scholarly journals Effective Novel Fto Inhibitors Show Potent Anti-Cancer Efficacy and Suppress Drug Resistance

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 233-233 ◽  
Author(s):  
Rui Su ◽  
Lei Dong ◽  
Yangchan Li ◽  
Li Han ◽  
Min Gao ◽  
...  

N 6-methyladenosine (m6A) represents the most abundant and prevalent internal modification in eukaryotic mRNA. Fat mass and obesity-associated protein (FTO) was identified as the first RNA demethylase that can remove m6A from RNA. Recently, we have reported that FTO is highly expressed in acute myeloid leukemia (AML) patients, and plays a critical role in leukemogenesis as an m6A demethylase (Li Z et al., Cancer Cell, 2017). Subsequently, we showed that FTO is a target of R-2-hydroxyglutarate (R-2HG; a metabolite produced IDH1/2 mutants), and by suppression of FTO activity, R-2HG displays intrinsic and broad ant-leukemia effects (Su R et al., Cell, 2018). Thus, our data suggest that FTO is a promising and druggable target in leukemia. To discover highly effective and selective inhibitors targeting FTO protein for AML therapy, we conducted a structure based virtual screening from NCI DTP compound library with more than 260,000 compounds (Fig. 1A). The top 370 candidates were requested from NCI and tested for their anti-leukemic efficacy in MONOMAC6 AML cell line (harboring t(9;11)/MLL-AF9) via MTT cell proliferation/viability assay. Then, the top 20 compounds with prominent inhibitory effects on AML cell viability (Fig. 1B) were chosen for further validations. First, we validated their anti-leukemia effects in two additional AML cell lines NOMO-1 (carrying MLL-AF9) and U937 (non-MLL-rearranged). Second, we assessed their potential suppressive efficacy on the demethylase activity of FTO protein in cell-free system. Finally, we found two compounds, namely CS1 and CS2, displayed consistently potent anti-leukemic effects in all the AML cell lines we tested, and also showed the most significant effects on inhibition of FTO's m6A demethylase activity (Fig. 1C). Besides our CS1 and CS2, FB23-2 (Huang Y et al., Cancer Cell, 2019) and MO-I-500 (Zheng G et al., ACS Chem Neurosci, 2014) were also reported as FTO inhibitors. Thus, we compared the anti-leukemic activity of the four FTO inhibitors together in AML cells. CS1 and CS2 displayed much higher activity in inhibiting cell viability, with IC50 values 10 to 30 times lower than FB23-2 or MO-I-500 (Fig. 1D). Through Drug Affinity Responsive Target Stability (DARTS, Fig. 1E) and Cellular Thermal Shift Assay (CETSA, Fig. 1F), we demonstrated the direct interactions between CS1/CS2 and FTO protein in cellulo. CS1 and CS2 treatment induced global increased m6A abundance at mRNA levels in various AML cell lines. Via RNA sequencing, we found that CS1 and CS2 exert their anti-leukemic effects through manipulating FTO-associated signaling pathways, such as MYC pathways. Moreover, gene-specific Cross-linking immunoprecipitation (CLIP)-qPCR results also demonstrated CS1 and CS2 treatment significantly inhibited the binding of FTO to its target mRNAs, such as MYC, CEBPA, and RARA. To assess the therapeutic effects of FTO inhibitors, CS1 (packaged in Micells) and CS2, in AML in vivo, we utilized xenograft mouse model with MA9.3ITD AML cells, patient-derived xenograft (PDX) AML mouse model, and secondary bone marrow transplantation (BMT) mouse model with primary MLL-AF9 (MA9) cells. Through bioluminescence imaging, we observed that treatment with either CS1 or CS2 remarkably inhibited leukemia progression and constantly reduced leukemia burden (Fig. 1G), and dramatically prolonged survival (P<0.0001 for either CS1 or CS2) in AML mice (Fig. 1H). In a PDX model (xeno-transplanted with a human relapsed primary AML sample), both CS1 and fCS2 showed more potent anti-AML efficacy than FB23-2, while all three significantly prolonged survival relative to the control (Ctrl) group (Ctrl vs CS1, P<0.0001; Ctrl vs CS2, P<0.0001; Ctrl vs FB23-2, P=0.0057; CS1 vs FB23-2, P<0.0001; CS2 vs FB23-2, P<0.0001) (Fig. 1I). Similarly, CS1 and CS2 significantly delayed onset of leukemia and prolonged overall survival in mice transplanted with MA9-induced murine AML cells (Ctrl vs CS1, P<0.0001; Ctrl vs CS2, P=0.0002). Our further studies discovered that our FTO inhibitors also sensitize AML cells to other therapeutic drugs, such as decitabine, tyrosine kinase inhibitor, and IDH2mut inhibitor, and overcome the corresponding drug resistance. Taken together, here we have identified two potent small-molecule FTO inhibitor compounds (i.e., CS1 and CS2), effectively and selectively targeting FTO protein and showing potent therapeutic efficacy in treating leukemia. Figure Disclosures Chen: Genovel Biotech Corp: Other: scientific founder and Chairman.

