Abstract

Signal transducer and activator of transcription 3 (STAT3) is an attractive cancer therapeutic target. Unfortunately, targeting STAT3 with small molecules has proven to be very challenging, and for full activation of STAT3, the cooperative phosphorylation of both tyrosine 705 (Tyr705) and serine 727 (Ser727) is needed. Further, a selective inhibitor of STAT3 dual phosphorylation has not been developed. Here, we identified a low nanomolar potency and highly selective small-molecule STAT3 inhibitor that simultaneously inhibits both STAT3 Tyr705 and Ser727 phosphorylation. YY002 potently inhibited STAT3-dependent tumor cell growth in vitro and achieved potent suppression of tumor growth and metastasis in vivo. More importantly, YY002 exhibited favorable pharmacokinetics, an acceptable safety profile, and superior antitumor efficacy compared to BBI608 (STAT3 inhibitor that has advanced into phase III trials). For the mechanism, YY002 is selectively bound to the STAT3 Src Homology 2 (SH2) domain over other STAT members, which strongly suppressed STAT3 nuclear and mitochondrial functions in STAT3-dependent cells. Collectively, this study suggests the potential of small-molecule STAT3 inhibitors as possible anticancer therapeutic agents.
Short abstract
YY002 is the highly selective and potent small molecular inhibitor that simultaneously inhibited STAT3 Tyr705 and Ser727 phosphorylation, demonstrating promising antitumor activity.
Introduction
Signal transducer and activator of transcription 3 (STAT3) integrates signals from cytokines and growth factors into transcriptional responses in target cells. It mediates a complex spectrum of cellular responses, including proliferation, differentiation, and apoptosis.1 STAT3 is a key element in multiple oncogenic signaling pathways, which activate STAT3 by canonical and noncanonical pathways.2 The canonical pathway of STAT3 is activated by both receptor tyrosine kinases and nonreceptor tyrosine kinases upon stimulation with various growth factors or cytokines. The activated receptors phosphorylate the Tyrosine705 (Tyr705) residue of STAT3, which leads to STAT3 homodimerization, nuclear translocation and transcriptional activation. It was originally proposed that the reduction of STAT3 Tyr705 phosphorylation would prevent STAT3 homodimerization and consequently inhibit the activation of its target genes. Thus, the majority of drug development efforts have been focused on STAT3-Tyr705 phosphorylation inhibition. However, these inhibitors have limited antitumor activities.3,4
Apart from the well-studied Tyr705 activation (canonical pathway), STAT3 undergoes alternative post-translational modifications like phosphorylation of Serine 727 (Ser727),2 which mediates STAT3 translocating into mitochondria, and subsequent binding to the electron transport chain to regulate mitochondrial respiration and oxidative phosphorylation (OXPHOS)5,6 The Warburg effect describes the phenomenon that tumors are highly reliant on glycolysis for generating the intermediate metabolites to meet the demands of the tumors’ high rate of proliferation.7 The recent research also demonstrated that many tumors employ high levels of OXPHOS for energy metabolism and tumor survival.8−10 OXPHOS is an emerging drug development target in refractory cancers such as acute myeloid leukemia (AML),11,12 pancreatic cancer,13,14SMARCA4 mutant tumors,15 and so forth. Some inhibitors target OXPHOS by directly interfering with the electron transport chain (ETC) complex. Among them, IACS-01075916 and metformin,17 advanced to clinic trials in several advanced tumors. However, directly targeting the ETC has some unexpected toxicities in the clinic.18 Thus, the inhibition of STAT3 Ser727 phosphorylation, in turn indirectly inhibiting OXPHOS may provide an alternative approach to achieve metabolic synthetic lethality in cancer cells. In conclusion, the phosphorylation of STAT3 at Tyr705 along with Ser727 is required for full activation of STAT3. Developing a STAT3 inhibitor that simultaneously inhibits STAT3 Tyr705 and Ser727 phosphorylation would achieve an adequate inhibition of STAT3, and may achieve a highly potent therapeutic benefit for cancer treatment.
Pancreatic cancer is an aggressive malignancy with a 5-year survival rate of less than 9%,19 which is hard to diagnose and has high liver and lymph node metastasis rates.20 Up to date, the therapies of advanced pancreatic cancer are extremely lacking, and the current approved drugs are with limited clinical benefits.21,22 Therefore, it is urgent to search, identify and develop novel targeted drug candidates for advanced pancreatic cancer. Overexpression and persistent activation of STAT3 is tightly relevant to the poor prognosis of pancreatic cancer.23−25 STAT3 is consistently activated in pancreatic cancer in multiple ways. For instance, myeloid cells produce IL-6 to activate the JAK-STAT3 pathway that promotes the development of pancreatic ductal adenocarcinoma (PDAC) in a KrasG12D-driven pancreatic tumor mouse model.24,26 Besides, pancreatic-specific Kras and P53 mutations induce an accumulation of reactive oxygen species (ROS) in mouse pancreatic tumors, leading to activation of STAT3. Moreover, genetic approaches to eliminate STAT3 expression or administration of inhibitors of JAK2 or STAT3 contribute to decreasing tumor fibrosis and pancreatic stellate cells and altering the type of immune cells infiltrating the tumor.27
In this study, we used two functional assays, the STAT3-Luciferase reporter assay (based on pTyr705 nuclear transcriptional function) and the mitochondrial respiration assay (based on pSer727 mitochondrial OXPHOS function), to screen inhibitors that abolished STAT3 pTyr705 and pSer727 concurrently. After several rounds of screening and rational drug modifications, we identified a series of small-molecule STAT3 inhibitors. The most promising lead, YY002, a highly potent and selective STAT3 inhibitor, inhibited STAT3 Tyr705 and Ser727 phosphorylation, thereby abrogating the STAT3 nuclear and mitochondrial functions. YY002 potently inhibited pancreatic cancer growth and metastasis in vitro and in vivo. Taken together, this work not only revealed a novel STAT3 Tyr705 and Ser727 phosphorylation inhibitor as a promising new therapeutic agent for pancreatic cancer, but also provided insights into approaches to the development of STAT3 Tyr705 and Ser727 phosphorylation inhibitors.
Material and Methods
Chemical, Cell Lines, Culture and Reagents
Detailed information on chemicals and cell lines is described in Supporting Information and Methods. All cell lines were maintained in incubators containing 5% CO2 at 37 °C. The AQueous One Solution Cell Proliferation Kit was purchased from Promega (MTS reagent, Madison, WI).
STAT3 Knockdown
The shRNAs for silencing STAT3 are listed in Supplementary Table S1. ShRNAs were respectively inserted into pLKO.1 vector, then were cotransfected into 293T cells with the packing plasmids pMD2G and pXPAX2, using Lipofectamine 2000 (Thermo Fisher Scientific, Beijing, China). After transfection for 72 h, the harvested lentiviruses were infected into tumor cell lines and puromycin was added to screen for stable STAT3 knockdown cells. Finally, the knockdown efficiency was detected by Western blot.
Plasmid and Protein Expression
STAT1 (amino acid residues 127–716), STAT2 (amino acid residues 138–702), STAT3 (amino acid residues 127–722), STAT4 (amino acid residues 133–705), STAT5B (amino acid residues 136–703) and STAT6 (amino acid residues 113–658) were cloned into the pET28A vector. STAT3-SH2 (amino acid residues 586–685) and the mutants of the STAT3-SH2 produced by site-directed mutagenesis were also cloned into the pET28A vector. All recombinant human fusion proteins were expressed in E. coli BL21 (DE3). Then, according to different induction conditions, the protein expression was induced. During the induction of the expression of STAT3-SH2 and all the mutants, STAT1, STAT2, STAT3 and STAT6, when the OD value reached about 0.6–1.0, the E. coli was transferred to 22 °C and 0.5 mM Isopropyl β-D-1-thiogalactopyranoside (IPTG) was added. For the expression of STAT4 and STAT5B when the OD value reached about 0.5–0.6, the E. coli was transferred to 30 °C for 2.5–3 h and 0.2 mM IPTG was added.
Docking Assay
Docking was performed by Schrödinger Glide software (New York, NY, USA). The induced fit docking (IFD) protocol was performed on compound YY002 using the induced fit tool in Maestro 11.5 from Schrödinger. IFD employed the use of Glide and Prime for ligand docking and protein refinement, respectively. The active site was centered on the Tyr705 phosphorylated site near the STAT3-SH2 domain (PDB code 6QHD. Deposited: 2019-01-16; Released: 2019-06-19). Induced fit protein–ligand complexes were generated using Prime and further subjected to side chain and backbone refinement.
Microscale Thermophoresis (MST) Assay
Microscale thermophoresis (MST) was conducted as previously described.28 Binding affinities of YY002, Stattic against purified STAT3127–722 and STAT3-SH2 protein or YY002 against STAT3-SH2 WT and mutants were measured by a Monolith NT.115 (Nanotemper Technologies). The proteins were fluorescently labeled according to the manufacturer’s procedure and kept in the MST buffer (50 mM HEPES, pH 7.0, 500 mM NaCl, 0.01% NP40, 50 mM l-argine) at a concentration of 200 nM. Next, the RED fluorescent dye was added, mixed and incubated for 30 min at 25 °C in the dark. For each assay, the labeled protein was mixed with the same volume of unlabeled compound at 16 different serially diluted concentrations at room temperature. The samples were then loaded into premium capillaries (NanoTemper Technologies) and measured at 25 °C using 20%–40% LED power and medium MST power. Each assay was repeated two or three times. Data analyses were performed using MO. Affinity Analysis v.2.2.4 software. All figures were made by GraphPad Prism 7.0.
