Metformin for Therapeutic Intervention In Acute Myeloid Leukemia

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4351-4351 ◽  
Author(s):  
Sarah Scotland ◽  
Elizabeth Micklow ◽  
Zhu Wang ◽  
Helena Boutzen ◽  
Christian Récher ◽  
...  

Abstract Abstract 4351 Metformin is a biguanide compound widely used for the treatment of type 2 diabetes. Several epidemiological studies have shown that metformin may reduce the risk of cancer in these patients and recent works in cancer suggest that this drug class may have anti-neoplastic activity. Metformin is known to have at least two mechanisms of action, which may be interrelated, inhibition of electron transport chain complex I and modulation of intracellular signal transduction pathways especially AMP Kinase. We hypothesized that acute myeloid leukemia (AML) cells may be sensitive to this agent and have studied its effects on cell survival and cellular metabolism in several AML cell lines. Metformin consistently and markedly decreased oxygen consumption of six leukemic cell lines in a concentration-dependent manner. However, only MOLM14 cells showed significant apoptosis when treated with metformin alone or in combination with a conventional chemotherapeutic agent (cytosine arabinoside). In addition, only MOLM14 cells exhibit a significant increase of the extracellular lactate level (Pasteur effect) in response to metformin-induced inhibition of the mitochondrial electron transport chain complex I. By contrast, U937 cells, another AML cell line are insensitive to metformin with a marked decrease of the Pasteur effect, suggesting that intrinsic metabolic differences may contribute to the cytotoxic effect of metformin in vitro. Interestingly, we first observed highest glucose consumption and glutathione content as well as differentially expressed genes encoding several enzymes that catalyze glycolytic and anapleurotic reactions in metformin-insensitive U937 cells compared to metformin-sensitive MOLM14 cells. Accordingly, treatment of U937 cells with an inhibitor of glycolysis sensitized U937 cells to metformin while their treatment with an inhibitor of the glutathione synthesis did not abrogate their insensitivity. Finally, treatment of insensitive HL60 cells with activators of mitochondrial oxygen consumption and cell differentiation sensitized these cells to metformin. Taken together, these findings suggest that a high glycolytic flux for production of ATP and biosynthetic precursors coupled to significant routing to the pentose phosphate pathway for NADPH for biosynthesis and GSH regeneration are key components which counterbalance the metformin-induced cytotoxic stress in U937 cells. Furthermore, based on these results, we can hypothesize that AML cell lines, and perhaps primary AML patient samples undergo a reprogramming of diverse metabolic pathways, which might be exploited by targeted therapies. Experiments on metabolic and signaling pathways as well as in vivo studies are in progress to better characterize alterations in different metabolic pathways which mediate the cytotoxic response of metformin in both AML cell lines and primary patient specimens, and thereby impact the therapeutic potential of metformin in vivo. Disclosures: Carroll: Cephalon Oncology: Consultancy; Sanofi Aventis Corporation: Research Funding; Agios Pharmaceuticals: Research Funding; Tetralogic Pharmaceuticals: Research Funding.

Cancer Cell ◽  
2019 ◽  
Vol 36 (1) ◽  
pp. 84-99.e8 ◽  
Author(s):  
Irène Baccelli ◽  
Yves Gareau ◽  
Bernhard Lehnertz ◽  
Stéphane Gingras ◽  
Jean-François Spinella ◽  
...  

2019 ◽  
Author(s):  
Irene Baccelli ◽  
Yves Gareau ◽  
Bernhard Lehnertz ◽  
Stéphane Gingras ◽  
Jean-François Spinella ◽  
...  

2019 ◽  
Author(s):  
Irène Baccelli ◽  
Yves Gareau ◽  
Bernhard Lehnertz ◽  
Stéphane Gingras ◽  
Jean-François Spinella ◽  
...  

AbstractInhibition of oxidative phosphorylation (OXPHOS) is a promising therapeutic strategy in Acute Myeloid Leukemia (AML), but patients respond heterogeneously. Through chemically interrogation of 200 sequenced specimens, we identified Mubritinib as a strong in vitro and in vivo anti-leukemic compound, acting through ubiquinone-dependent inhibition of Electron Transport Chain complex I (ETC1). ETC1 targeting showed selective toxicity against a subgroup of chemotherapy-resistant leukemias exhibiting OXPHOS hyperactivity, high expression of mitochondrial activity-related genes, and mutations affecting NPM1, FLT3 and DNMT3A. Altogether, our work thus identifies a novel ETC1 inhibitor with high clinical potential and reveals the landscape of OXPHOS dependency in AML.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1649-1649 ◽  
Author(s):  
Nobuaki Fukushima ◽  
Yosuke Minami ◽  
Fumihiko Hayakawa ◽  
Hitoshi Kiyoi ◽  
Anil Sadarangani ◽  
...  

