,
C.
and
Shi
,
Y.
(
2020
)
Irradiation induces cancer lung metastasis through activation of the cGAS–STING–CCL5 pathway in mesenchymal stromal cells
.
Cell Death Dis.
11
,
326
127
Zhou
,
C.
,
Chen
,
X.
,
Planells-Cases
,
R.
,
Chu
,
J.
,
Wang
,
L.
,
Cao
,
L.
et al (
2020
)
Transfer of cGAMP into bystander cells via LRRC8 volume-regulated anion channels augments STING-mediated interferon responses and anti-viral immunity
.
Immunity
52
,
767
781.e6
128
Lahey
,
L.J.
,
Mardjuki
,
R.E.
,
Wen
,
X.
,
Hess
,
G.T.
,
Ritchie
,
C.
,
Carozza
,
J.A.
et al (
2020
)
LRRC8A:C/E heteromeric channels are ubiquitous transporters of cGAMP
.
Mol. Cell
80
,
578
591.e5
129
Concepcion
,
A.R.
,
Wagner
,
L.E.
,
Zhu
,
J.
,
Tao
,
A.Y.
,
Yang
,
J.
,
Khodadadi-Jamayran
,
A.
et al (
2022
)
The volume-regulated anion channel LRRC8C suppresses T cell function by regulating cyclic dinucleotide transport and STING–p53 signaling
.
Nat. Immunol.
23
,
287
302
130
Maltbaek
,
J.H.
,
Cambier
,
S.
,
Snyder
,
J.M.
and
Stetson
,
D.B.
(
2022
)
ABCC1 transporter exports the immunostimulatory cyclic dinucleotide cGAMP
.
Immunity
55
,
1799
1812.e4
131
Ritchie
,
C.
,
Cordova
,
A.F.
,
Hess
,
G.T.
,
Bassik
,
M.C.
and
Li
,
L.
(
2019
)
SLC19A1 is an importer of the immunotransmitter cGAMP
.
Molecular Cell
75
,
372
381.e5
132
Luteijn
,
R.D.
,
Zaver
,
S.A.
,
Gowen
,
B.G.
,
Wyman
,
S.K.
,
Garelis
,
N.E.
,
Onia
,
L.
et al (
2019
)
SLC19A1 transports immunoreactive cyclic dinucleotides
.
Nature
573
,
434
438
133
Zhou
,
Y.
,
Fei
,
M.
,
Zhang
,
G.
,
Liang
,
W.-C.
,
Lin
,
W.
,
Wu
,
Y.
et al (
2020
)
Blockade of the phagocytic receptor MerTK on tumor-associated macrophages enhances P2X7R-dependent STING activation by tumor-derived cGAMP
.
Immunity
52
,
357
373.e9
134
Ablasser
,
A.
,
Schmid-Burgk
,
J.L.
,
Hemmerling
,
I.
,
Horvath
,
G.L.
,
Schmidt
,
T.
,
Latz
,
E.
et al (
2013
)
Cell intrinsic immunity spreads to bystander cells via the intercellular transfer of cGAMP
.
Nature
503
,
530
534
135
Pépin
,
G.
,
De Nardo
,
D.
,
Rootes
,
C.L.
,
Ullah
,
T.R.
,
Al-Asmari
,
S.S.
,
Balka
,
K.R.
et al (
2020
)
Connexin-dependent transfer of cGAMP to phagocytes modulates antiviral responses
.
mBio
11
,
e03187-19
136
Schadt
,
L.
,
Sparano
,
C.
,
Schweiger
,
N.A.
,
Silina
,
K.
,
Cecconi
,
V.
,
Lucchiari
,
G.
et al (
2019
)
Cancer-cell-intrinsic cGAS expression mediates tumor immunogenicity
.
Cell Rep.
29
,
1236
1248.e7
137
Luther
,
J.
,
Khan
,
S.
,
Gala
,
M.K.
,
Kedrin
,
D.
,
Sridharan
,
G.
,
Goodman
,
R.P.
et al (
2020
)
Hepatic gap junctions amplify alcohol liver injury by propagating cGAS-mediated IRF3 activation
.
Proc. Natl Acad. Sci. U.S.A.