Surface Plasmon Resonance (SPR) Assay
Surface plasmon resonance (SPR) experiments were performed with a Biacore 8K instrument (Cytiva) with CM5 sensor chip (Cytiva). To test YY002 binding of STAT3 protein, serially diluted concentrations of YY002 were injected into the flow system. Experiments were conducted using the buffer: 10 mM PBS, pH7.4, 137 mM NaCl, 2.7 mM KCl, 1 mM DTT, 0.05% P20, 5% DMSO. Recombinant full-length human STAT3 (Creative Biomart, Catalog: STAT3–001H, Batch number: PSS1072207) was immobilized on the sensor chip (CM5) using the amine-coupling method according to standard protocols. The analyte was injected at the flow rate of 30 μL/min. The association time was 180 s and the dissociation time was 180 s. Since YY002 was dissolved in PBS containing 5% dimethyl sulfoxide and a solvent correction assay was performed to adjust the results. YY002 at various concentrations was injected into the flow system. The KD values were calculated with the kinetics and affinity analysis option of Biacore evaluation software.
Colony Formation
Tumor cells were seeded into 6-well plates, allowing attachment overnight. The test compounds were added at indicated concentrations for 1 week of treatment. Then colonies were fixed by 4% paraformaldehyde (PFA) and then stained with 0.2% crystal violet. Images were photographed using a digital camera, and colonies were quantified by manual counting.
Western Blot
Pancreatic cancer cells were lysed in RIPA buffer supplemented with 1 mM phenylmethylsulfonyl fluoride (PMSF), a proteinase inhibitor cocktail and a phosphatase inhibitor cocktail (Sigma). Lysates were separated by 8–12% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to nitrocellulose. The blots were probed with specific antibodies followed by a secondary antibody, and then membranes were examined by the LI-COR Odyssey infrared imaging system (LI-COR Biotechnology, Lincoln NE). The antibodies used and their technical information are listed in Supplementary Table S2.
Real-Time Quantitative PCR (RT-qPCR)
Total RNA was isolated using TRIzol reagent (Takara, Japan). RNA was used to generate single-stranded cDNA according to the manufacturer’s protocols of the cDNA Reverse Transcription Kit (Takara, Japan) and then subjected to polymerase chain reaction (PCR) using SYBR Green Master Mix (Thermo Fisher Scientific). The expression of VEGF, Cyclin D1, c-Myc and Bcl-2 in Capan-2, HPAC, and BxPC-3 cells was validated by real-time quantitative PCR. The sequences of Real-time PCR primers are listed in Supplementary Table S3.
Luciferase Reporter Assay
The STAT3 luciferase plasmids (pGL4.47 luc2p/SIE/Hygro) vector (E4041, Promega) and Renilla plasmids were cotransfected into HEK293T with Lipofectamine 2000 reagent (Invitrogen) and incubated for 20–24 h. Then, the cells were seeded into 96-well plates at a cell density of 2 × 104 per well. After overnight attachment of the cells, compounds were added with concentration gradient, and IL-6 (20 ng/mL) was used as an activator of STAT3. Luciferase assays were performed by using the Dual Luciferase Reporter Assay System (Promega) according to manufacturer’s instructions. The firefly luciferase activities were normalized against Renilla luciferase activities.
Immunofluorescence Assay
BxPC-3 and Capan-2 cells were cultured on glass coverslips at a density of 105 per well of 24-well plate. After treating with YY002 for 24 h, IL-6 was added for 30 min stimulation. Then, the culture medium was removed, and the cell slides were fixed with 4% paraformaldehyde for 15 min and permeated by 0.2%Triton X-100 for 20 min. Next, after PBS wash and blocking in 0.5% BSA, primary antibodies were added to incubate with cells overnight at 4 °C, and then, cells were incubated with corresponding secondary antibodies for 1 h at room temperature. Finally, DAPI was added and a confocal microscope (Leica) was used to record images.
Mitochondrial Bioenergetics Analysis
Mitochondrial bioenergetics were evaluated by oxygen consumption rate (OCR) and extracellular acidification rate (ECAR), using the Seahorse XFe96 analyzer (Agilent Technologies). OCR reflected oxidative phosphorylation, and during the experiment, 1 × 104 to 2 × 104 cells per well were seeded onto the cell culture plates in the kit. After treatment with YY002 for 1 h, the medium in the plates was replaced by the newly configured detection medium through the liquid exchange program of the Seahorse instrument. Then, following basal respiration, the mitochondrial effectors (oligomycin, FCCP, and a mixture of rotenone and antimycin A) were injected sequentially according to the manufacturer’s protocol to measure the ATP-linked respiration and spare respiratory capacity. During the process of ECAR (which reflected glycolysis), the density of cells per well and the time of YY002 treatment were consistent with experiments to measure OCR. Glucose, oligomycin and 2-DG were injected sequentially to evaluate the level of cellular glycolysis and the ability of cells to restore glycolysis.
Animal Studies
The mice were obtained from the Animal Center of East China Normal University. All animal experiments conformed to the Animal Investigation Committee of the Institute of Biomedical Sciences, East China Normal University. NOD/SCID, male, 6–8-week-old mice were injected subcutaneously with 5 × 106 PANC-1 and MIA Paca-2 cells in a mixture of PBS with 50% Matrigel for tumor growth. Female Balb/c Nude was inoculated with 1 × 107 MDA-MB-231 tumor cells (in 0.15 mL, 1:1 with Matrigel) at the right flank. Male CB17 SCID was inoculated with 3 × 106 SUDHL-1 tumor cells (in 0.1 mL, 1:1 with Matrigel) at the right flank. Tumor-bearing mice was randomly assigned to different experimental groups for experiments after the tumor nodules grew to 100–200 mm3 and the dose formulation of YY002 was as indicated. The tumor volume and mouse body weight were measured every 4–5 days. The tumor volume was calculated using the following equation: tumor volume (V) = length × width × width × 0.52. In each group, the relative tumor volume (RTV) was calculated as RTV = Vt/V0 (Vt represents the tumor volume at the end of the experiment, V0 represents the tumor volume at the beginning of the treatment). The tumor growth inhibition (TGI) was calculated using the following formula: TGI = (1 – RTVthe treated group /RTV the control group) × 100%. At the indicated time-points, mice were euthanized and tissue samples were collected for analyses.
Orthotopic Pancreatic Cancer Tumor Model
Murine pancreatic cancer cells, PAN02-luciferase, suspended into the precooled Matrigel (BD biosciences) were injected (5 × 105 cells per mouse) into the pancreas of male, 6–8-week-old, C57BL/6 mice. The fluorescence was detected 1 week after tumor cell implantation by an IVIS Imaging System (Xenogen Corporation, Alameda, CA) and the mice were divided into 4 groups according to the fluorescence value, and keep the average luminescence values of each group consistent when mice were grouped. The average luminescence values of each group when grouped were as follows: 307714.29 photon/sec2/cm2/sr (Control), 301142.86 photon/sec2/cm2/sr (5 mg/kg), 302500 photon/sec2/cm2/sr (10 mg/kg), 303414.29 photon/sec2/cm2/sr (10 mg/kg). The dose formulation of YY002 was as indicated. Then, the bioluminescence of cancer cells and body weight of mice were observed once a week during the treatment by IVIS. In addition, we used AsPC-1 cells to construct another orthotopic model of pancreatic cancer (similar to PAN02-Luci), and 2 × 106 cells were injected into male, 6–8-week-old Balb/c mouse. Another independent animal experiment was performed to determine whether YY002 prolongs the survival of mice.
Liver Metastasis Model of Human Pancreatic Cancer
Murine pancreatic cancer cells, PAN02-luc, suspended into the precooled Matrigel (BD Biosciences) were injected (5 × 105 cells per mouse) into the spleen of male, 6–8-week-old, C57BL/6 mice. The fluorescence was detected after tumor cell implantation for 1 week by an IVIS Imaging System (Xenogen Corporation, Alameda, CA), and the mice were divided into 4 groups according to the fluorescence value. The average luminescence values of each group were kept consistent when mice were grouped. The average luminescence values of each group when grouped were as follows: 45328.57 photon/sec2/cm2/sr (Control), 44522.29 photon/sec2/cm2/sr (5 mg/kg), 45885.71 photon/sec2/cm2/sr (10 mg/kg), 45742.86 photon/sec2/cm2/sr (10 mg/kg). The dose formulation of YY002 was as indicated. Then, the bioluminescence and body weight of mice were observed once a week during the treatment by IVIS. Another independent animal experiment was performed to determine whether YY002 prolongs the survival of mice.
Statistical Analysis
Experiments were carried out with three or more replicates. Statistical analyses were done by Student’s t test. P values < 0.05 were considered significant. The differences between control and experimental groups were determined by one-way ANOVA. Since treatment and time course were investigated, two-way ANOVA followed by post hoc test was also applied. Data were expressed as means and 95% confidence intervals and P < 0.05 was considered significant. All analyses were performed using Microsoft Excel 2010 and GraphPad Prism 7 software.
