Stimulator of interferon genes (STING) is an innate immune sensor of cytoplasmic dsDNA originating from microorganisms and host cells. STING plays an important role in the regulation of murine graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT) and may be similarly activated during other transplantation modalities. In this review, we discuss STING in allo-HSCT and its prospective involvement in autologous HSCT (auto-HSCT) and solid organ transplantation (SOT), highlighting its unique role in nonhematopoietic, hematopoietic, and malignant cell types.

Stimulator of interferon genes (STING) is an endoplasmic reticulum protein that acts as an indirect sensor of cytoplasmic double-stranded DNA (dsDNA).1  The sources of DNA that induce cyclic dinucleotides (CDNs) include the genomes of invading pathogens, including herpes simplex virus 1 (HSV-1) and cytomegalovirus, whereas certain bacteria can secrete CDNs (eg, Listeria monocytogenes) after infection of the host.2-4  STING signaling has now been shown to be essential for protecting the cell against a variety of pathogens and even against the development of cancer by promoting antitumor immune responses.5,6  The canonical ligand for STING is the CDN 2′,3′-cGAMP which is produced as a result of DNA sensing by the protein cyclic GMP-AMP synthase (cGAS). STING can also be activated by other CDNs, including cyclic di-AMP (c-di-AMP) and cyclic di-GMP (c-di-GMP).7,8  After binding to CDNs, STING recruits TBK1 and traffics to perinuclear regions.9  Downstream STING signaling results in the activation of the transcription factors interferon regulatory factor 3 (IRF3) and NF-κB, which induce type I interferons (T1IFNs) and additional inflammatory molecules (Figure 1A).10  Notably, STING alleles in the human population have been identified that encode proteins that are largely unresponsive to CDNs, raising the notion that there may be clinical implications for individuals with these genotypes.11  Patients are administered liquid (bone marrow transplants) and solid organ transplants (SOTs) to replace damaged tissue compartments. Continuing efforts to develop clinically useful STING agonists and antagonists may prove effective as therapies to reduce or prevent allograft rejection (host-versus-graft) in SOT and graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Notably, complications in transplant recipients include an array of infections and hematopoietic malignancies, and therefore regulating STING may affect immune defense against microbial pathogens and cancer in such patients. In this review, we focus on current literature regarding STING and its role in allo-HSCT, which is currently the predominant transplantation arena where it has begun to be studied. We also speculate on the involvement of STING in autologous HSCT (auto-HSCT) and SOT and highlight recent work using new STING compounds, which are bringing STING-targeted agents closer to implementation in the clinic.

Figure 1.

The STING pathway in NH and HCs: prospective contribution to transplant outcome. (A) Left: As a result of transplantation-related damage, dsDNA from host cells (eg, dendritic cells, epithelial cells) or viruses induce cyclic dinucleotide (CDN) production by cGAS which bind to and activate STING. Bacterial CDNs can also activate STING directly. Downstream of STING signaling, activation of IRF3 and NF-κB induce the production of cytokines, which regulate immune activation and reparative processes such as epithelial regeneration. Right: STING activation in the NH or hematopoietic compartments during SOT could increase T-cell responses against donor grafts. STING activation in the NH compartment during allo-HSCT increases aGVHD in MHC-matched transplant recipients; however, expression of STING in this compartment decreases aGVHD in MHC-mismatched transplant recipients. The potential role of STING in auto-HSCT is unclear. Promotion of epithelial repair in the GI tract and/or increased anti-pathogen T-cell responses could diminish recipient transplant-related mortality (TRM). Alternatively, increased cytokine production as a result of conditioning could elevate recipient TRM. (B) STING-targeted agents (see Table 1) can be combined with tumor vaccination and checkpoint blockade to promote tumor-specific responses without exacerbating GVHD. Application of this strategy could be combined with prophylactic GVHD regimens involving posttransplant cyclophosphamide to reduce potential exacerbation of donor antihost alloreactivity in addition to direct cytotoxic effects on the tumor. EBV, Epstein-Barr virus.

Figure 1.

The STING pathway in NH and HCs: prospective contribution to transplant outcome. (A) Left: As a result of transplantation-related damage, dsDNA from host cells (eg, dendritic cells, epithelial cells) or viruses induce cyclic dinucleotide (CDN) production by cGAS which bind to and activate STING. Bacterial CDNs can also activate STING directly. Downstream of STING signaling, activation of IRF3 and NF-κB induce the production of cytokines, which regulate immune activation and reparative processes such as epithelial regeneration. Right: STING activation in the NH or hematopoietic compartments during SOT could increase T-cell responses against donor grafts. STING activation in the NH compartment during allo-HSCT increases aGVHD in MHC-matched transplant recipients; however, expression of STING in this compartment decreases aGVHD in MHC-mismatched transplant recipients. The potential role of STING in auto-HSCT is unclear. Promotion of epithelial repair in the GI tract and/or increased anti-pathogen T-cell responses could diminish recipient transplant-related mortality (TRM). Alternatively, increased cytokine production as a result of conditioning could elevate recipient TRM. (B) STING-targeted agents (see Table 1) can be combined with tumor vaccination and checkpoint blockade to promote tumor-specific responses without exacerbating GVHD. Application of this strategy could be combined with prophylactic GVHD regimens involving posttransplant cyclophosphamide to reduce potential exacerbation of donor antihost alloreactivity in addition to direct cytotoxic effects on the tumor. EBV, Epstein-Barr virus.