Author(s):  
Yung-Hsing Huang ◽  
Mohammad Vakili ◽  
Ommoleila Molavi ◽  
Yuen Morrissey ◽  
Chengsheng Wu ◽  
...  

STAT3 is an oncoprotein which has been shown to contribute to drug resistance in multiple myeloma (MM). Nonetheless, the clinical utility of STAT3 inhibitors in treating MM has been limited, partly related to some of their pharmacologic properties. To overcome these challenges, our group had previously packaged STAT3 inhibitors using a novel formulation of nanoparticles (NP) and found encouraging results. In this study, we aimed to further improve the pharmacologic properties of these NP by decorating them with monoclonal anti-CD38 antibodies. NP loaded with S3I-1757 (a STAT3 inhibitor), labeled as S3I-NP, were generated. S3I-NP decorated with anti-CD38 (labeled as CD38-S3I-NP) were found to have a similar nanoparticular size, drug encapsulation and loading as S3I-NP. The release of S3I-1757 at 24 hours was also similar between the two formulations. Using Cy5.5 labeling of the NP, we found that the decoration of anti-CD38 on these NP significantly increased the cellular uptake by two MM cell lines (p<0.001). Accordingly, CD38-S3I-NP showed a significantly lower inhibitory concentration at 50% (IC50) compared to S3I-NP in two IL6-stimulated MM cell lines (p<0.001). In a xenograft mouse model, CD38-S3I-NP significantly reduced the tumor size by 4 folds compared to S3I-NP on day 12 after drug administration (p=0.006). The efficacy of CD38-S3I-NP in suppressing STAT3 phosphorylation in the xenografts was confirmed by using immunocytochemistry and western blot analysis. In conclusion, our study suggests that the decoration of anti-CD38 on NP loaded with STAT3 inhibitors can further improve their therapeutic effects against MM.


Cancers ◽  
2019 ◽  
Vol 11 (2) ◽  
pp. 248 ◽  
Author(s):  
Yung-Hsing Huang ◽  
Mohammad Vakili ◽  
Ommoleila Molavi ◽  
Yuen Morrissey ◽  
Chengsheng Wu ◽  
...  

STAT3 is an oncoprotein which has been shown to contribute to drug resistance in multiple myeloma (MM). Nonetheless, the clinical utility of STAT3 inhibitors in treating MM has been limited, partly related to some of their pharmacologic properties. To overcome these challenges, our group had previously packaged STAT3 inhibitors using a novel formulation of nanoparticles (NP) and found encouraging results. In this study, we aimed to further improve the pharmacologic properties of these NP by decorating them with monoclonal anti-CD38 antibodies. NP loaded with S3I-1757 (a STAT3 inhibitor), labeled as S3I-NP, were generated. S3I-NP decorated with anti-CD38 (labeled as CD38-S3I-NP) were found to have a similar nanoparticular size, drug encapsulation, and loading as S3I-NP. The release of S3I-1757 at 24 h was also similar between the two formulations. Using Cy5.5 labeling of the NP, we found that the decoration of anti-CD38 on these NP significantly increased the cellular uptake by two MM cell lines (p < 0.001). Accordingly, CD38-S3I-NP showed a significantly lower inhibitory concentration at 50% (IC50) compared to S3I-NP in two IL6-stimulated MM cell lines (p < 0.001). In a xenograft mouse model, CD38-S3I-NP significantly reduced the tumor size by 4-fold compared to S3I-NP on day 12 after drug administration (p = 0.006). The efficacy of CD38-S3I-NP in suppressing STAT3 phosphorylation in the xenografts was confirmed by using immunocytochemistry and Western blot analysis. In conclusion, our study suggests that the decoration of anti-CD38 on NP loaded with STAT3 inhibitors can further improve their therapeutic effects against MM.