Results
Development of Highly Potent and Selective STAT3 Inhibitors
To specifically identify STAT3 inhibitors, structure-based virtual screening and computer-aided drug design were utilized. And then, we generated a STAT3-specific luciferase reporter system to confirm compounds capable of inhibiting STAT3 Tyr705 and Ser727 phosphorylation (Supplementary Figure S1A–C) and a mitochondrial respiration assay (Supplementary Figure S1D, S1E). Through these preliminary assays, the small molecule YY002 (Figure 1A, Supplementary Figure S1) exhibited the best inhibitory activity among the investigated compounds. YY002 inhibited STAT3 luciferase reporter expression (Figure 1B), ATP production (Figure 1C) and OXPHOS (Figure 1D) in a concentration-dependent manner, with an inhibitory concentration (IC50) ranging from 1 to 10 nM. We next purified the STAT3-SH2 domain (SH2D) and STAT3127–722 to confirm whether YY002 directly interacted with STAT3. In the Microscale Thermophoresis (MST) assay, YY002 directly bound STAT3SH2 and STAT3127–722 with a KD of 2.24 ± 0.55 nM and 18.27 ± 4.04 nM, respectively (Figure 1E). The surface plasmon resonance (SPR) assay also proved that YY002 bound to STAT3 with high affinity (Figure 1F). To further determine whether YY002 selectively bind to STAT3 over STAT family proteins, we purified STAT family proteins and performed direct binding assays between YY002 and STATs protein in MST assays. YY002 exhibited a high selectivity against STAT3 with no significant binding affinities to other STAT family proteins (Figure 1G). To further evaluate the dependency of YY002 action on STAT3, we constructed shRNA-mediated STAT3 knockdown stable cell lines, and in cell viability assays, both lines of shSTAT3 PDAC cells (Capan-2, PANC-1) saw a loss of YY002 treatment efficacy upon STAT3 knockdown (Figure 1H–J). This indicates a general lack of off-target effects of YY002 on cell viability. Subsequently, we identified the JAK-STAT3 signaling pathway as significantly affected upon YY002 treatment (Figure 1K). In brief, these results demonstrated that YY002 selectively suppressed the proliferation of tumor cells by targeting STAT3.
Figure 1.
YY002 selectively targets STAT3. (A) Structure of YY002. (B) YY002 inhibited the STAT3 luciferase reporter activity. IL-6 was used as an activator of STAT3 (n = 2). (C) YY002 inhibited the ATP production (n = 2). (D) YY002 inhibited the OXPHOS rate (OCR = oxygen consumption rate, n = 2). (E) Direct binding of YY002 to STAT3SH2 or STAT3127–722 was determined by MST experiments (n = 3). (F) The binding affinities between YY002 and STAT3 was detected by surface plasmon resonance (SPR) assay. (G) YY002 selectively bound to STAT3 and other STAT members (n = 3). (H) The shRNA knock-down efficiency in PANC-1 and Capan-2 was confirmed by Western Blot. (I, J) The pancreatic cancer cell lines shNC, shSTAT3-1# and shSTAT3-2# of PANC-1 (I) and Capan-2 (J) were treated with YY002, and the proliferation was determined after 72 h of treatment (n = 3). (K) BxPC3 cells were treated with 50 nM YY002 for 24 h, and then RNA-Seq assay was performed. The results were analyzed by gene set enrichment analysis (GSEA). (L) Capan-2 cells were treated with different concentrations of STAT3 inhibitors or YY002, and the proliferation was tested by MTS (n = 2). (M) YY002 and Stattic directly bound to STAT3127–722, The binding affinities were determined by MST experiments (n = 3). Data shown as mean ± SD ns, P > 0.05, *P < 0.05, **P < 0.01, *** P < 0.001 and **** P < 0.0001 by one-way ANOVA following multiple comparison.
Currently, the majority of STAT3 inhibitors merely inhibited STAT3 Tyr705 phosphorylation, and most of them had poor inhibitory activities. So, we compared the inhibitory activity of a STAT3 dual-phosphorylation inhibitor YY002, with the several representative STAT3 inhibitors including STAT3-SH2D inhibitors (C188-9, BP-1–102, Stattic, FLLL32), JAK2 inhibitor (AZD1480), and STAT3 and cancer stemness inhibitor (BBI-608). YY002 exhibited markedly potent antitumor activity and direct binding affinities than the above STAT3 inhibitors; these activities were approximately 1000-fold better than the tested alternatives (Figure 1L,M, Supplementary Figure S2A).
YY002 Directly Binds to the STAT3-SH2 Domain
To illustrate a structural basis for the selective and high binding affinity between YY002 and STAT3, we performed a computational docking and molecular dynamics simulation assay of YY002 and the STAT3 Src homology 2 domain (SH2D), which revealed that the predominant binding mode was within the STAT3-SH2D (Figure 2A). Besides, YY002 had a broader interaction interface with STAT3-SH2D compared with the existing STAT3 inhibitors (Supplementary Figure S2B,C). Specifically, YY002 formed an extensive hydrogen-bonding network with the side chains of the residues of Ser636, Val637, Glu638, Asp647, and Ile659. The residues of Trp623, Ile634, and Gln644 may interact with YY002 through hydrophobic interactions (Figure 2A). Subsequently, we validated the predicted amino acids that YY002 directly bound to by site-directed mutagenesis and determining the binding affinities between the STAT3-SH2D mutants and YY002. As our results showed, the binding affinities between YY002 and the SH2D mutants including K626A, Y640A, and Q644A were only modestly changed compared with the binding affinities of YY002 to SH2D-WT (Figure 2B,C). In contrast, YY002 did not bind several mutants including S636A, V637A, E638A, N647A, I634A and I659A (Figure 2B,C). Thus, computational modeling and direct binding results demonstrated that YY002 directly bound to the residues of S636, V637, E638, N647, I634, and I659. To further confirm whether YY002-binding amino acids were responsible for the anticancer activity of YY002, we overexpressed each mutant in STAT3 stable knock-down PANC-1 cells and treated the above cells with YY002, however, overexpression of the mutants, except for an activated STAT3 mutation N647I29 that harbors high pSTAT3, failed to restore the anticancer capacity of YY002 (Figure 2D,E), demonstrating YY002-bound amino acids directly regulated its antipancreatic cancer activity. Furthermore, this led us to investigate if these amino acids also play a crucial role in the function of STAT3. Thus, we performed a STAT3 reporter luciferase assay and found that some mutants such as N647A, I659A, E638A, and V637A failed to respond to IL-6 stimulation of STAT3 transcription (Supplementary Figure S2D), indicating that these amino acid residues are critical for STAT3 transcription.
Figure 2.
YY002 directly binds to STAT3 SH2 domain. (A) Computer docking model predicting that YY002 bound to the STAT3-SH2 domain. (B, C) YY002 binding to STAT3-SH2 and its indicated mutants. The binding affinities were measured by MST experiments (n = 3). (D) PANC-1 shSTAT3-1# cells that were infected with indicated lentivirus vectors to reintroduce specific SH2 mutants were treated with indicated YY002 concentrations, and cell viability was measured (n = 3). Data shown as mean ± SD. (E) Re-expression of STAT3 mutant in STAT3 knockdown PDAC cells. The shSTAT3-1# PANC-1 cells that were infected with the indicated lentivirus expression vectors, and the transfection efficacy was detected by Western blots. (F) In vitro enzyme inhibition assays.
Taken together, YY002 had a broader interaction interface with STAT3-SH2D, the binding residues and model of YY002 to STAT3-SH2 domain were clarified by computational modeling and direct binding assays. The further results demonstrated that these amino acids were essential for the YY002 binding capabilities and antitumor functions. Moreover, owing to the specific binding between YY002 and STAT3, YY002 also showed no obvious inhibition on tyrosine kinases or serine kinases (Figure 2F).
YY002 Inhibits Cancer Cell Proliferation, STAT3 Tyr705 Phosphorylation and STAT3 Nuclear Functions
Given that the rapid proliferation of cancer cells is one of the main drivers responsible for tumor progression, we next investigated the antitumor activity of YY002 against several pancreatic cancer cell lines. Because STAT3 is frequently activated in human tumors, it is conceivable that high pSTAT3 (Tyr705 and Ser727) cell lines are more strongly dependent on STAT3 signaling for survival, and that inhibition of STAT3 by YY002 may achieve strong growth inhibitory activity. YY002 potently suppressed pancreatic cancer cells that were with high pSTAT3 (Tyr705 and Ser727), with inhibitory concentrations (IC50) ranging from 3 to 11 nM (Figure 3A,B). While, in the low level of p-STAT3 cell lines, including two cancer cell line PC3 and KG1, and normal cell line HUVEC and HAF, YY002 was relative insensitive to these cell lines (Figure 3A,B). Moreover, YY002 inhibited colony formation of multiple STAT3-dependent PDAC cells at concentration of 1 to 10 nM (Supplementary Figure S3A), while YY002 had little impact on STAT3 negative PC3 cells (Supplementary Figure S3B). These results further demonstrated that inhibition of phosphorylated STAT3 (Tyr705 and Ser727) byYY002 is responsible for the anticancer activity of YY002.
Figure 3.