Close modal

Despite the widespread use of allo-HSCT for more than 5 decades, GVHD that is driven by major and minor histocompatibility antigen disparities between donor and recipient remains a major cause of nonrelapse morbidity and mortality.12-14  GVHD is potentiated when innate immune sensors, mainly in hematopoietic and nonhematopoietic antigen-presenting cells (APCs), are activated in response to pathogen-associated molecular patterns and damage-associated molecular patterns released by pretransplant chemotherapy or irradiation that result in upregulating certain cytokines.15  Recent work by our laboratory and others has established that 1 such innate sensor, STING, is a potent regulator of GVHD.16,17  The initial studies that demonstrated the ability of STING to regulate GVHD used murine models of major histocompatibility complex (MHC)–mismatched HSCT and recipients (B6-Goldenticket, B6-STINGgt/gt) expressing a missense mutation in the STING gene.18  Transplants into B6-STINGgt/gt recipients resulted in higher lethality compared with B6 wild-type recipients, which has been attributed to the reduced ability of STING-deficient recipients to regenerate their intestinal epithelium after pretransplant conditioning.16 

Recently, by using an independently derived STING-deficient mouse (B6-STING−/−), the initial report that murine STING deficiency results in worse GVHD after MHC-mismatched allo-HSCT was independently corroborated.17  These studies also examined MHC-matched HSCT and discovered that STING deficiency ameliorated GVHD in these matched unrelated donor models.17  Donor CD4+ or CD8+ T cells often predominate in preclinical graft-versus-host responses (ie, CD4+ in MHC-mismatched vs CD8+ in MHC-matched).19  Subset studies revealed that STING deficiency protected against GVHD when only donor CD8+ T cells were transplanted into either MHC-mismatched or -matched recipients, thereby reconciling the seemingly disparate results in these models. Interestingly, T1IFN was also shown to differentially regulate CD4+ vs CD8+ T-cell–mediated GVHD.20  Moreover, T1IFN enhanced MHC class I expression on recipient hematopoietic APCs, which are critical for the induction of GVHD.17  In contrast, hematopoietic APCs responding to T1IFNs have diminished allogenicity, suggesting a protective role for STING in MHC-mismatched GVHD.21 

Nonhematopoietic (NH) cells are important for the initiation of gut GVHD.22  Hematopoietic APCs seem to play a role after STING signaling following allo-HSCT, and STING expression in NH cells protected against CD4+ T-cell–mediated GVHD but enhanced CD8+ T-cell–mediated GVHD regardless of the allo-HSCT model.17  The importance of the NH compartment was also demonstrated in knockout mice that lacked mitochondrial antiviral-signaling protein, a critical molecule involved in cytoplasmic RNA sensing.16  The specific NH cell types in which STING expression is important for the regulation of GVHD remains unknown, but STING activation in NH cells has been shown to be responsible for the initiation of autoimmune disease involving activated T cells.23  We speculate that in non-hematolymphoid tissue compartments, STING activation within NH-APCs leading to production of T1IFNs and inflammatory cytokines could target parenchymal populations that are distinct from those in the hematopoietic tissues. For example, IFNs could upregulate MHC class I on tissue-resident cells which would alter their susceptibility as target populations. NH-APC cytokine/chemokine (eg, MCP-1/CCL2) production downstream of T1IFNs could promote the trafficking of hematopoietic cell (HC)-APCs into lymphoid tissues thus augmenting alloreactive donor T-cell responses. NH-APCs after HSCT could engage CD8+ effector cells and activate resident memory populations. Activation of the STING pathway in any recipient cell could occur via several routes, depending on the host compartment. After conditioning involving chemotherapy and irradiation, virtually all cells could activate STING in response to dying cells via uptake of dsDNA.24,25 

Intestinal microbiota may also play a role in activating STING in recipient cells after HSCT, because pretransplant conditioning directly induces intestinal permeability, which provides exposure of bacterial dsDNA to numerous cell populations.26,27  Consistent with such a pathway, gut microbiota are capable of initiating lethal GVHD through upregulation of MHC class II on NH intestinal epithelial cells.22  Therefore, the extent to which conditioning regimens damage the gastrointestinal (GI) tract may correlate with the level of STING activation in this compartment. In addition, recent studies have reported emerging pathways and new mechanisms leading to STING activation involving noncanonical signaling complexes and micronuclei in damaged cells.28-30  Further studies are required to identify the precise mechanisms by which recipient STING is activated before and during HSCT to better understand how STING signaling differentially regulates GVHD. It should be noted that preclinical mouse studies are typically performed under specific pathogen-free conditions and thus do not mimic the opportunistic viral (eg, reactivation of cytomegalovirus), bacterial, and fungal infections that frequently occur in allo-HSCT patients and can cause mortality (Figure 1A).31-33  Diminishing or augmenting STING signaling could alter the course of such infections, which can influence GVHD. For example, decreasing STING signaling could diminish responses against such microbial pathogens. Alternatively, augmenting STING signaling could enhance anti-pathogen immunity thereby preventing pathogen promotion of GVHD.34,35  Overall, both the timing of STING activation and the transplant parameters will have an impact on how this pathway regulates GVHD to ultimately enable STING agonists and/or antagonists to benefit patients receiving allo-HSCTs.

Allogeneic HSCT is most often used to treat hematologic malignancies, primarily leukemias, to exploit the accompanying antitumor (graft-versus-leukemia [GVL]) response. Patients who develop acute GVHD (aGVHD) or chronic GVHD (cGVHD) have a lower frequency of leukemia relapse because of immune activation against alloantigens expressed on both normal and malignant cells.36  STING agonists can promote GVHD,17  and therefore this pathway could augment beneficial GVL responses by facilitating the activation of donor T cells that target alloantigens and tumor antigens (Figure 1B). The outcome of STING activation can reportedly be either immunogenic or tolerogenic. Thus, it is possible that the former would result in increased graft-versus-host and GVL responses, whereas the latter may diminish these responses.37  Alternatively, STING antagonists could diminish GVL by decreasing donor alloantigen-specific or tumor-specific T cells resulting in increased risk of relapse. Several studies have demonstrated that STING-activating agents can improve antitumor T-cell responses in both solid and liquid tumor models.38-40  In solid tumors, in which the majority of STING-related cancer research has been performed, defective STING signaling in tumors has been shown to correlate with increased tumorigenesis in colorectal carcinoma and ovarian cancer.41,42  Interestingly, tumor-derived 2′,3′-cGAMP can trigger STING-mediated T1IFN in neighboring cells as long as cGAS is functional through mechanisms involving the transfer of CDNs from one cell to another.43 