117
,
11667
11673
138
Torralba
,
D.
,
Baixauli
,
F.
,
Villarroya-Beltri
,
C.
,
Fernández-Delgado
,
I.
,
Latorre-Pellicer
,
A.
,
Acín-Pérez
,
R.
et al (
2018
)
Priming of dendritic cells by DNA-containing extracellular vesicles from activated T cells through antigen-driven contacts
.
Nat. Commun.
9
,
2658
139
Ahn
,
J.
,
Xia
,
T.
,
Rabasa Capote
,
A.
,
Betancourt
,
D.
and
Barber
,
G.N.
(
2018
)
Extrinsic phagocyte-dependent STING signaling dictates the immunogenicity of dying cells
.
Cancer Cell
33
,
862
873.e5
140
Xu
,
M.M.
,
Pu
,
Y.
,
Han
,
D.
,
Shi
,
Y.
,
Cao
,
X.
,
Liang
,
H.
et al (
2017
)
Dendritic cells but not macrophages sense tumor mitochondrial DNA for cross-priming through signal regulatory protein α signaling
.
Immunity
47
,
363
373.e5
141
de Mingo Pulido
,
Á.
,
Hänggi
,
K.
,
Celias
,
D.P.
,
Gardner
,
A.
,
Li
,
J.
,
Batista-Bittencourt
,
B.
et al (
2021
)
The inhibitory receptor TIM-3 limits activation of the cGAS-STING pathway in intra-tumoral dendritic cells by suppressing extracellular DNA uptake
.
Immunity
54
,
1154
1167.e7
142
Arwert
,
E.N.
,
Milford
,
E.L.
,
Rullan
,
A.
,
Derzsi
,
S.
,
Hooper
,
S.
,
Kato
,
T.
et al (
2020
)
STING and IRF3 function in stromal fibroblasts enables sensing of genomic stress in cancer cells thereby undermining oncolytic viral therapy
.
Nat. Cell Biol.
22
,
758
766
143
Mekers
,
V.E.
,
Kho
,
V.M.
,
Ansems
,
M.
and
Adema
,
G.J.
(
2022
)
cGAS/cGAMP/STING signal propagation in the tumor microenvironment: key role for myeloid cells in antitumor immunity
.
Radiother. Oncol.
174
,
158
167
144
Li
,
L.
,
Yin
,
Q.
,
Kuss
,
P.
,
Maliga
,
Z.
,
Millán
,
J.L.
,
Wu
,
H.
et al (
2014
)
Hydrolysis of 2′3′-cGAMP by ENPP1 and design of nonhydrolyzable analogs
.
Nat. Chem. Biol.
10
,
1043
1048
145
Carozza
,
J.A.
,
Böhnert
,
V.
,
Nguyen
,
K.C.
,
Skariah
,
G.
,
Shaw
,
K.E.
,
Brown
,
J.A.
et al (
2020
)
Extracellular cGAMP is a cancer-cell-produced immunotransmitter involved in radiation-induced anticancer immunity
.
Nat. Cancer
1
,
184
196
146
Vijayan
,
D.
,
Young
,
A.
,
Teng
,
M.W.L.
and
Smyth
,
M.J.
(
2017
)
Targeting immunosuppressive adenosine in cancer
.
Nat. Rev. Cancer
17
,
709
724
147
Li
,
J.
,
Duran
,
M.A.
,
Dhanota
,
N.
,
Chatila
,
W.K.
,
Bettigole
,
S.E.
,
Kwon
,
J.
et al (
2021
)
Metastasis and immune evasion from extracellular cGAMP hydrolysis
.
Cancer Discov.
11
,
1212
1227
148
Benmerzoug
,
S.
,
Rose
,
S.
,
Bounab
,
B.
,
Gosset
,
D.
,
Duneau
,
L.
,
Chenuet
,
P.
et al (
2018
)
STING-dependent sensing of self-DNA drives silica-induced lung inflammation
.
Nat. Commun.
9
,
5226
149
Sisirak
,
V.
,
Sally
,
B.
,
D'Agati
,
V.
,
Martinez-Ortiz
,
W.
,
Özçakar
,
Z.B.
,
David
,
J.
et al (
2016
)
Digestion of chromatin in apoptotic cell microparticles prevents autoimmunity
.