Author(s):  
Yuru Shang ◽  
Xianbin Zhang ◽  
Lili Lu ◽  
Ke Jiang ◽  
Mathias Krohn ◽  
...  

Abstract Background Recent evidence proves that intravenous human immunoglobulin G (IgG) can impair cancer cell viability. However, no study evaluated whether IgG application benefits cancer patients receiving chemotherapeutics. Methods Influence of pharmaceutical-grade human IgG on the viability of a series of patient-derived colon cancer cell lines with and without chemotherapeutic intervention was determined. Cell death was analysed flow cytometrically. In addition, the influence of oxaliplatin and IgG on the ERK1/2-signalling pathway was evaluated by western blots. Results We evaluated the effects of pharmaceutical IgG, such as PRIVIGEN® IgG and Tonglu® IgG, in combination with chemotherapeutics. We did not observe any significant effects of IgG on tumour cell viability directly; however, human IgG significantly impaired the anti-tumoral effects of oxaliplatin. Primary cancer cell lines express IgG receptors and accumulate human IgG intracellularly. Moreover, while oxaliplatin induced the activation of ERK1/2, the pharmaceutical IgG inhibited ERK1/2 activity. Conclusions The present study demonstrates that pharmaceutical IgG, such as PRIVIGEN® IgG and Tonglu® IgG, can impair the anti-carcinoma activity of oxaliplatin. These data strongly suggest that therapeutic IgG as co-medication might have harmful side effects in cancer patients. The clinical significance of these preclinical observations absolutely advises further preclinical, as well as epidemiological and clinical research.


Cancers ◽  
2021 ◽  
Vol 13 (15) ◽  
pp. 3790
Author(s):  
Gro Elise Rødland ◽  
Sissel Hauge ◽  
Grete Hasvold ◽  
Lilli T. E. Bay ◽  
Tine T. H. Raabe ◽  
...  

Inhibitors of WEE1 and ATR kinases are considered promising for cancer treatment, either as monotherapy or in combination with chemo- or radiotherapy. Here, we addressed whether simultaneous inhibition of WEE1 and ATR might be advantageous. Effects of the WEE1 inhibitor MK1775 and ATR inhibitor VE822 were investigated in U2OS osteosarcoma cells and in four lung cancer cell lines, H460, A549, H1975, and SW900, with different sensitivities to the WEE1 inhibitor. Despite the differences in cytotoxic effects, the WEE1 inhibitor reduced the inhibitory phosphorylation of CDK, leading to increased CDK activity accompanied by ATR activation in all cell lines. However, combining ATR inhibition with WEE1 inhibition could not fully compensate for cell resistance to the WEE1 inhibitor and reduced cell viability to a variable extent. The decreased cell viability upon the combined treatment correlated with a synergistic induction of DNA damage in S-phase in U2OS cells but not in the lung cancer cells. Moreover, less synergy was found between ATR and WEE1 inhibitors upon co-treatment with radiation, suggesting that single inhibitors may be preferable together with radiotherapy. Altogether, our results support that combining WEE1 and ATR inhibitors may be beneficial for cancer treatment in some cases, but also highlight that the effects vary between cancer cell lines.