STING signaling in tumor cells also correlates with their antigenicity and immunogenicity.44,45  Increased tumor cell immunogenicity can result from tumor-derived DNA, which activates STING signaling in both tumor and dendritic cells (DCs) and leads to the production of T1IFN and increased antitumor immunity.46  STING signaling in solid tumors can also be initiated by irradiation-induced DNA damage, and liquid tumors may be similarly activated in response to pre-HSCT chemotherapy.47  Notably, the dose of irradiation can regulate this response, because higher doses attenuate STING activation as a result of the upregulation of 3′ repair exonuclease 1 (TREX1), which directly competes with cGAS by degrading cytoplasmic dsDNA.48  It is therefore likely that the precise conditioning regimen used for HSCT is in part responsible for the level of STING signaling in response to chemotherapy and irradiation. STING signaling reportedly slows tumor proliferation via control of NF-κB and p53-driven activation of p21.49  In contrast, STING activation has also been shown to be immunosuppressive via the recruitment of myeloid-derived suppressor cells to the tumor site via CCR2.50  Thus, STING signaling in tumors is complex, and in transplant recipients, downstream effects of STING activation are likely to be highly dependent upon both the type of tumor and the conditioning regimen.

Unfortunately, primary disease is still the most common cause of mortality after allo-HSCT.51  Activation of STING in donor T cells can induce strong antitumor responses despite the antiproliferative effects of T1IFN.52,53  Moreover, STING-induced T1IFN is effective at promoting natural killer cell antitumor activity.54  Strategies to combine HSCT with posttransplant cellular therapy are being developed that may improve transplant outcomes.55  Notably, STING in DCs is reportedly activated by tumor mitochondrial DNA and can be enhanced by tumor CD47 blockade.56  On the basis of such types of findings, it may be noteworthy that pulsed DC vaccines have been shown to be effective at inducing antigen-specific immune responses.57  Nanovaccines targeting STING have also been developed that potentiate antitumor T-cell activation.58,59  DCs pulsed with such vaccines and added to the donor graft may be effective as a novel cellular therapy to promote effective antitumor responses against minimal residual disease after HSCT. Some DC subsets have been identified as strong responders to STING-targeted agents and therefore could be isolated and/or expanded for adoptive cellular therapy accompanying HSCT.60  T1IFN has potent adjuvant activity in combinatorial approaches using DNA vaccines that target immature DCs.61  Interestingly, adjuvant activity of STING-targeted agents may be dependent on tumor necrosis factor α,62  an important cytokine produced after HSCT conditioning and during GVHD.63  Because STING has been shown to be a potent target for enhancing antitumor immunity, there is provocative rationale for developing cellular therapy approaches that use STING-targeted agents as adjuvants to eradicate residual tumor after HSCT.

There is much diversity in the types of conditioning regimens used throughout various centers that perform allo-HSCT.64  We anticipate that, as the regimens involving radiation and/or chemotherapy (busulfan, fludarabine, and cyclophosphamide) are lessened, correspondingly less DNA damage will result which would decrease potential self-DNA–induced STING activation. In addition, reduced intensity regimens will result in diminished GI damage by decreasing leakage of bacterial products that activate STING.65  In contrast, high-dose myeloablative conditioning is the predominant choice for auto-HSCT conditioning.66,67  Because of cell death and accompanying GI damage resulting from myeloablative conditioning, STING signaling is likely pertinent in auto-HSCT and, lacking alloreactive T cells, would result from effects on host HCs and NH cells (Figure 1), including tumor cells. However, STING regulation at the time of auto-HSCT may suppress inflammation and epithelial repair, both potentially influencing nonrelapse mortality. STING agonists might also be used in combination with tumor vaccination to promote GVL in the auto-HSCT setting.68 

Although it has not been explored, activation of the STING pathway would not be surprising after SOT. Cell death or DNA damage after SOT is likely less than that after HSCT but could provide a dsDNA source to activate STING as could bacterial CDNs after prolonged immunosuppression. Ischemia or reperfusion injury occurring during graft harvesting, cold storage, or surgery are underlying causes of inflammation and graft dysfunction.69-72  Subsequent activation of STING in HCs as well as NH-APCs in the tissue graft or draining nodes could drive host T-cell antidonor alloantigen rejection responses. Chronic rejection remains the most problematic challenge confronting transplantation physicians. Infection resulting from long-term immunosuppression provides potential for bacterial STING stimulation. The involvement of STING in the fate of solid tissue grafts could differ, depending on the location and type of transplanted tissue. For example, orthotopic ocular, skin, and GI allografts provide an environment that may readily expose the transplanted tissue cells to bacterial signals activating the STING pathway. In addition, use of immune suppressive treatment to prolong graft survival could, over time, lead to activation of host T-effector or memory cell populations that take up residence in the graft by persisting donor NH-APCs. It will be interesting to determine whether STING contributes via HCs and/or NH-APCs to SOT rejection and, if so, whether its targeting may be useful for prolonging graft survival.

aGVHD and cGVHD represent 2 distinct clinical diseases and have been demarcated by onset <100 and >100 days from allo-HSCT, respectively. However, it is well appreciated that aGVHD may not predate cGVHD and late-onset (>100 days) GVHD can be indistinguishable from aGVHD.73,74  Nonetheless, transition from aGVHD to cGVHD occurs and remains unexplained. Patients with defective immune systems who become exposed to infectious agents could experience sporadic or sustained STING activation weeks or months after HSCT that provokes alloantigen or autoantigen responses contributing to cGVHD.