YY002 inhibits pancreatic cancer cell growth, STAT3 Tyr705 phosphorylation and STAT3 nuclear function. (A) YY002 inhibited the proliferation of pancreatic cancer cell lines but only had a limited effect on normal cells (n = 2). (B) The protein expression level of STAT3, p-STAT3Tyr705 and p-STAT3Ser727 in different cell lines. (C) STAT3 phosphorylation and downstream gene expression were measured by Western Blot in Capan-2, HPAC and BxPC-3 cells after treatment with YY002. (D) Down-stream gene expression was measured by quantitative real-time PCR in Capan-2, HPAC and BxPC-3 cells after treatment with YY002 (n = 2). (E) YY002 inhibited the entry of STAT3 into the nucleus of BxPC3, Capan-2 and PANC-1 cells. Scale bar, 50 μm. Data shown as mean ± sd. ns, P > 0.05, *P < 0.05, **P < 0.01, ***P < 0.001 and ****P < 0.0001 by one-way ANOVA following multiple comparison.
We next evaluated the efficacy of YY002 on STAT3 phosphorylation and the expression of its downstream genes on several pancreatic cancer cells. It is widely believed that Tyr705 phosphorylation is closely involved in STAT3 canonical nuclear function, whereas Ser727 phosphorylation regulates mitochondrial electron transfer chain activity. Distinct from most of the reported STAT3 inhibitors, YY002 showed concurrent inhibition of STAT3 Tyr705 and Ser727 phosphorylation (Figure 3C). At the same time, YY002 significantly suppressed STAT3 downstream gene expression in pancreatic cancer cells (Capan-2, HPAC and BxPC-3) (Figure 3C,D). Next, we deduced that YY002 bound to STAT3-SH2D suppressed STAT3 dimerization and reduced STAT3 nuclear translocation. We validated this hypothesis by immunofluorescence experiments. The classical STAT3 stimulatory molecule IL6 promoted STAT3 entry into the nucleus, whereas YY002 treatment significantly inhibited STAT3 nuclear translocation (Figure 3E). In summary, YY002 selectively inhibited pancreatic cancer cell proliferation and suppressed STAT3 Tyr705 and Ser727 phosphorylation. Finally, YY002 inhibited STAT3 classical nuclear function, thus restraining the oncology-related gene expression.
YY002 Inhibits STAT3 Ser727 Phosphorylation and Mitochondrial OXPHOS
YY002 significantly inhibited STAT3-Ser727 phosphorylation and mitochondrial STAT3 was prevalently phosphorylated at Ser727, which enhances its mitochondrial functions. We speculated that YY002 would inhibit the STAT3 functions in the mitochondria by suppressing pSTAT3-Ser727. We first investigated the effects of YY002 treatment on pancreatic cancer’s two major energy-generating pathways: mitochondrial oxidative phosphorylation (OXPHOS) and glycolysis. To this end, we measured the oxygen consumption rate (OCR), an indicator of OXPHOS, and the lactate production rate, an indicator of glycolysis (extracellular acidification rate; ECAR). As shown in Figure 4A, YY002 suppressed OXPHOS in pancreatic cancer cells in a concentration-dependent manner.
Figure 4.
YY002 inhibits STAT3 Ser727 phosphorylation and mitochondrial OXPHOS. (A) Oxygen consumption rate (OCR) was evaluated by the Seahorse XF96 extracellular flux analyzer and OXPHOS was inhibited in BxPC-3 (n = 2), CFPAC-1 (n = 3) and Capan-2 (n = 4) cells treated with YY002. (B) Knockdown of STAT3 in CFPAC-1 inhibited OXPHOS (n = 3). (C) CFPAC-1 shSTAT3-1# cells were treated with YY002 or vehicle (DMSO), and the level of OXPHOS was measured (n = 3). (D, E) The glycolysis level of BxPC-3 (D) and CFPAC-1 (E) cells treated with different concentrations of YY002 was determined (n = 3). (F) A brief description of the mitochondrial electron transport chain. (G, H) Capan-2 cells were treated with 50 nM YY002 or vehicle control and injected with the indicated drugs 1, 2, 3, 4 sequentially, and the mitochondrial respiration was determined by Seahorse instrument assay (n = 3). Data shown as mean ± sd.
To confirm whether the above inhibitory effect was due to the direct inhibition of STAT3, STAT3 stable knockdown pancreatic cancer cells were constructed, and the OXPHOS was significantly inhibited upon STAT3 stable knockdown (Figure 4B). Further, we treated STAT3 knockdown cells with YY002, and showed no significant inhibition of OXPHOS (Figure 4C). Subsequently, we reintroduced wild-type STAT3 or the indicated SH2 mutants in stable STAT3 knock-down cells. In WT-STAT3 re-expression cells, YY002 inhibitory capacity was restored, and this effect was specific because it did not fully restore upon expression of the indicated SH2 mutant or vector plasmids (Supplementary Figure S4A, S4B). Moreover, mitochondrial membrane potential (MMP) was also drastically affected in cells treated with YY002 (Supplementary Figure S4C). In contrast, YY002 showed no significant impact on glycolysis in PDAC cells (Figure 4D,E). Taken together, YY002 inhibited STAT3 Ser727 phosphorylation, and then interfered with mitoSTAT3 and its mitochondrial function.
Recent studies showed that STAT3 enters mitochondria and directly binds to electron transport chain complexes to modulate mitochondrial oxidative phosphorylation (Figure 4F). We next confirmed whether YY002 specifically impaired electron transfer chain complexes to inhibit OXPHOS. The electron transfer chain experiments confirmed that treatment of detergent-permeabilized cells with YY002 in medium supplemented with pyruvate and malate (to generate NADH for use by complex I) resulted in an attenuated oxygen consumption rate, whereas when the medium was supplemented with succinic acid to supply complex II, the OCR was not affected by YY002 treatment, thus bypassing the requirement for complex I function, all these results indicated that YY002 selectively impaired complex I functions in PDAC cells (Figure 4G,H). Studies have shown that the inhibition of complex I significantly induces the production of high levels of reactive oxygen species (ROS), leading to oxidative stress.30 We used the ROS dye 2′,7′-Dichlorodihydrofluorescein diacetate (DCFH-DA) to further investigate the effect of YY002 on ROS levels. As expected, YY002 significantly increased ROS levels in PDAC cells (Supplementary Figure S4D).
YY002 induction of mitochondrial STAT3 dysfunction as well as suppression of cancer cell OXPHOS suggested that cancer cells may be more sensitive to the cytotoxic effects of YY002 under conditions of increased dependence on mitochondrial activity. Indeed, combination YY002 with an inhibitor of glucose uptake and metabolism, 2-deoxy-d-glucose (2-DG), increased its cytotoxic effects in pancreatic cancer cells (Supplementary Figure S4E,F). Besides, YY002 robustly inhibited PDAC cells growth in galactose-containing medium, wherein cells were rendered dependent on OXPHOS for survival31 (Supplementary Figure S4G).
In summary, YY002 targeted STAT3 to inhibit oxidative phosphorylation, but not glycolysis, and further experiments demonstrated that YY002 directly inhibited the function of ETC complex I.
YY002 Potently Suppresses the Pancreatic Tumor Growth in Vivo
To evaluate the antitumor effect of YY002 in vivo, we first determined the pharmacokinetic properties of YY002. The plasma exposure and pharmacokinetic parameters were detected following oral and intravenous administration at 10 and 1 mg/kg, respectively. The Cmax of YY002 was 4307.80 ng/mL in mice under oral administration. The t1/2 value of oral administration of YY002 was 14.30 h. After intravenous administration, YY002 Cmax was 8154.40 ng/mL at a mean t1/2 of 16.60 h (Supplementary Figure S5A). These results proved that YY002 had an acceptable biological half-life and a moderate time–concentration curve. Importantly, YY002 exhibited well oral bioavailability with a rate of 31.30% in mice (Supplementary Figure S5A).
The drug-like properties of YY002 were also evaluated. For instance, YY002 was stable in human, murine, canine, rat, and monkey plasma, as well as in liver microsomes (Supplementary Figure S5B). In a general safety panel, YY002 showed no obvious inhibition of the human Ether-à-go-go-Related Gene (hERG) and cytochrome P450 enzymes (Supplementary Figure S5C).
To evaluate the antitumor growth activities of YY002 against pancreatic cancer in vivo, we established an animal model of pancreatic cancer using PANC-1, and set the dose of YY002 from 5 mg/kg/d to 20 mg/kg/d, which was administered orally. As the results show, both the tumor volume (Figure 5A,B) and tumor weight (Figure 5C) were significantly inhibited after YY002 treatment. At the dose of 20 mg/kg, YY002 caused a significant potent tumor regression in vivo, and the tumor growth inhibition (TGI) rate was 93.56 ± 1.58%. In another mouse xenograft model, similar results were observed. YY002 suppressed MIA PaCa-2 tumor growth in a concentration-dependent manner (Figure 5D–F). The TGI rates of each dose were 61.24 ± 9.68% (20 mg/kg), 70.91 ± 7.84% (10 mg/kg), and 85.06 ± 9.28% (5 mg/kg). FLLL32 was a represented STAT3 inhibitor that suppressed the tumor growth of breast cancer, PDAC in vitro and in vivo.32 To examine the efficacy of YY002 and FLLL32 at inhibiting tumor growth in vivo, we established an animal model of pancreatic cancer using BxPC3 cells. Both of YY002 or FLLL32 treatment caused a significant inhibition in tumor growth compared with vehicle treatment. Moreover, YY002 was orally available, and also achieved superior inhibition of tumor growth in vivo upon comparing with FLLL32 treatment groups (Supplementary Figure S6).
