Abstract
Background
TP53 mutations in acute myeloid leukemia (AML) are associated with resistance to
chemotherapy and poor prognosis. Patients with TP53-mutated AML respond to DNA
methyltransferase inhibitors (DNMTis) but responses are brief. Immunotherapeutic approaches
are being explored.
Stimulator of interferon genes (STING) is a key innate immune driver that activates interferon
(IFN) and nuclear factor Kappa B (NFkB)/tumor necrosis factor-alpha (TNF) signaling. STING
is regulated epigenetically, and we previously reported that DNMTi treatment increases basal
STING transcripts and epigenetically-silenced endogenous retroviruses (ERVs), increasing
cytosolic dsRNA and IFN signaling through a mechanism known as viral mimicry. Thus,
increasing basal STING expression with DNMTI treatment should increase substrate for
activation by STING agonists, making this a potentially attractive novel combination therapy in
AML. STING activation was also recently shown to trigger p53-independent apoptosis, and
STING agonists have shown a synergistic effect with BH3-mimetics in TP53-mutated AML cells.
Here we studied the next-generation allosteric STING agonist C92 from Curadev Pharma, which
potently binds and activates all human STING variants, in combination with the DNMTi
decitabine (DAC) in TP53-mutated AML.
Methods
To test the hypothesis that DNMTis synergize with STING agonists, we treated AML cell lines
with wild-type (WT) (MOLM-14, OCI AML1) and mutated TP53 (KG1, KASUMI, U937) and
patient samples (N=5-10) with C92 in combination with decitabine (DAC). We also utilized
CRISPR-Cas9 gene editing to knock out TP53 in MOLM-14 and OCI AML1 in order to confirm
observed results from mutated cell lines. To assess synergy of the two drugs, we utilized MTS
assays and the Chou-Talalay combination index. To identify the molecular pathways and genes
activated by the drug combination in TP53-mutated vs -WT AML, we performed ribosomal
depleted RNA-seq after C92 and/or DAC treatment. We validated gene expression using qPCR
of RNA, Western blotting of proteins and cytokine (including CXCL10, IFN a, b g, TNFa)
release by ELISA assays. To investigate apoptosis in TP53 KO vs WT, we measured Annexin V
labeling by flow cytometry pre and post treatment with the 2 drugs, administered alone and in
combination in the presence of the pan-caspase apoptosis inhibitor Z-VAD-FMK, the JAK/STAT
inhibitor ruxilitinib and the TBK1 inhibitor amlexanox. Finally, to test efficacy of the combination
therapy in vivo, we injected a humanized AML mouse model with MOLM-14 cells expressing
luciferase, and measured leukemia burden by non-invasive luciferin imaging. To measure
cytokines, T cells and myeloid in the leukemia microenvironment (LME), we performed IHC in
spleen and bone marrow, pre and post treatment. We also quantitated cytokines in the plasma
extracted from peripheral blood using ELISA assays.
Results
The STING agonist C92 was highly synergistic with DAC in AML cells, with combination index of
<1 in all AML cell lines, ranging from 0.1-0.8. This drug combination induces an interferon
signature in TP53-mutated AML cells, compared with TP53-WT, with increased expression
(p<0.05) of Th1 type chemokines (CXCL10, CXCL11), RNA detection- (DHX58, EIFAK2, IFIH1,
OASL, PARP9, RIGI), and interferon cell death-associated genes (IFI27, IFI44L, IFIT2, MX1,
OAS1, OAS2, OAS3, and TNFSF10). TP53-mutated AML cells, compared with TP53-WT,
induce interferon-driven cell death by apoptosis, as demonstrated by Z-VAD-FMK, ruxilitinib and
amlexanox treatment (p<0.05). Finally, C92 and DAC combination decreased the AML burden in
vivo and increased cytokines and T cells in the LME, compared to single-agent therapy and
vehicle controls (p<0.05).
Conclusion
Our results demonstrate that next-generation STING agonist C92 in combination with DNA
methyltransferase inhibitor induces interferon-driven apoptosis in TP53-mutated acute myeloid
leukemia, compared with TP53-WT AML. The more advanced analogue CRD3874-SI is
currently in a Phase I clinical trial in relapsed/refractory AML at UMGCCC, and our data here
support development of a clinical trial combining CRD3874-SI with DAC for patients with TP53-
mutated AML.
This feature is available to Subscribers Only
Sign In or Create an Account Close Modal