Preclinical studies in mice have taken advantage of gene deletion to generate STING-deficient animals that have enabled significant discoveries to improve understanding of the signaling pathway and its importance in host defenses. Interestingly, human STING alleles are heterogeneous, and R71H-G230A-R293Q (HAQ), the second most common STING allele, is associated with decreased STING function.11,75,76  STING can influence vaccine effectiveness because lower antibody responses were reported in STING-deficient mice.77  Our recent pneumococcal vaccine investigation identified diminished antibody responses in individuals with a single HAQ copy.75  With regard to targeting the STING pathway, although the initial compound targeting the ligand-binding region, 5,6-dimethylxanthenone-4-acetic acid (DMXAA), is ineffective in humans,78  chitosan, α-mangostin, and amidobenzimidazole-based compounds have been identified that agonize the STING pathway in humans and mice (Table 1).79-81  Studies investigating specific CDN family members have identified c-di-GMP and the novel synthetic CDN ML-RR-S2 c-di-AMP as potent STING activators.82,83  Nitrofuran-based small molecules can also inhibit human and mouse STING by targeting its transmembrane region (Table 1).84  Regarding clinical translation of mouse studies, STING’s broad expression and intracellular location will likely require adoptive cell therapy and/or targeting agonists and antagonists across the cell membrane to specific cell populations. For example, activating the STING pathway in APCs ex vivo before adoptive transfer to drive immune responses and targeting cells using nanoparticles directed to specific cell surface receptors that contain STING activators have been reported.85,86  These types of strategies would also diminish global and off-target effects.

Table 1.

Compounds (published or in trials) directed to agonizing or antagonizing the STING pathway for application to preclinical and clinical studies

graphic
 
graphic
 

CCCP, carbonyl cyanide 3-chlorophenylhydrazone; CDA, c-di-AMP; CMA, 10-carboxymethyl-9-acridanone; CTD, C-terminal domain; DRP1, dynamin-related protein 1.

STING agonists have also been successful as adjuvants for tumor vaccines.83,87  Regarding enhancement of GVL, delivery of STING agonists via nanoshells before chemotherapy can promote antitumor activity by placing tumor antigen and STING-based adjuvant in the same physical location.88  Liposomal nanoparticles loaded with CDNs are more effective at activating STING within tumor cells compared with naked CDNs, likely because of the impermeability of CDNs to the cell membrane.89  Combinatorial approaches that activate STING may be synergistic or additive with inhibitors of the noncanonical NF-κB, absent in melanoma 2 (AIM2) inflammasome, or caspase 1–mediated cell death pathways.90,91  STING activation via microbiota or agonists could also improve antitumor responses when combined with CD47, glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR), or programmed cell death protein 1 (PD-1) checkpoint blockade.92,93  Murine studies have shown promising results for STING-targeting cancer therapeutics, and multiple trials implementing STING targeting for cancer immunotherapy of solid tumors are underway (E7766: NCT04109092, ADU-S100: NCT02675439, MK1454: NCT03010176) (Table 1). These agonists and others have not yet been tested for treatment of blood cancers and the promotion of the GVL effect. The use of posttransplant cyclophosphamide has been shown to diminish antirecipient alloreactivity.94,95  Administration of a tumor vaccine combined with STING agonists in patients at some time point after posttransplant cyclophosphamide could promote an antitumor-specific response without exacerbating GVHD alloreactivity.

In conclusion, isolating the absence of STING both singularly and in combination with donor cells, host HCs, host NH cells, or tumor cells will provide a way to carefully interrogate its significance in different tissues after experimental transplantation. Studies using STING pathway–specific agonists or antagonists could then be performed to corroborate the precise significance of the pathway in vivo and further define the location and sources of STING signals that influence the outcome of liquid and solid transplants.

The authors thank Zhibin Chen, Lazaros Lekakis, Sabrina Copsel, and Dietlinde Wolf for their critiques and helpful discussions regarding the manuscript.

This work was supported by grants from the National Institutes of Health (NIH), National Eye Institute (R01EY024484, R01EY030283), the Sylvester Comprehensive Cancer Center (R.B.L.), the NIH National Institute of Allergy and Infectious Diseases (R01AI110606, R21AI125999) (L.J.), and the NIH National Cancer Institute (F31CA216999, F99CA245728) (C.S.B.).

Contribution: C.S.B., L.J., and R.B.L. wrote the manuscript.

Conflict-of-interest disclosure: R.B.L. served as a consultant or advisory board member for and equity holder in Heat Biologics. The remaining authors declare no competing financial interests.

Correspondence: Robert B. Levy, Department of Microbiology and Immunology, University of Miami Miller School of Medicine, R-138, 1600 NW 10th Ave, Miami, FL 33136; e-mail: rlevy@med.miami.edu.