Figure 5.
YY002 inhibits pancreatic cancer growth in vivo. (A, B) The PANC-1 tumor volumes of mice were recorded every 4–5 days (n = 8). Data shown as mean ± SEM * P < 0.05, * * P < 0.01 and **** P < 0.0001 by One-way ANOVA followed multiple comparison. (C) At the end of the experiment, the tumors in each group were excised, weighed and counted. (D-E) MIA PaCa-2 cells were injected into mice which were treated with different concentrations of YY002 orally (n = 8). (F) The tumor volume was recorded every 4 days and after 24 days of administration, the tumors were harvested. Data shown as mean ± SEM * P < 0.05, * * P < 0.01 and **** P < 0.0001 by One-way ANOVA followed multiple comparison. At the end of the experiment, the tumors in each group were excised, weighed and counted. (G) Quantification of STAT3, pSTAT3Tyr705, and pSTAT3Ser727 immuno-staining and representative images of PANC-1 tumor.
Of note, the effect of YY002 therapy was well tolerated, since no animals in any group exhibited systematic toxicity in these studies (Supplementary Figure 7A,B). The liver enzymes, such as alanine aminotransferase (ALT) and aspartate aminotransferase (AST), were detected in two groups of mice treated with vehicle and YY002, respectively. The data showed that YY002 did not induce hepatic toxicity after treatment with YY002 (Supplementary Figure 7C). Besides, there was also no significant difference in serum blood urea nitrogen (BUN) and creatinine (CREA) levels between vehicle and YY002 groups (Supplementary Figure 7C). The hematology analyses also revealed that YY002 treatment did not have a significantly influence in the counts of white blood cells, lymphocytes, monocytes and neutrophils versus control mice (Supplementary Figure 7D). Altogether, YY002 is well tolerated in vivo.
To further investigate whether YY002 inhibits STAT3 phosphorylation in vivo, we detected STAT3, pSTAT3 (Tyr705) and pSTAT3 (Ser727) in PACN-1 tumors. Pharmacodynamic (PD) analysis demonstrated that YY002 did not suppress STAT3 expression in vivo (Figure 5G, Supplementary Figure S7E). Besides, a p.o. dose of YY002 at 5 mg/kg to 20 mg/kg significantly suppressed the phosphorylation of STAT3 at Tyr705 and Ser727 in PDAC tumors (Figure 5G, Supplementary Figure S7F,G). These effects were consistent with the results of YY002 on pancreatic cancer cell lines. Taken together, these data suggest that YY002 results in strong growth inhibitory activities in PDAC tumor models, and our PD analysis demonstrated that YY002 simultaneously suppressed the phosphorylation of STAT3 at Tyr705 and Ser727 in vivo.
YY002 Inhibits Pancreatic Cancer Metastasis
Owning to the potent antitumor growth effects in vivo, and recent reports suggesting that STAT3 is involved in key steps of pancreatic cancer metastasis,33,34 we next tested the effects of YY002 administration on pancreatic tumors in vivo using an orthotopic pancreatic cancer mouse model and a liver metastatic mouse model. In the orthotopic pancreatic cancer mouse model constructed by PAN02, YY002 inhibited tumor growth and metastasis in a concentration-dependent manner (Figure 6A). Since YY002 markedly inhibited tumor growth and metastasis in an orthotopic pancreatic cancer mouse model, we therefore carried out further experiments to confirm whether YY002 inhibited liver metastasis of pancreatic tumors. Advanced pancreatic cancer frequently carries fatal liver metastases with no effective treatment available. By injecting PAN02 cells into the mouse spleen, the tumor directed metastasis to the liver. And YY002 significantly inhibited tumor signal (Figure 6B). We hypothesized that YY002-mediated suppression of metastasis of pancreatic cancer would prolong survival of tumor-bearing mice. To assess this, we determined survival rates of mice that received vehicle or drug treatments in the independent pancreatic mouse metastatic models. YY002 significantly prolonged the survival of the tumor-bearing mice that constructed by PAN02 cells (Figure 6C,D). Besides, YY002 also significantly increased overall survival in the orthotopic model constructed from another highly metastatic human pancreatic cancer cell line AsPC-1, and 60% of mice in the high concentration group survived (Figure 6E). In conclusion, we demonstrated that YY002 strongly inhibited pancreatic tumor metastasis in vivo and significantly prolonged the survival of mice with pancreatic tumor metastasis in a preclinical pancreatic cancer mouse model.
Figure 6.
YY002 inhibits metastasis of pancreatic cancer in vivo. (A) PAN02-Luciferase cells were implanted orthotopically into the pancreas tails of male C57/BL6 mice. Different concentrations of YY002 were given orally for 4 weeks. Quantification of bioluminescence in pancreatic orthotopic xenograft model(n = 7). (B) PAN02-Luciferase cells were inoculated intravenously into male C57/BL6 mice. Quantification of bioluminescence in pancreatic cancer liver metastasis mouse model (n = 8). (C) Overall survival rates of the additional independent orthotopic of pancreatic cancer. Log-rank (mantel-cox) test was used (n = 9 in Control, 10 mg/kg, 20 mg/kg groups; n = 8 in 5 mg/kg group). (D) Overall survival rates of the additional independent liver metastatic models of pancreatic cancer. Log-rank (mantel-cox) test was used (n = 8 in each group). (E) AsPC-1 cells were implanted orthotopically into the pancreas tails of mice, and then mice were treated with different concentrations of YY002 for 4 weeks, The survival time was counted continuously (n = 9 in Control, 10 mg/kg, 20 mg/kg groups; n = 8 in 5 mg/kg group). Data shown as mean ± sd. * P < 0.05, ** P < 0.01, *** P < 0.001 and **** P < 0.0001 by One-way ANOVA followed multiple comparison. (F-J). YY002 induced the death of other STAT3-dependent tumors. (F) YY002 induced various STAT3-dependent cancer cells death. The tumor cells treated with YY002 in different concentrations, the cell viabilities were determined by MTS assays (n = 2). (G, H) The STAT3 phosphorylation and the expression of STAT3 were measured by Western Blot in MDA-MB-231 and SUDHL-1 cells after treatment with YY002. (I) MDA-MB-231 cells were injected into mice which were treated with YY002 or BBI608 orally (n = 10). (J) SUDHL-1 cells were injected into mice which were treated with YY002 or BBI608 orally (n = 10).
YY002 Induced the Death of Other STAT3-Dependent Tumors in Vitro and in Vivo
Considering the critical role of STAT3 for the tumor progression, targeted STAT3 signaling represents a promising therapeutic approach for several tumors, including triple negative breast cancer (TNBC),35,36 acute myeloid leukemia (AML),37 and peripheral T-cell lymphoma (PTCL).38 We then selected six non-PDAC cell lines (MOLM16, OCI-AML3 AML cell lines, Karpas-299, SUDHL-1 PTCL cell lines; MDA-MB-231, MDA-MB-468 TNBC cell lines) representing other human tumor types that highly depended on STAT3. As results shown, YY002 treatment led to the specific death of STAT3-dependent cell lines derived from various tumor types (Figure 6F). Moreover, YY002 potently inhibited STAT3 Tyr705 and Ser727 phosphorylation in TNBC cells (MDA-MB-231) and PTCL cells (SUDHL-1) (Figure 6G,H). Importantly, our study demonstrates YY002′s potent antitumor efficacy in vivo, notably causing significant regression in STAT3-dependent tumors (Figure 6I,J).
There are several ongoing clinical stage STAT3 inhibitors currently being investigated in advanced tumor patients. For instance, Napabucasin (BBI608) is an orally available STAT3 and cancer cell stemness inhibitor,39 which has finished phase III studies in patients with advanced tumor in combination with standard chemotherapies.40 In TNBC and PTCL models, oral administration of YY002 demonstrated superior antitumor efficacy compared to BBI608. These findings underscore the immense therapeutic potential of YY002 for treating STAT3-dependent tumors (Figure 6I,J).
Discussion
STAT3 is a member of the STAT transcription factor family, which promotes tumor progression by regulating the expression of genes related to cell proliferation, survival, metastasis, and immune evasion.41 STAT3 is abnormally constitutively activated in various tumors,42−47 such as pancreatic cancer, liver cancer, gastric cancer, and acute myeloid leukemia, which contributes to the poor prognosis of these patients. Thus, STAT3 is a promising drug target for cancer treatment. However, despite over 20 years of effort, the development of STAT3 inhibitors has proven to be particularly challenging. One major challenge is the difficulty of acquiring highly selective STAT3 inhibitors that discriminate between the highly structurally homologous STAT family members. The other major challenge is that phosphorylation of STAT3 at Tyr705 along with Ser727 is required for full activation of STAT3; most reported STAT3 inhibitors antagonize STAT3-Tyr705 phosphorylation while ignoring the contribution of Ser727 phosphorylation. Herein, we present a highly potent and selective small-molecule STAT3 inhibitor, and validated its on-target efficacy using the following approaches. First, we confirmed that the compound inhibited STAT3 Tyr705 and Ser727 phosphorylation and the corresponding transcriptional activities and mitochondrial OXPHOS. Second, the compound selectively bound STAT3 over other STAT members. The binding domain was located in the SH2 domain, and binding residues and a binding model was also further clarified by computational modeling and direct binding assays. Third, we characterized the selective killing profile of this small-molecule STAT3 inhibitor by testing the anticancer activities of STAT3-negative PC3 cells and a variety of STAT3-dependent tumor cells, including PDAC cells, and liver cancer cells. YY002 activity depended on STAT3, as confirmed by our cell viability assay using stable shNC and shSTAT3 cells. Taken together, this study presented a novel small-molecule STAT3 inhibitor, which is highly selective for STAT3, and able to potently suppress STAT3 Tyr705 and Ser727 phosphorylation in vitro and in vivo.