2010 ◽  
Vol 24 (6) ◽  
pp. 1287-1296 ◽  
Author(s):  
Susan Holbeck ◽  
Jianjun Chang ◽  
Anne M. Best ◽  
Angie L. Bookout ◽  
David J. Mangelsdorf ◽  
...  

Abstract We profiled the expression of the 48 human nuclear receptors (NRs) by quantitative RT-PCR in 51 human cancer cell lines of the NCI60 collection derived from nine different tissues. NR mRNA expression accurately classified melanoma, colon, and renal cancers, whereas lung, breast, prostate, central nervous system, and leukemia cell lines exhibited heterogeneous receptor expression. Importantly, receptor mRNA levels faithfully predicted the growth-inhibitory qualities of receptor ligands in nonendocrine tumors. Correlation analysis using NR expression profiles and drug response information across the cell line panel uncovered a number of new potential receptor-drug interactions, suggesting that in these cases, individual receptor levels may predict response to chemotherapeutic interventions. Similarly, by cross-comparing receptor levels within our expression dataset and relating these profiles to existing microarray gene expression data, we defined interactions among receptors and between receptors and other genes that can now be mechanistically queried. This work supports the strategy of using NR expression profiling to classify various types of cancer, define NR-drug interactions and receptor-gene networks, predict cancer-drug sensitivity, and identify druggable targets that may be pharmacologically manipulated for potential therapeutic intervention.


Oncotarget ◽  
2017 ◽  
Vol 8 (30) ◽  
pp. 49944-49958 ◽  
Author(s):  
Radosław Januchowski ◽  
Karolina Sterzyńska ◽  
Piotr Zawierucha ◽  
Marcin Ruciński ◽  
Monika Świerczewska ◽  
...  

Metabolites ◽  
2020 ◽  
Vol 10 (7) ◽  
pp. 280
Author(s):  
Laila Naif Al-Harbi ◽  
Pandurangan Subash-Babu ◽  
Manal Abdulaziz Binobead ◽  
Maha Hussain Alhussain ◽  
Sahar Abdulaziz AlSedairy ◽  
...  

Controlled production of cyclin dependent kinases (CDK) and stabilization of tumor suppressor genes are the most important factors involved in preventing carcinogenesis. The present study aimed to explore the cyclin dependent apoptotic effect of nymphayol on breast cancer MCF-7 cells. In our previous study, we isolated the crystal from a chloroform extract of Nymphaea stellata flower petals and it was confirmed as nymphayol (17-(hexan-2-yl)-10,13-dimethylhexadecahydro-1H-cyclopenta[a]phenanthren-3-ol) using x-ray diffraction (XRD), Fourier transform infrared (FTIR), and mass spectroscopy (MS) methods. The cytotoxic effect of nymphayol on MCF-7 cells were analyzed using the 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. The cellular and nuclear damage was determined using propidium iodide (PI) and acridine orange/ethidium bromide (AO/ErBr) staining. Tumor suppressor and apoptosis related mRNA transcript levels were determined using real-time polymerase chain reaction (RT-PCR). Nymphayol potentially inhibits MCF-7 cell viability up to 78%, and the IC50 value was observed as 2.8 µM in 24 h and 1.4 µM in 48 h. Treatment with nymphayol significantly increased reactive oxygen species (ROS) level and the tunnel assay confirmed DNA damage. We found characteristically 76% apoptotic cells and 9% necrotic cells in PI and AO/ErBr staining after 48 h treatment with 2.8 µM of nymphayol. Gene expression analysis confirmed significantly (p ≤ 0.001) increased mRNA levels of cyclin dependent kinase inhibitor 2A (Cdkn2a), retinoblastoma protein 2 (pRb2), p53, nuclear factor erythroid 2-factor 2 (Nrf2), caspase-3, and decreased B-cell lymphoma 2 (Bcl-2), murine double minute 2 (mdm2), and proliferating cell nuclear antigen (PCNA) expression after 48 h. Nymphayol effectively inhibited breast cancer cell viability, and is associated with early expression of Cdkn2a, pRb2, and activation of p53 and caspases.


Sign in / Sign up

Export Citation Format

Share Document