1.
Ishikawa
H
,
Barber
GN
.
STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling
.
Nature
.
2008
;
455
(
7213
):
674
-
678
.
2.
Ishikawa
H
,
Ma
Z
,
Barber
GN
.
STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity
.
Nature
.
2009
;
461
(
7265
):
788
-
792
.
3.
Sauer
JD
,
Pereyre
S
,
Archer
KA
, et al
.
Listeria monocytogenes engineered to activate the Nlrc4 inflammasome are severely attenuated and are poor inducers of protective immunity
.
Proc Natl Acad Sci U S A
.
2011
;
108
(
30
):
12419
-
12424
.
4.
Ma
Z
,
Damania
B
.
The cGAS-STING defense pathway and its counteraction by viruses
.
Cell Host Microbe
.
2016
;
19
(
2
):
150
-
158
.
5.
Woo
SR
,
Corrales
L
,
Gajewski
TF
.
The STING pathway and the T cell-inflamed tumor microenvironment
.
Trends Immunol
.
2015
;
36
(
4
):
250
-
256
.
6.
Barber
GN
.
STING-dependent cytosolic DNA sensing pathways
.
Trends Immunol
.
2014
;
35
(
2
):
88
-
93
.
7.
Burdette
DL
,
Monroe
KM
,
Sotelo-Troha
K
, et al
.
STING is a direct innate immune sensor of cyclic di-GMP
.
Nature
.
2011
;
478
(
7370
):
515
-
518
.
8.
Diner
EJ
,
Burdette
DL
,
Wilson
SC
, et al
.
The innate immune DNA sensor cGAS produces a noncanonical cyclic dinucleotide that activates human STING
.
Cell Rep
.
2013
;
3
(
5
):
1355
-
1361
.
9.
Barber
GN
.
STING: infection, inflammation and cancer
.
Nat Rev Immunol
.
2015
;
15
(
12
):
760
-
770
.
10.
Liu
S
,
Cai
X
,
Wu
J
, et al
.
Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation
.
Science
.
2015
;
347
(
6227
):
aaa2630
.
11.
Patel
S
,
Blaauboer
SM
,
Tucker
HR
, et al
.
The common R71H-G230A-R293Q human TMEM173 is a null allele
.
J Immunol
.
2017
;
198
(
2
):
776
-
787
.
12.
Zeiser
R
,
Blazar
BR
.
Acute graft-versus-host disease - biologic process, prevention, and therapy
.
N Engl J Med
.
2017
;
377
(
22
):
2167
-
2179
.
13.
Zeiser
R
,
Blazar
BR
.
Pathophysiology of chronic graft-versus-host disease and therapeutic targets
.
N Engl J Med
.
2017
;
377
(
26
):
2565
-
2579
.
14.
Dickinson
AM
,
Norden
J
,
Li
S
, et al
.
Graft-versus-leukemia effect following hematopoietic stem cell transplantation for leukemia
.
Front Immunol
.
2017
;
8
:
496
.
15.
Heidegger
S
,
van den Brink
MR
,
Haas
T
,
Poeck
H
.
The role of pattern-recognition receptors in graft-versus-host disease and graft-versus-leukemia after allogeneic stem cell transplantation
.
Front Immunol
.
2014
;
5
:
337
.
16.
Fischer
JC
,
Bscheider
M
,
Eisenkolb
G
, et al
.
RIG-I/MAVS and STING signaling promote gut integrity during irradiation- and immune-mediated tissue injury
.
Sci Transl Med
.
2017
;
9
(
386
):
eaag2513
.
17.
Bader
CS
,
Barreras
H
,
Lightbourn
CO
, et al
.
STING differentially regulates experimental GVHD mediated by CD8 versus CD4 T cell subsets
.
Sci Transl Med
.
2020
;
12
(
552
):
eaay5006
.
18.
Sauer
JD
,
Sotelo-Troha
K
,
von Moltke
J
, et al
.
The N-ethyl-N-nitrosourea-induced Goldenticket mouse mutant reveals an essential function of Sting in the in vivo interferon response to Listeria monocytogenes and cyclic dinucleotides
.
Infect Immun
.
2011
;
79
(
2
):
688
-
694
.
19.
Reddy
P
,
Negrin
R
,
Hill
GR
.
Mouse models of bone marrow transplantation
.
Biol Blood Marrow Transplant
.
2008
;
14
(
1 suppl 1
):
129
-
135
.
20.
Robb
RJ
,
Kreijveld
E
,
Kuns
RD
, et al
.
Type I-IFNs control GVHD and GVL responses after transplantation
.
Blood
.
2011
;
118
(
12
):
3399
-
3409
.
21.
Fischer
JC
,
Bscheider
M
,
Göttert
S
, et al
.
Type I interferon signaling before hematopoietic stem cell transplantation lowers donor T cell activation via reduced allogenicity of recipient cells
.
Sci Rep
.
2019
;
9
(
1
):
14955
.
22.
Koyama
M
,
Mukhopadhyay
P
,
Schuster
IS
, et al
.
MHC class II antigen presentation by the intestinal epithelium initiates graft-versus-host disease and is influenced by the microbiota
.
Immunity
.
2019
;
51
(
5
):
885
-
898.e7
.
23.
Gall
A
,
Treuting
P
,
Elkon
KB
, et al
.
Autoimmunity initiates in nonhematopoietic cells and progresses via lymphocytes in an interferon-dependent autoimmune disease
.
Immunity
.
2012
;
36
(
1
):
120
-
131
.
24.
Ahn
J
,
Xia
T
,
Rabasa Capote
A
,
Betancourt
D
,
Barber
GN
.
Extrinsic phagocyte-dependent STING signaling dictates the immunogenicity of dying cells
.
Cancer Cell
.
2018
;
33
(
5
):
862
-
873.e5
.
25.
Klarquist
J
,
Hennies
CM
,
Lehn
MA
,
Reboulet
RA
,
Feau
S
,
Janssen
EM
.
STING-mediated DNA sensing promotes antitumor and autoimmune responses to dying cells
.
J Immunol
.
2014
;
193
(
12
):
6124
-
6134
.
26.
Gutierrez-Merino
J
,
Isla
B
,
Combes
T
,
Martinez-Estrada
F
,
Maluquer De Motes
C
.
Beneficial bacteria activate type-I interferon production via the intracellular cytosolic sensors STING and MAVS
.
Gut Microbes
.
2020
;