Our study has a number of important implications. First, this study may provide a basis for the evaluation of STAT3 inhibitors in PDAC treatment. As numerous researchers reported, STAT3 is persistently activated in pancreatic cancer and is associated with poor prognosis of disease.42,48,49 Inhibition of STAT3 expression or blocking its activities by signaling pathway inhibitors suppresses tumor growth and metastasis.50,51 Conditional knockdown of STAT3 in the mouse pancreas had no significant effect on the pancreas development and function,52 demonstrating STAT3 is a favorable safety target for pancreatic cancer. Our results also further clarify that inhibition of Ser727 phosphorylation by YY002 induced mitochondrial dysfunction, leading to metabolic synthetic lethality in cancer cells. Since it induces a significant inhibition of OXPHOS, YY002 is expected to be a promising therapeutic strategy for OXPHOS-addicted tumors. In a subpopulation of brain tumors, due to the mutation or deletion of glycolysis-related genes such as ENO1,49,53 the tumors are glycolysis-deficient, thus heavily dependent on OXPHOS. Meanwhile, SWF/SNF mutations, which frequently occur in pancreatic cancer, melanoma, NSCLC, and other tumors,15,54−56 induce a targetable dependence on OXPHOS. Thus, through the development and characterization of YY002, we now provide an alternative therapeutic approach for OXPHOS-additive tumors.
Indeed, our work still has several limitations needing further exploration. For instance, the effect of YY002 on the tumor microenvironment merits further studies. Accumulating evidence suggests that STAT3 Tyr705 phosphorylation regulates the tumor microenvironment by enhancing immune evasion, cross-talk between tumor cells and cancer-associated fibroblasts (CAFs), and the remodeling of stromal cells.1,57 Also, the role of Ser727 phosphorylation in regulation of the tumor microenvironment remains poor understood. Hence, we will further explore the impact of YY002 on the tumor microenvironment, and also investigate the distinctive role of STAT3 Tyr705 or Ser727 phosphorylation in modulating the tumor microenvironment. Additionally, we have confirmed that YY002 exhibits specific binding to the STAT3 SH2 domain, identifying its potential binding amino acids. Importantly, these amino acids might play a pivotal role in STAT3′s functional modulation (Supplementary Figure S2D). Consequently, in our subsequent research, we will persist in optimizing and modifying the compound to effectively target these crucial regions. Upon YY002 binding with STAT3, a noticeable inhibition of STAT3′s dual phosphorylation was observed. We speculate whether this mechanism aligns with that of other STAT3 inhibitors. Studies have been revealed that several compounds bind to STAT3, inducing the inhibition of dual STAT3 phosphorylation58−61 Notably, some compounds can solely inhibit the STAT3 Ser727 phosphorylation.62 Furthermore, we have ruled out the potential impact of YY002 on upstream kinases, as our results indicate that YY002 does not significantly affect the activity of upstream kinases of STAT3 (Figure 2F). Therefore, our hypothesis suggests that YY002 may not directly inhibit kinase activity. Instead, after binding to STAT3, YY002 could potentially influence the interactions between STAT3 and kinases related to Tyr705 (e.g., JAK, EGFR) and Ser727 phosphorylation (e.g., ERK1, JNK1), ultimately leading to a reduction in STAT3 phosphorylation. To validate these hypotheses, additional comprehensive investigations are warranted in future studies.
In summary, we present a novel small-molecule STAT3 inhibitor YY002, which simultaneously inhibited STAT3 Tyr705 and Ser727 phosphorylation. YY002 potently inhibited pancreatic tumor growth and metastasis, and also exhibited favorable pharmacokinetics and an acceptable safety profile. Therefore, this study not only evaluates the potential value of the YY002 for the treatment of PDAC tumors, but also provides new insights to develop the novel STAT3 inhibitors targeting STAT3 Tyr705 and Ser727 phosphorylation.
Acknowledgments
We thank Dr. Stefan Siwko (Texas A&M University, USA) for revising the paper. We thank the Instruments Sharing Platform of School of Life Sciences, East China Normal University for providing technical support and assistance in data collection. We thank the staff members of the Large-scale Protein Preparation System at the National Facility for Protein Science in Shanghai (NFPS), Zhangjiang Lab, China for providing technical support and assistance in data collection and analysis. This work was supported by the grants from National Natural Science Foundation of China (82373146, 82073310 to Z. Yi; 81973160 to Y. Chen; 82202897 to H. Chen), The Science and Technology Commission of Shanghai Municipality (20JC1417900 to Z. Yi; 21S11902000 to Z. Sun; 21S11907800 to Y. Chen), Shanghai Rising-Star Program (23QB1405600 to H. Chen), ECNU Construction Fund of Innovation and Entrepreneurship Laboratory (44400-20201-532300/021 to Z. Yi).
Data Availability Statement
All the data supporting the findings of this study are available from the corresponding author on reasonable request.
Supporting Information Available
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acscentsci.3c01440.
Experimental procedures for the synthesis of YY002; additional materials and methods; and Figures S1–S7, Tables S1–S3 (PDF)
Author Contributions
# H.C., A.W.B., W.B.Z. and Y.M. contributed equally to this work. Study design: H.C., Z.F.Y., Y.H.C. and M.Y.L.; Experiments conduction: H.C, A.W.B, and Y.M; Compound design and synthesis: W.B.Z and Y.H.C; Data analysis and interpretation: H.C; Figures preparation: H.C; Manuscript draft: H.C, A.W.B, Y.M, J. N. Y, Q. S. Z., W.B.Z, Z.L.S and C.W.T; Manuscript edition and approve: Y.H.C, Z.F.Y and M.Y.L. All of the authors approve the submitted manuscript.
The authors declare no competing financial interest.