Cell
166
,
88
101
150
An
,
X.
,
Zhu
,
Y.
,
Zheng
,
T.
,
Wang
,
G.
,
Zhang
,
M.
,
Li
,
J.
et al (
2019
)
An analysis of the expression and association with immune cell infiltration of the cGAS/STING pathway in pan-cancer
.
Mol. Ther. Nucleic Acids
14
,
80
89
151
Liang
,
H.
,
Deng
,
L.
,
Hou
,
Y.
,
Meng
,
X.
,
Huang
,
X.
,
Rao
,
E.
et al (
2017
)
Host STING-dependent MDSC mobilization drives extrinsic radiation resistance
.
Nat. Commun.
8
,
1736
152
Shae
,
D.
,
Becker
,
K.W.
,
Christov
,
P.
,
Yun
,
D.S.
,
Lytton-Jean
,
A.K.R.
,
Sevimli
,
S.
et al (
2019
)
Endosomolytic polymersomes increase the activity of cyclic dinucleotide STING agonists to enhance cancer immunotherapy
.
Nat. Nanotechnol.
14
,
269
278
153
Kho
,
V.M.
,
Mekers
,
V.E.
,
Span
,
P.N.
,
Bussink
,
J.
and
Adema
,
G.J.
(
2021
)
Radiotherapy and cGAS/STING signaling: impact on MDSCs in the tumor microenvironment
.
Cell. Immunol.
362
,
104298
154
Ni
,
H.
,
Zhang
,
H.
,
Li
,
L.
,
Huang
,
H.
,
Guo
,
H.
,
Zhang
,
L.
et al (
2022
)
T cell-intrinsic STING signaling promotes regulatory T cell induction and immunosuppression by upregulating FOXP3 transcription in cervical cancer
.
J. Immunother. Cancer
10
,
e005151
155
Benmerzoug
,
S.
,
Bounab
,
B.
,
Rose
,
S.
,
Gosset
,
D.
,
Biet
,
F.
,
Cochard
,
T.
et al (
2019
)
Sterile lung inflammation induced by silica exacerbates mycobacterium tuberculosis infection via STING-dependent type 2 immunity
.
Cell Rep.
27
,
2649
2664.e5
156
Ma
,
R.
,
Ji
,
T.
,
Chen
,
D.
,
Dong
,
W.
,
Zhang
,
H.
,
Yin
,
X.
et al (
2016
)
Tumor cell-derived microparticles polarize M2 tumor-associated macrophages for tumor progression
.
OncoImmunology
5
,
e1118599
157
Larkin
,
B.
,
Ilyukha
,
V.
,
Sorokin
,
M.
,
Buzdin
,
A.
,
Vannier
,
E.
and
Poltorak
,
A.
(
2017
)
Cutting edge: activation of STING in T cells induces type I IFN responses and cell death
.
J. Immunol.
199
,
397
402
158
Cerboni
,
S.
,
Jeremiah
,
N.
,
Gentili
,
M.
,
Gehrmann
,
U.
,
Conrad
,
C.
,
Stolzenberg
,
M.-C.
et al (
2017
)
Intrinsic antiproliferative activity of the innate sensor STING in T lymphocytes
.
J. Exp. Med.
214
,
1769
1785
159
Imanishi
,
T.
,
Unno
,
M.
,
Kobayashi
,
W.
,
Yoneda
,
N.
,
Matsuda
,
S.
,
Ikeda
,
K.
et al (
2019
)
Reciprocal regulation of STING and TCR signaling by mTORC1 for T-cell activation and function
.
Life Sci. Alliance
2
,
e201800282
160
Wu
,
J.
,
Dobbs
,
N.
,
Yang
,
K.
and
Yan
,
N.
(
2020
)
Interferon-independent activities of mammalian STING mediate antiviral response and tumor immune evasion
.
Immunity
53
,
115
126.e5
161
Wu
,
J.
,
Chen
,
Y.-J.
,
Dobbs
,
N.
,
Sakai
,
T.
,
Liou
,
J.
,
Miner
,
J.J.
et al (
2019
)
STING-mediated disruption of calcium homeostasis chronically activates ER stress and primes T cell death
.