11
(
4
):
771
-
788
.
27.
Fischer
JC
,
Wintges
A
,
Haas
T
,
Poeck
H
.
Assessment of mucosal integrity by quantifying neutrophil granulocyte influx in murine models of acute intestinal injury
.
Cell Immunol
.
2017
;
316
:
70
-
76
.
28.
Dunphy
G
,
Flannery
SM
,
Almine
JF
, et al
.
Non-canonical activation of the DNA sensing adaptor STING by ATM and IFI16 mediates NF-κB signaling after nuclear DNA damage
.
Mol Cell
.
2018
;
71
(
5
):
745
-
760.e5
.
29.
Harding
SM
,
Benci
JL
,
Irianto
J
,
Discher
DE
,
Minn
AJ
,
Greenberg
RA
.
Mitotic progression following DNA damage enables pattern recognition within micronuclei
.
Nature
.
2017
;
548
(
7668
):
466
-
470
.
30.
Mackenzie
KJ
,
Carroll
P
,
Martin
CA
, et al
.
cGAS surveillance of micronuclei links genome instability to innate immunity
.
Nature
.
2017
;
548
(
7668
):
461
-
465
.
31.
Camargo
JF
,
Wieder
ED
,
Kimble
E
, et al
.
Deep functional immunophenotyping predicts risk of cytomegalovirus reactivation after hematopoietic cell transplantation
.
Blood
.
2019
;
133
(
8
):
867
-
877
.
32.
Camargo
JF
,
Kimble
E
,
Rosa
R
, et al
.
Impact of cytomegalovirus viral load on probability of spontaneous clearance and response to preemptive therapy in allogeneic stem cell transplantation recipients
.
Biol Blood Marrow Transplant
.
2018
;
24
(
4
):
806
-
814
.
33.
Camargo
JF
,
Komanduri
KV
.
Emerging concepts in cytomegalovirus infection following hematopoietic stem cell transplantation
.
Hematol Oncol Stem Cell Ther
.
2017
;
10
(
4
):
233
-
238
.
34.
Hong
J
,
Moon
SM
,
Ahn
HK
, et al
.
Comparison of characteristics of bacterial bloodstream infection between adult patients with allogeneic and autologous hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2013
;
19
(
6
):
994
-
999
.
35.
Fuji
S
,
Kim
SW
,
Fukuda
T
, et al
.
Preengraftment serum C-reactive protein (CRP) value may predict acute graft-versus-host disease and nonrelapse mortality after allogeneic hematopoietic stem cell transplantation
.
Biol Blood Marrow Transplant
.
2008
;
14
(
5
):
510
-
517
.
36.
Scott
BL
,
Pasquini
MC
,
Logan
BR
, et al
.
Myeloablative versus reduced-intensity hematopoietic cell transplantation for acute myeloid leukemia and myelodysplastic syndromes
.
J Clin Oncol
.
2017
;
35
(
11
):
1154
-
1161
.
37.
Zhu
Y
,
An
X
,
Zhang
X
,
Qiao
Y
,
Zheng
T
,
Li
X
.
STING: a master regulator in the cancer-immunity cycle
.
Mol Cancer
.
2019
;
18
(
1
):
152
.
38.
Corrales
L
,
Glickman
LH
,
McWhirter
SM
, et al
.
Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity
.
Cell Rep
.
2015
;
11
(
7
):
1018
-
1030
.
39.
Curran
E
,
Chen
X
,
Corrales
L
, et al
.
STING pathway activation stimulates potent immunity against acute myeloid leukemia
.
Cell Rep
.
2016
;
15
(
11
):
2357
-
2366
.
40.
Tang
CH
,
Zundell
JA
,
Ranatunga
S
, et al
.
Agonist-mediated activation of STING induces apoptosis in malignant B cells
.
Cancer Res
.
2016
;
76
(
8
):
2137
-
2152
.
41.
Xia
T
,
Konno
H
,
Ahn
J
,
Barber
GN
.
Deregulation of STING signaling in colorectal carcinoma constrains DNA damage responses and correlates with tumorigenesis
.
Cell Rep
.
2016
;
14
(
2
):
282
-
297
.
42.
de Queiroz
NMGP
,
Xia
T
,
Konno
H
,
Barber
GN
.
Ovarian cancer cells commonly exhibit defective STING signaling which affects sensitivity to viral oncolysis
.
Mol Cancer Res
.
2019
;
17
(
4
):
974
-
986
.
43.
Marcus
A
,
Mao
AJ
,
Lensink-Vasan
M
,
Wang
L
,
Vance
RE
,
Raulet
DH
.
Tumor-derived cGAMP triggers a STING-mediated interferon response in non-tumor cells to activate the NK cell response
.
Immunity
.
2018
;
49
(
4
):
754
-
763.e4
.
44.
Woo
SR
,
Fuertes
MB
,
Corrales
L
, et al
.
STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors
.
Immunity
.
2014
;
41
(
5
):
830
-
842
.
45.
Falahat
R
,
Perez-Villarroel
P
,
Mailloux
AW
, et al
.
STING signaling in melanoma cells shapes antigenicity and can promote antitumor T-cell activity
.
Cancer Immunol Res
.
2019
;
7
(
11
):
1837
-
1848
.
46.
Andzinski
L
,
Spanier
J
,
Kasnitz
N
, et al
.
Growing tumors induce a local STING dependent Type I IFN response in dendritic cells
.
Int J Cancer
.
2016
;
139
(
6
):
1350
-
1357
.
47.
Deng
L
,
Liang
H
,
Xu
M
, et al
.
STING-dependent cytosolic DNA sensing promotes radiation-induced Type I interferon-dependent antitumor immunity in immunogenic tumors
.
Immunity
.
2014
;
41
(
5
):
843
-
852
.
48.
Vanpouille-Box
C
,
Alard
A
,
Aryankalayil
MJ
, et al
.
DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity
.
Nat Commun
.
2017
;
8
(
1
):
15618
.
49.
Ranoa
DRE
,
Widau
RC
,
Mallon
S
, et al
.
STING promotes homeostasis via regulation of cell proliferation and chromosomal stability
.
Cancer Res
.
2019
;
79
(
7
):
1465
-
1479
.
50.
Liang
H
,
Deng
L
,
Hou
Y
, et al