Abstract Background Aberrant activation of the Hedgehog (Hh) signaling pathway is involved in a variety of cancers, and required for maintenance of the leukemic stem cell (LSC) populations in several experimental systems. Cumulative evidence suggests that dormant self-renewing LSC contribute to acute myeloid leukemia (AML) propagation and relapse by evading conventional chemotherapies that target cycling cells. PF-04449913 (PF) is a novel oral small molecule inhibitor that selectively binds and targets Smoothened (SMO), a membrane protein regulating the Hh pathway. Treatment with PF has shown promising Results regarding safety, tolerability, and early signs of efficacy in a phase 1 dose-escalation study of hematologic malignancies including AML (Jamieson C, et al. ASH, 2011). On the basis of these encouraging Results, phase 1b and phase 2 studies of PF in combination with chemotherapies have been planned in patients with AML. However, the detailed mechanisms and biomarkers remain to be elucidated in AML therapy with Hh pathway inhibitors. Research Aims and Methods We used AML cell lines and patient-derived primary AML cells in order to evaluate the efficacy and elucidate detailed mechanisms and biomarkers in the Hh antagonist, PF treatment. Using the co-culturing system with HS-5 stromal cells, the colony assay system, and the immunodeficient NOD/SCID/IL2rgnull (NOG) mouse model serially xenotransplanted with primary AML cells, we examined the effects of PF on LSC population and AML propagation. Results Using FACS sorting and RQ-PCR assays of AML patient-derived primary cells, the Hh signaling pathway was activated more in CD34-positive cells than CD34-negative cells. Ex vivo-treatment with PF inhibited proliferation and induced minimal cell death in leukemia cell lines and primary AML cells. However, in vivo-treatment with PF attenuated leukemia-initiation potential in AML cells through the serial transplantation system, while limiting reduction of tumor burden in the primary leukemia system. Also in the colony-assay system using primary AML cells, treatment with PF reduced serially colony formation. In MOLM-14 cells, treatment with PF down-regulated mRNA encoding downstream effector GLIs and GLI-targeting molecules in the canonical Hh pathway using RQ-PCR assays, and decreased nuclear expression of GLI-2 using immunofluorescence assays. In addition, treatment with PF remarkably decreased the quiescent (Hoechst-33342low/Pyronin-Ylow) cell population and increased cycling cell population. In the in vivo-NOG mouse system, comprehensive Gene Set Enrichment Analysis (GSEA) revealed that PF treatment modulated cell cycle regulation and self-renewal signaling in primary AML cells. Moreover, combined treatment with PF abrogated resistance to Ara-C in AML cell lines co-cultured with HS-5 stromal cells and sensitized primary AML cells to Ara-C in the colony-assay system. We are also investigating toxicity for normal cord blood cells with PF treatment. Conclusions Our findings imply that selective Hh inhibitor, PF treatment can attenuate the leukemia-initiation potential in AML cells by modulation of cell cycle regulation and self-renewal signaling, and can also improve AML therapy through sensitizing dormant LSC to chemotherapy and overcoming the resistance in the bone marrow microenvironment. Disclosures: Kiyoi: Kyowa Hakko Kirin Co. Ltd.: Research Funding; Novartis Pharma: Research Funding; Chugai Pharmaceutical Co., Ltd.: Research Funding; Bristol-Myers Squibb: Research Funding.


2011 ◽  
Vol 22 (13) ◽  
pp. 2235-2245 ◽  
Author(s):  
Zhongyan Zhang ◽  
Nobunao Wakabayashi ◽  
Junko Wakabayashi ◽  
Yasushi Tamura ◽  
Woo-Jin Song ◽  
...  