J. Exp. Med.
216
,
867
883
162
Du
,
S.-S.
,
Chen
,
G.-W.
,
Yang
,
P.
,
Chen
,
Y.-X.
,
Hu
,
Y.
,
Zhao
,
Q.-Q.
et al (
2022
)
Radiation therapy promotes hepatocellular carcinoma immune cloaking via PD-L1 upregulation induced by cGAS-STING activation
.
Int. J. Radiat. Oncol. Biol. Phys.
112
,
1243
1255
163
Cheng
,
A.N.
,
Cheng
,
L.-C.
,
Kuo
,
C.-L.
,
Lo
,
Y.K.
,
Chou
,
H.-Y.
,
Chen
,
C.-H.
et al (
2020
)
Mitochondrial Lon-induced mtDNA leakage contributes to PD-L1–mediated immunoescape via STING-IFN signaling and extracellular vesicles
.
J. Immunother. Cancer
8
,
e001372
164
Ma
,
H.
,
Kang
,
Z.
,
Foo
,
T.K.
,
Shen
,
Z.
and
Xia
,
B.
(
2022
)
Disrupted BRCA1-PALB2 interaction induces tumor immunosuppression and T-lymphocyte infiltration in HCC through cGAS-STING pathway
.
Hepatology
77
,
33
47
165
Garcia-Diaz
,
A.
,
Shin
,
D.S.
,
Moreno
,
B.H.
,
Saco
,
J.
,
Escuin-Ordinas
,
H.
,
Rodriguez
,
G.A.
et al (
2017
)
Interferon receptor signaling pathways regulating PD-L1 and PD-L2 expression
.
Cell Rep.
19
,
1189
1201
166
Parkes
,
E.E.
,
Savage
,
K.I.
,
Lioe
,
T.
,
Boyd
,
C.
,
Halliday
,
S.
,
Walker
,
S.M.
et al (
2022
)
Activation of a cGAS-STING-mediated immune response predicts response to neoadjuvant chemotherapy in early breast cancer
.
Br. J. Cancer
126
,
247
258
167
Parkes
,
E.E.
,
Humphries
,
M.P.
,
Gilmore
,
E.
,
Sidi
,
F.A.
,
Bingham
,
V.
,
Phyu
,
S.M.
et al (
2021
)
The clinical and molecular significance associated with STING signaling in breast cancer
.
NPJ Breast Cancer
7
,
81
168
Davoli
,
T.
,
Uno
,
H.
,
Wooten
,
E.C.
and
Elledge
,
S.J.
(
2017
)
Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy
.
Science
355
,
eaaf8399
169
Taylor
,
A.M.
,
Shih
,
J.
,
Ha
,
G.
,
Gao
,
G.F.
,
Zhang
,
X.
,
Berger
,
A.C.
et al (
2018
)
Genomic and functional approaches to understanding cancer aneuploidy
.
Cancer Cell
33
,
676
689.e3
170
Tripathi
,
R.
,
Modur
,
V.
,
Senovilla
,
L.
,
Kroemer
,
G.
and
Komurov
,
K.
(
2019
)
Suppression of tumor antigen presentation during aneuploid tumor evolution contributes to immune evasion
.
OncoImmunology
8
,
1657374
171
Kabashima
,
A.
,
Matsuo
,
Y.
,
Ito
,
S.
,
Akiyama
,
Y.
,
Ishii
,
T.
,
Shimada
,
S.
et al (
2022
)
cGAS-STING signaling encourages immune cell overcoming of fibroblast barricades in pancreatic cancer
.
Sci. Rep.
12
,
10466
172
Demaria
,
O.
,
De Gassart
,
A.
,
Coso
,
S.
,
Gestermann
,
N.
,
Di Domizio
,
J.
,
Flatz
,
L.
et al (
2015
)
STING activation of tumor endothelial cells initiates spontaneous and therapeutic antitumor immunity
.
Proc. Natl Acad. Sci. U.S.A.
112
,
15408
15413
173
Yang
,
H.
,
Lee
,
W.S.
,
Kong
,
S.J.
,
Kim
,
C.G.
,
Kim
,
J.H.