.
Host STING-dependent MDSC mobilization drives extrinsic radiation resistance
.
Nat Commun
.
2017
;
8
(
1
):
1736
.
51.
D’Souza
A
,
Fretham
C
,
Lee
SJ
, et al
.
Current use of and trends in hematopoietic cell transplantation in the United States
.
Biol Blood Marrow Transplant
.
2020
;
26
(
8
):
e177
-
e182
.
52.
Imanishi
T
,
Unno
M
,
Kobayashi
W
, et al
.
Reciprocal regulation of STING and TCR signaling by mTORC1 for T-cell activation and function
.
Life Sci Alliance
.
2019
;
2
(
1
):
e201800282
.
53.
Cerboni
S
,
Jeremiah
N
,
Gentili
M
, et al
.
Intrinsic antiproliferative activity of the innate sensor STING in T lymphocytes
.
J Exp Med
.
2017
;
214
(
6
):
1769
-
1785
.
54.
Nicolai
CJ
,
Wolf
N
,
Chang
IC
, et al
.
NK cells mediate clearance of CD8+ T cell-resistant tumors in response to STING agonists
.
Sci Immunol
.
2020
;
5
(
45
):
eaaz2738
.
55.
Singh
AK
,
McGuirk
JP
.
Allogeneic stem cell transplantation: A historical and scientific overview
.
Cancer Res
.
2016
;
76
(
22
):
6445
-
6451
.
56.
Xu
MM
,
Pu
Y
,
Han
D
, et al
.
Dendritic cells but not macrophages sense tumor mitochondrial DNA for cross-priming through signal regulatory protein α signaling
.
Immunity
.
2017
;
47
(
2
):
363
-
373.e5
.
57.
Chen
W
,
Zhang
Z
,
Shi
M
, et al
.
Activated plasmacytoid dendritic cells act synergistically with hepatitis B core antigen-pulsed monocyte-derived dendritic cells in the induction of hepatitis B virus-specific CD8 T-cell response
.
Clin Immunol
.
2008
;
129
(
2
):
295
-
303
.
58.
Miao
L
,
Li
L
,
Huang
Y
, et al
.
Delivery of mRNA vaccines with heterocyclic lipids increases anti-tumor efficacy by STING-mediated immune cell activation
.
Nat Biotechnol
.
2019
;
37
(
10
):
1174
-
1185
.
59.
Luo
M
,
Wang
H
,
Wang
Z
, et al
.
A STING-activating nanovaccine for cancer immunotherapy
.
Nat Nanotechnol
.
2017
;
12
(
7
):
648
-
654
.
60.
Mansouri
S
,
Patel
S
,
Katikaneni
DS
, et al
.
Immature lung TNFR2- conventional DC 2 subpopulation activates moDCs to promote cyclic di-GMP mucosal adjuvant responses in vivo
.
Mucosal Immunol
.
2019
;
12
(
1
):
277
-
289
.
61.
Gordy
JT
,
Luo
K
,
Kapoor
A
, et al
.
Treatment with an immature dendritic cell-targeting vaccine supplemented with IFN-α and an inhibitor of DNA methylation markedly enhances survival in a murine melanoma model
.
Cancer Immunol Immunother
.
2020
;
69
(
4
):
569
-
580
.
62.
Blaauboer
SM
,
Gabrielle
VD
,
Jin
L
.
MPYS/STING-mediated TNF-α, not type I IFN, is essential for the mucosal adjuvant activity of (3′-5′)-cyclic-di-guanosine-monophosphate in vivo
.
J Immunol
.
2014
;
192
(
1
):
492
-
502
.
63.
Levine
JE
.
Implications of TNF-α in the pathogenesis and management of GVHD
.
Int J Hematol
.
2011
;
93
(
5
):
571
-
577
.
64.
Pingali
SR
,
Champlin
RE
.
Pushing the envelope-nonmyeloablative and reduced intensity preparative regimens for allogeneic hematopoietic transplantation
.
Bone Marrow Transplant
.
2015
;
50
(
9
):
1157
-
1167
.
65.
Sengsayadeth
S
,
Savani
BN
,
Blaise
D
,
Malard
F
,
Nagler
A
,
Mohty
M
.
Reduced intensity conditioning allogeneic hematopoietic cell transplantation for adult acute myeloid leukemia in complete remission - a review from the Acute Leukemia Working Party of the EBMT
.
Haematologica
.
2015
;
100
(
7
):
859
-
869
.
66.
Gyurkocza
B
,
Sandmaier
BM
.
Conditioning regimens for hematopoietic cell transplantation: one size does not fit all
.
Blood
.
2014
;
124
(
3
):
344
-
353
.
67.
Negrin
RS
.
Preparative regimens for hematopoietic cell transplantation
. .
68.
Porrata
LF
.
Autologous graft-versus-tumor effect: Reality or fiction?
Adv Hematol
.
2016
;
2016
:
5385972
.
69.
Busuttil
RW
,
Tanaka
K
.
The utility of marginal donors in liver transplantation
.
Liver Transpl
.
2003
;
9
(
7
):
651
-
663
.
70.
Mendes-Braz
M
,
Elias-Miró
M
,
Jiménez-Castro
MB
,
Casillas-Ramírez
A
,
Ramalho
FS
,
Peralta
C
.
The current state of knowledge of hepatic ischemia-reperfusion injury based on its study in experimental models
.
J Biomed Biotechnol
.
2012
;
2012
:
298657
.
71.
Ozier
Y
,
Klinck
JR
.
Anesthetic management of hepatic transplantation
.
Curr Opin Anaesthesiol
.
2008
;
21
(
3
):
391
-
400
.
72.
Lord
JM
,
Midwinter
MJ
,
Chen
YF
, et al
.
The systemic immune response to trauma: an overview of pathophysiology and treatment
.
Lancet
.
2014
;
384
(
9952
):
1455
-
1465
.
73.
Toubai
T
,
Sun
Y
,
Reddy
P
.
GVHD pathophysiology: is acute different from chronic?
Best Pract Res Clin Haematol
.
2008
;
21
(
2
):
101
-
117
.
74.
Lee
SJ
,
Wolff
D
,
Kitko
C
, et al
.
Measuring therapeutic response in chronic graft-versus-host disease. National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: IV. The 2014 Response Criteria Working Group report
.