Previous studies using in vitro cell culture systems have shown the role of the dynamin-related GTPase Opa1 in apoptosis prevention and mitochondrial DNA (mtDNA) maintenance. However, it remains to be tested whether these functions of Opa1 are physiologically important in vivo in mammals. Here, using the Cre-loxP system, we deleted mouse Opa1 in pancreatic beta cells, in which glucose-stimulated ATP production in mitochondria plays a key role in insulin secretion. Beta cells lacking Opa1 maintained normal copy numbers of mtDNA; however, the amount and activity of electron transport chain complex IV were significantly decreased, leading to impaired glucose-stimulated ATP production and insulin secretion. In addition, in Opa1-null beta cells, cell proliferation was impaired, whereas apoptosis was not promoted. Consequently, mice lacking Opa1 in beta cells develop hyperglycemia. The data suggest that the function of Opa1 in the maintenance of the electron transport chain is physiologically relevant in beta cells.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 791-791
Author(s):  
Ly P Vu ◽  
Brian F Pickering ◽  
Yuanming Cheng ◽  
Zaccara Sara ◽  
Diu Nguyen ◽  
...  

Abstract N 6-methyladenosine (m6A) is a nucleotide modification in mRNA that is required for the acquisition of cell fate in embryonic stem cells. Recent studies have indicated that methylation writers can act as both oncogenes and tumor suppressor genes. Here we show that m6A is a critical regulator of myeloid differentiation of human hematopoietic stem and progenitors and myeloid leukemia cells. Depletion of the m6A-forming enzyme METTL3 with shRNAs in human cord blood derived CD34+ hematopoietic stem/progenitor cells (HSPCs) decreased global m6A levels, promoted differentiation, and reduced cell proliferation and colony formation. Conversely, overexpression of wild type METTL3-WT, but not the catalytic dead form METTL3-CD, in HSPCs increased global m6A levels, inhibited differentiation, and promoted cell growth. We surveyed METTL3 expression across different cancers and found that METTL3 is expressed at the highest level in acute myeloid leukemia (AML) compared to a broad range of solid tumors. METTL3 was highly expressed in wide variety of human AML cell lines (11/11) and in primary AML patients (3/3). Utilizing 2D-Thin layer chromatography (TLC), we found that m6A levels in poly(A) mRNAs were significantly increased in the MOLM13 leukemia cells and primary AML patient cells compared to normal HSPCs. To directly address the role of m6A in human myeloid leukemia cells, we demonstrated that METTL3 depletion with shRNAs resulted in reduced cell proliferation and the induction of apoptosis in human AML cell lines (MOLM13, NOMO-1 and KASUMI-1), and delayed leukemia in vivo (MOLM13 cells - median survival of 23 days control versus 35 and 36 days shRNA#9 and shRNA#12 respectively - in vivo knockdown was negatively selected as determined by immunoblotting). Validating an on-target effect, expression of shRNA-resistant form of METTL3 rescued both the apoptotic and differentiation phenotypes in METTL3-depleted cells and CRISPR/Cas9 mediate depletion of METTL3 (using two independent guide RNAs) resulted in similar phenotypes. Additionally, we looked into the rank of m6A "writers" and "erasers" in the genome-wide CRISPR-based screen (Wang et al. Cell 2017) for genes essential for survival in 14 AML cell lines. Interestingly, while all members of the "writer" complexes, METTL3, METTL14, WTAP and KIAA1429, were scored highly (at the top 10%), the eraser ALKBH5 showed no essentiality and FTO is only important for survival of EOL-1 cells. This data suggests that the m6A writer complex is required for leukemia. To gain insights into the mechanism of why m6A and METTL3 are required in leukemia, we performed single-nucleotide resolution mapping of m6A in MOLM13 cells using miCLIP, gene expression analysis, ribosome profiling and reverse phase protein arrays in MOLM13 cells depleted for METTL3. We profiled mRNAs targeted for m6A modifications and found that transcripts with m6A were more stable but were translated with less efficiency. Gene Set Enrichment Analysis revealed negative enrichment of MYC and ESCs gene sets. We further validated c-MYC, BCL2 and PTEN as targets for m6A modifications. We performed meRIP-qPCR and showed that knockdown of METTL3 specifically reduced enrichment of m6A at mapped sites of these transcripts. We demonstrated that c-MYC, BCL2 and PTEN protein expression were reduced despite a 2-5 log2 fold increase in mRNA expression after METTL3 depletion. The effects were reversed in cells overexpressing METTL3-WT, but not the METTL3-CD. Importantly, we observed a robust activation of p-AKT upon METTL3 knockdown, and treatment with inhibitors of PI3K and AKT partially rescued the differentiation effects in METTL3 depleted cells. Overall, m6A is critical for maintaining the differentiation program in the hematopoietic system and that this process is dysregulated in myeloid leukemia. Our data provides a rationale for targeting the mRNA methylation program in myeloid leukemia. Disclosures Carroll: Astellas Pharmaceuticals: Research Funding; Incyte Pharmaceuticals: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document