Supplementary Material
References
- Yu H.; Pardoll D.; Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat. Rev. Cancer 2009, 9 (11), 798–809. 10.1038/nrc2734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huynh J.; Chand A.; Gough D.; Ernst M. Therapeutically exploiting STAT3 activity in cancer - using tissue repair as a road map. Nat. Rev. Cancer 2019, 19 (2), 82–96. 10.1038/s41568-018-0090-8. [DOI] [PubMed] [Google Scholar]
- Wong A. L. A.; Hirpara J. L.; Pervaiz S.; Eu J. Q.; Sethi G.; Goh B. C. Do STAT3 inhibitors have potential in the future for cancer therapy?. Expert opinion on investigational drugs 2017, 26 (8), 883–887. 10.1080/13543784.2017.1351941. [DOI] [PubMed] [Google Scholar]
- Dong J.; Cheng X. D.; Zhang W. D.; Qin J. J. Recent Update on Development of Small-Molecule STAT3 Inhibitors for Cancer Therapy: From Phosphorylation Inhibition to Protein Degradation. J. Med. Chem. 2021, 64 (13), 8884–8915. 10.1021/acs.jmedchem.1c00629. [DOI] [PubMed] [Google Scholar]
- Wegrzyn J.; Potla R.; Chwae Y. J.; Sepuri N. B.; Zhang Q.; Koeck T.; Derecka M.; Szczepanek K.; Szelag M.; Gornicka A.; et al. Function of mitochondrial Stat3 in cellular respiration. Science 2009, 323 (5915), 793–797. 10.1126/science.1164551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee M.; Hirpara J. L.; Eu J. Q.; Sethi G.; Wang L.; Goh B. C.; Wong A. L. Targeting STAT3 and oxidative phosphorylation in oncogene-addicted tumors. Redox biology 2019, 25, 101073 10.1016/j.redox.2018.101073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liberti M. V.; Locasale J. W. The Warburg Effect: How Does it Benefit Cancer Cells?. Trends in biochemical sciences 2016, 41 (3), 211–218. 10.1016/j.tibs.2015.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sancho P.; Barneda D.; Heeschen C. Hallmarks of cancer stem cell metabolism. British journal of cancer 2016, 114 (12), 1305–1312. 10.1038/bjc.2016.152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schopf B.; Weissensteiner H.; Schafer G.; Fazzini F.; Charoentong P.; Naschberger A.; Rupp B.; Fendt L.; Bukur V.; Giese I.; et al. OXPHOS remodeling in high-grade prostate cancer involves mtDNA mutations and increased succinate oxidation. Nat. Commun. 2020, 11 (1), 1487. 10.1038/s41467-020-15237-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zacksenhaus E.; Shrestha M.; Liu J. C.; Vorobieva I.; Chung P. E. D.; Ju Y.; Nir U.; Jiang Z. Mitochondrial OXPHOS Induced by RB1 Deficiency in Breast Cancer: Implications for Anabolic Metabolism, Stemness, and Metastasis. Trends in cancer 2017, 3 (11), 768–779. 10.1016/j.trecan.2017.09.002. [DOI] [PubMed] [Google Scholar]
- Lagadinou E. D.; Sach A.; Callahan K.; Rossi R. M.; Neering S. J.; Minhajuddin M.; Ashton J. M.; Pei S.; Grose V.; O’Dwyer K. M.; et al. BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell stem cell 2013, 12 (3), 329–341. 10.1016/j.stem.2012.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar B. Harnessing the Metabolic Vulnerabilities of Leukemia Stem Cells to Eradicate Acute Myeloid Leukemia. Frontiers in oncology 2021, 11, 632789 10.3389/fonc.2021.632789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Masoud R.; Reyes-Castellanos G.; Lac S.; Garcia J.; Dou S.; Shintu L.; Abdel Hadi N.; Gicquel T.; El Kaoutari A.; Dieme B.; et al. Targeting Mitochondrial Complex I Overcomes Chemoresistance in High OXPHOS Pancreatic Cancer. Cell Rep. Med. 2020, 1 (8), 100143 10.1016/j.xcrm.2020.100143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Valle S.; Alcala S.; Martin-Hijano L.; Cabezas-Sainz P.; Navarro D.; Munoz E. R.; Yuste L.; Tiwary K.; Walter K.; Ruiz-Canas L.; et al. Exploiting oxidative phosphorylation to promote the stem and immunoevasive properties of pancreatic cancer stem cells. Nat. Commun. 2020, 11 (1), 5265. 10.1038/s41467-020-18954-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lissanu Deribe Y.; Sun Y.; Terranova C.; Khan F.; Martinez-Ledesma J.; Gay J.; Gao G.; Mullinax R. A.; Khor T.; Feng N.; et al. Mutations in the SWI/SNF complex induce a targetable dependence on oxidative phosphorylation in lung cancer. Nat. Med. 2018, 24 (7), 1047–1057. 10.1038/s41591-018-0019-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Molina J. R.; Sun Y.; Protopopova M.; Gera S.; Bandi M.; Bristow C.; McAfoos T.; Morlacchi P.; Ackroyd J.; Agip A. A.; et al. An inhibitor of oxidative phosphorylation exploits cancer vulnerability. Nat. Med. 2018, 24 (7), 1036–1046. 10.1038/s41591-018-0052-4. [DOI] [PubMed] [Google Scholar]
- Sica V.; Bravo-San Pedro J. M.; Stoll G.; Kroemer G. Oxidative phosphorylation as a potential therapeutic target for cancer therapy. International journal of cancer 2020, 146 (1), 10–17. 10.1002/ijc.32616. [DOI] [PubMed] [Google Scholar]
- Ashton T. M.; McKenna W. G.; Kunz-Schughart L. A.; Higgins G. S. Oxidative Phosphorylation as an Emerging Target in Cancer Therapy. Clin. Cancer Res. 2018, 24 (11), 2482–2490. 10.1158/1078-0432.CCR-17-3070. [DOI] [PubMed] [Google Scholar]
- Siegel R. L.; Miller K. D.; Fuchs H. E.; Jemal A. Cancer Statistics, 2021. CA: a cancer journal for clinicians 2021, 71 (1), 7–33. 10.3322/caac.21654. [DOI] [PubMed] [Google Scholar]
- Kleeff J.; Korc M.; Apte M.; La Vecchia C.; Johnson C. D.; Biankin A. V.; Neale R. E.; Tempero M.; Tuveson D. A.; Hruban R. H.; et al. Pancreatic cancer. Nature reviews. Disease primers 2016, 2, 16022. 10.1038/nrdp.2016.22. [DOI] [PubMed] [Google Scholar]
- Ryan D. P.; Hong T. S.; Bardeesy N. Pancreatic adenocarcinoma. New England journal of medicine 2014, 371 (11), 1039–1049. 10.1056/NEJMra1404198. [DOI] [PubMed] [Google Scholar]
- Hidalgo M. Pancreatic cancer. New England journal of medicine 2010, 362 (17), 1605–1617. 10.1056/NEJMra0901557. [DOI] [PubMed] [Google Scholar]
- Wu P.; Wu D.; Zhao L.; Huang L.; Shen G.; Huang J.; Chai Y. Prognostic role of STAT3 in solid tumors: a systematic review and meta-analysis. Oncotarget 2016, 7 (15), 19863–19883. 10.18632/oncotarget.7887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lesina M.; Kurkowski M. U.; Ludes K.; Rose-John S.; Treiber M.; Kloppel G.; Yoshimura A.; Reindl W.; Sipos B.; Akira S.; et al. Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer. Cancer cell 2011, 19 (4), 456–469. 10.1016/j.ccr.2011.03.009. [DOI] [PubMed] [Google Scholar]
- Corcoran R. B.; Contino G.; Deshpande V.; Tzatsos A.; Conrad C.; Benes C. H.; Levy D. E.; Settleman J.; Engelman J. A.; Bardeesy N. STAT3 plays a critical role in KRAS-induced pancreatic tumorigenesis. Cancer research 2011, 71 (14), 5020–5029. 10.1158/0008-5472.CAN-11-0908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li N.; Grivennikov S. I.; Karin M. The unholy trinity: inflammation, cytokines, and STAT3 shape the cancer microenvironment. Cancer cell 2011, 19 (4), 429–431. 10.1016/j.ccr.2011.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wormann S. M.; Song L.; Ai J.; Diakopoulos K. N.; Kurkowski M. U.; Gorgulu K.; Ruess D.; Campbell A.; Doglioni C.; Jodrell D.; et al. Loss of P53 Function Activates JAK2-STAT3 Signaling to Promote Pancreatic Tumor Growth, Stroma Modification, and Gemcitabine Resistance in Mice and Is Associated With Patient Survival. Gastroenterology 2016, 151 (1), 180–193 e112. 10.1053/j.gastro.2016.03.010. [DOI] [PubMed] [Google Scholar]
- Chen H.; Bian A.; Yang L. F.; Yin X.; Wang J.; Ti C.; Miao Y.; Peng S.; Xu S.; Liu M.; et al. Targeting STAT3 by a small molecule suppresses pancreatic cancer progression. Oncogene 2021, 40 (8), 1440–1457. 10.1038/s41388-020-01626-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koskela H. L.; Eldfors S.; Ellonen P.; van Adrichem A. J.; Kuusanmaki H.; Andersson E. I.; Lagstrom S.; Clemente M. J.; Olson T.; Jalkanen S. E.; et al. Somatic STAT3 mutations in large granular lymphocytic leukemia. New England journal of medicine 2012, 366 (20), 1905–1913. 10.1056/NEJMoa1114885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schockel L.; Glasauer A.; Basit F.; Bitschar K.; Truong H.; Erdmann G.; Algire C.; Hagebarth A.; Willems P. H.; Kopitz C.; et al. Targeting mitochondrial complex I using BAY 87–2243 reduces melanoma tumor growth. Cancer Metab 2015, 3, 11. 10.1186/s40170-015-0138-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robinson B. H.; Petrova-Benedict R.; Buncic J. R.; Wallace D. C. Nonviability of cells with oxidative defects in galactose medium: a screening test for affected patient fibroblasts. Biochem Med. Metab Biol. 1992, 48 (2), 122–126. 10.1016/0885-4505(92)90056-5. [DOI] [PubMed] [Google Scholar]