Biol Blood Marrow Transplant
.
2015
;
21
(
6
):
984
-
999
.
75.
Sebastian
M
,
Hsiao
CJ
,
Futch
HS
, et al
.
Obesity and STING1 genotype associate with 23-valent pneumococcal vaccination efficacy
.
JCI Insight
.
2020
;
5
(
9
):
e136141
.
76.
Yi
G
,
Brendel
VP
,
Shu
C
,
Li
P
,
Palanathan
S
,
Cheng Kao
C
.
Single nucleotide polymorphisms of human STING can affect innate immune response to cyclic dinucleotides
.
PLoS One
.
2013
;
8
(
10
):
e77846
.
77.
Walker
MM
,
Crute
BW
,
Cambier
JC
,
Getahun
A
.
B cell-intrinsic STING signaling triggers cell activation, synergizes with B cell receptor signals, and promotes antibody responses
.
J Immunol
.
2018
;
201
(
9
):
2641
-
2653
.
78.
Conlon
J
,
Burdette
DL
,
Sharma
S
, 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
.
2013
;
190
(
10
):
5216
-
5225
.
79.
Carroll
EC
,
Jin
L
,
Mori
A
, et al
.
The vaccine adjuvant chitosan promotes cellular immunity via DNA sensor cGAS-STING-dependent induction of Type I interferons
.
Immunity
.
2016
;
44
(
3
):
597
-
608
.
80.
Ramanjulu
JM
,
Pesiridis
GS
,
Yang
J
, et al
.
Design of amidobenzimidazole STING receptor agonists with systemic activity
.
Nature
.
2018
;
564
(
7736
):
439
-
443
.
81.
Zhang
Y
,
Sun
Z
,
Pei
J
, et al
.
Identification of α-mangostin as an agonist of human STING
.
ChemMedChem
.
2018
;
13
(
19
):
2057
-
2064
.
82.
Blaauboer
SM
,
Mansouri
S
,
Tucker
HR
,
Wang
HL
,
Gabrielle
VD
,
Jin
L
.
The mucosal adjuvant cyclic di-GMP enhances antigen uptake and selectively activates pinocytosis-efficient cells in vivo
.
eLife
.
2015
;
4
:
e06670
.
83.
Fu
J
,
Kanne
DB
,
Leong
M
, et al
.
STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade
.
Sci Transl Med
.
2015
;
7
(
283
):
283ra52
.
84.
Haag
SM
,
Gulen
MF
,
Reymond
L
, et al
.
Targeting STING with covalent small-molecule inhibitors
.
Nature
.
2018
;
559
(
7713
):
269
-
273
.
85.
Zhang
H
,
Tang
K
,
Zhang
Y
, et al
.
Cell-free tumor microparticle vaccines stimulate dendritic cells via cGAS/STING signaling
.
Cancer Immunol Res
.
2015
;
3
(
2
):
196
-
205
.
86.
Huang
KW
,
Hsu
FF
,
Qiu
JT
, et al
.
Highly efficient and tumor-selective nanoparticles for dual-targeted immunogene therapy against cancer
.
Sci Adv
.
2020
;
6
(
3
):
eaax5032
.
87.
Kinkead
HL
,
Hopkins
A
,
Lutz
E
, et al
.
Combining STING-based neoantigen-targeted vaccine with checkpoint modulators enhances antitumor immunity in murine pancreatic cancer
.
JCI Insight
.
2018
;
3
(
20
):
e122857
.
88.
Chattopadhyay
S
,
Liu
YH
,
Fang
ZS
, et al
.
Synthetic immunogenic cell death mediated by intracellular delivery of STING agonist nanoshells enhances anticancer chemo-immunotherapy
.
Nano Lett
.
2020
;
20
(
4
):
2246
-
2256
.
89.
Cheng
N
,
Watkins-Schulz
R
,
Junkins
RD
, et al
.
A nanoparticle-incorporated STING activator enhances antitumor immunity in PD-L1-insensitive models of triple-negative breast cancer
.
JCI Insight
.
2018
;
3
(
22
):
e120638
.
90.
Hou
Y
,
Liang
H
,
Rao
E
, et al
.
Non-canonical NF-κB antagonizes STING sensor-mediated DNA sensing in radiotherapy
.
Immunity
.
2018
;
49
(
3
):
490
-
503.e4
.
91.
Corrales
L
,
Woo
SR
,
Williams
JB
,
McWhirter
SM
,
Dubensky
TW
Jr.
,
Gajewski
TF
.
Antagonism of the STING pathway via activation of the AIM2 inflammasome by intracellular DNA
.
J Immunol
.
2016
;
196
(
7
):
3191
-
3198
.
92.
Shi
Y
,
Zheng
W
,
Yang
K
, et al
.
Intratumoral accumulation of gut microbiota facilitates CD47-based immunotherapy via STING signaling
.
J Exp Med
.
2020
;
217
(
5
):
e20192282
.
93.
Sallets
A
,
Robinson
S
,
Kardosh
A
,
Levy
R
.
Enhancing immunotherapy of STING agonist for lymphoma in preclinical models
.
Blood Adv
.
2018
;
2
(
17
):
2230
-
2241
.
94.
Wolf
D
,
Bader
CS
,
Barreras
H
, et al
.
Superior immune reconstitution using Treg-expanded donor cells versus PTCy treatment in preclinical HSCT models
.
JCI Insight
.
2018
;
3
(
20
):
e121717
.
95.
Luznik
L
,
O’Donnell
PV
,
Fuchs
EJ
.
Post-transplantation cyclophosphamide for tolerance induction in HLA-haploidentical bone marrow transplantation
.
Semin Oncol
.
2012
;
39
(
6
):
683
-
693
.
96.
Gao
P
,
Ascano
M
,
Wu
Y
, et al
.
Cyclic [G(2′,5′)pA(3′,5′)p] is the metazoan second messenger produced by DNA-activated cyclic GMP-AMP synthase
.
Cell
.
2013
;
153
(
5
):
1094
-
1107
.
97.
Cavlar
T
,
Deimling
T
,
Ablasser
A
,
Hopfner
KP
,
Hornung
V
.
Species-specific detection of the antiviral small-molecule compound CMA by STING
.
EMBO J
.
2013
;
32
(
10
):
1440
-
1450
.
98.
Kwon
D
,
Park
E
,
Sesaki
H
,
Kang
SJ
.
Carbonyl cyanide 3-chlorophenylhydrazone (CCCP) suppresses STING-mediated DNA sensing pathway through inducing mitochondrial fission
.
Biochem Biophys Res Commun
.
2017
;
493
(
1
):
737
-
743
.
Sign in via your Institution