,
Chang
,
S.K.
et al (
2019
)
STING activation reprograms tumor vasculatures and synergizes with VEGFR2 blockade
.
J. Clin. Invest.
129
,
4350
4364
174
Campisi
,
M.
,
Sundararaman
,
S.K.
,
Shelton
,
S.E.
,
Knelson
,
E.H.
,
Mahadevan
,
N.R.
,
Yoshida
,
R.
et al (
2020
)
Tumor-derived cGAMP regulates activation of the vasculature
.
Front. Immunol.
11
,
2090
175
Laughney
,
A.M.
,
Elizalde
,
S.
,
Genovese
,
G.
and
Bakhoum
,
S.F.
(
2015
)
Dynamics of tumor heterogeneity derived from clonal karyotypic evolution
.
Cell Rep.
12
,
809
820
176
Andor
,
N.
,
Graham
,
T.A.
,
Jansen
,
M.
,
Xia
,
L.C.
,
Aktipis
,
C.A.
,
Petritsch
,
C.
et al (
2016
)
Pan-cancer analysis of the extent and consequences of intratumor heterogeneity
.
Nat. Med.
22
,
105
113
177
Gusev
,
Y.
,
Kagansky
,
V.
and
Dooley
,
W.C.
(
2001
)
Long-term dynamics of chromosomal instability in cancer: a transition probability model
.
Math. Comput. Model.
33
,
1253
1273
178
Silk
,
A.D.
,
Zasadil
,
L.M.
,
Holland
,
A.J.
,
Vitre
,
B.
,
Cleveland
,
D.W.
and
Weaver
,
B.A.
(
2013
)
Chromosome missegregation rate predicts whether aneuploidy will promote or suppress tumors
.
Proc. Natl Acad. Sci. U.S.A.
110
,
E4134
E4141
179
Zasadil
,
L.M.
,
Britigan
,
E.M.C.
,
Ryan
,
S.D.
,
Kaur
,
C.
,
Guckenberger
,
D.J.
,
Beebe
,
D.J.
et al (
2016
)
High rates of chromosome missegregation suppress tumor progression but do not inhibit tumor initiation
.
Mol. Biol. Cell
27
,
1981
1989
180
Jamal-Hanjani
,
M.
,
A'Hern
,
R.
,
Birkbak
,
N.J.
,
Gorman
,
P.
,
Grönroos
,
E.
,
Ngang
,
S.
et al (
2015
)
Extreme chromosomal instability forecasts improved outcome in ER-negative breast cancer: a prospective validation cohort study from the TACT trial
.
Ann. Oncol.
26
,
1340
1346
181
van Jaarsveld
,
R.H.
and
Kops
,
G.J.P.L.
(
2016
)
Difference makers: chromosomal instability versus aneuploidy in cancer
.
Trends Cancer
2
,
561
571
182
Bakhoum
,
S.F.
,
Kabeche
,
L.
,
Wood
,
M.D.
,
Laucius
,
C.D.
,
Qu
,
D.
,
Laughney
,
A.M.
et al (
2015
)
Numerical chromosomal instability mediates susceptibility to radiation treatment
.
Nat. Commun.
6
,
5990
183
Swanton
,
C.
,
Nicke
,
B.
,
Schuett
,
M.
,
Eklund
,
A.C.
,
Ng
,
C.
,
Li
,
Q.
et al (
2009
)
Chromosomal instability determines taxane response
.
Proc. Natl Acad. Sci. U.S.A.
106
,
8671
8676
184
Cohen-Sharir
,
Y.
,
McFarland
,
J.M.
,
Abdusamad
,
M.
,
Marquis
,
C.
,
Bernhard
,
S.V.
,
Kazachkova
,
M.
et al (
2021
)
Aneuploidy renders cancer cells vulnerable to mitotic checkpoint inhibition
.
Nature
590
,
486
491
185
Quinton
,
R.J.
,
DiDomizio
,
A.
,
Vittoria
,
M.A.
,
Kotýnková
,
K.
,
Ticas
,
C.J.
,
Patel
,
S.
et al (
2021
)
Whole-genome doubling confers unique genetic vulnerabilities on tumour cells
.
Nature
590
,
492
497
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY). Open access for this article was enabled by the participation of University of Oxford in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with JISC.