- Lin L.; Hutzen B.; Zuo M.; Ball S.; Deangelis S.; Foust E.; Pandit B.; Ihnat M. A.; Shenoy S. S.; Kulp S.; et al. Novel STAT3 phosphorylation inhibitors exhibit potent growth-suppressive activity in pancreatic and breast cancer cells. Cancer research 2010, 70 (6), 2445–2454. 10.1158/0008-5472.CAN-09-2468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee J. W.; Stone M. L.; Porrett P. M.; Thomas S. K.; Komar C. A.; Li J. H.; Delman D.; Graham K.; Gladney W. L.; Hua X.; et al. Hepatocytes direct the formation of a pro-metastatic niche in the liver. Nature 2019, 567 (7747), 249–252. 10.1038/s41586-019-1004-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maitra A. Molecular envoys pave the way for pancreatic cancer to invade the liver. Nature 2019, 567 (7747), 181–182. 10.1038/d41586-019-00710-z. [DOI] [PubMed] [Google Scholar]
- Qin J. J.; Yan L.; Zhang J.; Zhang W. D. STAT3 as a potential therapeutic target in triple negative breast cancer: a systematic review. J. Exp Clin Cancer Res. 2019, 38 (1), 195. 10.1186/s13046-019-1206-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang X.; Yue P.; Page B. D.; Li T.; Zhao W.; Namanja A. T.; Paladino D.; Zhao J.; Chen Y.; Gunning P. T.; et al. Orally bioavailable small-molecule inhibitor of transcription factor Stat3 regresses human breast and lung cancer xenografts. Proc. Natl. Acad. Sci. U. S. A. 2012, 109 (24), 9623–9628. 10.1073/pnas.1121606109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bai L.; Zhou H.; Xu R.; Zhao Y.; Chinnaswamy K.; McEachern D.; Chen J.; Yang C. Y.; Liu Z.; Wang M.; et al. A Potent and Selective Small-Molecule Degrader of STAT3 Achieves Complete Tumor Regression In Vivo. Cancer cell 2019, 36 (5), 498–511 e417. 10.1016/j.ccell.2019.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han J. J.; O’Byrne M.; Stenson M. J.; Maurer M. J.; Wellik L. E.; Feldman A. L.; McPhail E. D.; Witzig T. E.; Gupta M. Prognostic and therapeutic significance of phosphorylated STAT3 and protein tyrosine phosphatase-6 in peripheral-T cell lymphoma. Blood Cancer J. 2018, 8 (11), 110. 10.1038/s41408-018-0138-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y.; Rogoff H. A.; Keates S.; Gao Y.; Murikipudi S.; Mikule K.; Leggett D.; Li W.; Pardee A. B.; Li C. J. Suppression of cancer relapse and metastasis by inhibiting cancer stemness. Proc. Natl. Acad. Sci. U. S. A. 2015, 112 (6), 1839–1844. 10.1073/pnas.1424171112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sonbol M. B.; Ahn D. H.; Goldstein D.; Okusaka T.; Tabernero J.; Macarulla T.; Reni M.; Li C. P.; O’Neil B.; Van Cutsem E.; et al. CanStem111P trial: a Phase III study of napabucasin plus nab-paclitaxel with gemcitabine. Future Oncol 2019, 15 (12), 1295–1302. 10.2217/fon-2018-0903. [DOI] [PubMed] [Google Scholar]
- Yu H.; Lee H.; Herrmann A.; Buettner R.; Jove R. Revisiting STAT3 signalling in cancer: new and unexpected biological functions. Nat. Rev. Cancer 2014, 14 (11), 736–746. 10.1038/nrc3818. [DOI] [PubMed] [Google Scholar]
- Nagathihalli N. S.; Castellanos J. A.; Shi C.; Beesetty Y.; Reyzer M. L.; Caprioli R.; Chen X.; Walsh A. J.; Skala M. C.; Moses H. L.; et al. Signal Transducer and Activator of Transcription 3, Mediated Remodeling of the Tumor Microenvironment Results in Enhanced Tumor Drug Delivery in a Mouse Model of Pancreatic Cancer. Gastroenterology 2015, 149 (7), 1932–1943 e1939. 10.1053/j.gastro.2015.07.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He G.; Karin M. NF-kappaB and STAT3 - key players in liver inflammation and cancer. Cell Res. 2011, 21 (1), 159–168. 10.1038/cr.2010.183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim D. Y.; Cha S. T.; Ahn D. H.; Kang H. Y.; Kwon C. I.; Ko K. H.; Hwang S. G.; Park P. W.; Rim K. S.; Hong S. P. STAT3 expression in gastric cancer indicates a poor prognosis. J. Gastroenterol Hepatol 2009, 24 (4), 646–651. 10.1111/j.1440-1746.2008.05671.x. [DOI] [PubMed] [Google Scholar]
- Jiang Y. Z.; Ma D.; Suo C.; Shi J.; Xue M.; Hu X.; Xiao Y.; Yu K. D.; Liu Y. R.; Yu Y.; et al. Genomic and Transcriptomic Landscape of Triple-Negative Breast Cancers: Subtypes and Treatment Strategies. Cancer cell 2019, 35 (3), 428–440 e425. 10.1016/j.ccell.2019.02.001. [DOI] [PubMed] [Google Scholar]
- Shallis R. M.; Wang R.; Davidoff A.; Ma X.; Zeidan A. M. Epidemiology of acute myeloid leukemia: Recent progress and enduring challenges. Blood Rev. 2019, 36, 70–87. 10.1016/j.blre.2019.04.005. [DOI] [PubMed] [Google Scholar]
- Benekli M.; Baumann H.; Wetzler M. Targeting signal transducer and activator of transcription signaling pathway in leukemias. J. Clin Oncol 2009, 27 (26), 4422–4432. 10.1200/JCO.2008.21.3264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fukuda A.; Wang S. C.; Morris J. P. t.; Folias A. E.; Liou A.; Kim G. E.; Akira S.; Boucher K. M.; Firpo M. A.; Mulvihill S. J.; et al. Stat3 and MMP7 contribute to pancreatic ductal adenocarcinoma initiation and progression. Cancer cell 2011, 19 (4), 441–455. 10.1016/j.ccr.2011.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leonard P. G.; Satani N.; Maxwell D.; Lin Y. H.; Hammoudi N.; Peng Z.; Pisaneschi F.; Link T. M.; Lee G. R. t.; Sun D.; et al. SF2312 is a natural phosphonate inhibitor of enolase. Nat. Chem. Biol. 2016, 12 (12), 1053–1058. 10.1038/nchembio.2195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arpin C. C.; Mac S.; Jiang Y.; Cheng H.; Grimard M.; Page B. D.; Kamocka M. M.; Haftchenary S.; Su H.; Ball D. P.; et al. Applying Small Molecule Signal Transducer and Activator of Transcription-3 (STAT3) Protein Inhibitors as Pancreatic Cancer Therapeutics. Molecular cancer therapeutics 2016, 15 (5), 794–805. 10.1158/1535-7163.MCT-15-0003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi Y.; Gao W.; Lytle N. K.; Huang P.; Yuan X.; Dann A. M.; Ridinger-Saison M.; DelGiorno K. E.; Antal C. E.; Liang G.; et al. Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring. Nature 2019, 569 (7754), 131–135. 10.1038/s41586-019-1130-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee J. Y.; Hennighausen L. The transcription factor Stat3 is dispensable for pancreatic beta-cell development and function. Biochemical and biophysical research communications 2005, 334 (3), 764–768. 10.1016/j.bbrc.2005.06.162. [DOI] [PubMed] [Google Scholar]
- Muller F. L.; Colla S.; Aquilanti E.; Manzo V. E.; Genovese G.; Lee J.; Eisenson D.; Narurkar R.; Deng P.; Nezi L.; et al. Passenger deletions generate therapeutic vulnerabilities in cancer. Nature 2012, 488 (7411), 337–342. 10.1038/nature11331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shain A. H.; Giacomini C. P.; Matsukuma K.; Karikari C. A.; Bashyam M. D.; Hidalgo M.; Maitra A.; Pollack J. R. Convergent structural alterations define SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeler as a central tumor suppressive complex in pancreatic cancer. Proc. Natl. Acad. Sci. U. S. A. 2012, 109 (5), E252–259. 10.1073/pnas.1114817109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ying H.; Dey P.; Yao W.; Kimmelman A. C.; Draetta G. F.; Maitra A.; DePinho R. A. Genetics and biology of pancreatic ductal adenocarcinoma. Genes & development 2016, 30 (4), 355–385. 10.1101/gad.275776.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fischer G. M.; Jalali A.; Kircher D. A.; Lee W. C.; McQuade J. L.; Haydu L. E.; Joon A. Y.; Reuben A.; de Macedo M. P.; Carapeto F. C. L.; et al. Molecular Profiling Reveals Unique Immune and Metabolic Features of Melanoma Brain Metastases. Cancer Discov 2019, 9 (5), 628–645. 10.1158/2159-8290.CD-18-1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bournazou E.; Bromberg J. Targeting the tumor microenvironment: JAK-STAT3 signaling. JAKSTAT 2013, 2 (2), e23828 10.4161/jkst.23828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schust J.; Sperl B.; Hollis A.; Mayer T. U.; Berg T. Stattic: a small-molecule inhibitor of STAT3 activation and dimerization. Chem. Biol. 2006, 13 (11), 1235–1242. 10.1016/j.chembiol.2006.09.018. [DOI] [PubMed] [Google Scholar]
- Boengler K.; Ungefug E.; Heusch G.; Schulz R. The STAT3 inhibitor stattic impairs cardiomyocyte mitochondrial function through increased reactive oxygen species formation. Curr. Pharm. Des 2013, 19 (39), 6890–6895. 10.2174/138161281939131127115940. [DOI] [PubMed] [Google Scholar]
- Lachance C.; Goupil S.; Leclerc P. Stattic V, a STAT3 inhibitor, affects human spermatozoa through regulation of mitochondrial activity. J. Cell Physiol 2013, 228 (4), 704–713. 10.1002/jcp.24215. [DOI] [PubMed] [Google Scholar]
- Cai G.; Yu W.; Song D.; Zhang W.; Guo J.; Zhu J.; Ren Y.; Kong L. Discovery of fluorescent coumarin-benzo[b]thiophene 1, 1-dioxide conjugates as mitochondria-targeting antitumor STAT3 inhibitors. Eur. J. Med. Chem. 2019, 174, 236–251. 10.1016/j.ejmech.2019.04.024. [DOI] [PubMed] [Google Scholar]
- Mackenzie G. G.; Huang L.; Alston N.; Ouyang N.; Vrankova K.; Mattheolabakis G.; Constantinides P. P.; Rigas B. Targeting mitochondrial STAT3 with the novel phospho-valproic acid (MDC-1112) inhibits pancreatic cancer growth in mice. PloS one 2013, 8 (5), e61532 10.1371/journal.pone.0061532. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
All the data supporting the findings of this study are available from the corresponding author on reasonable request.






