Abstract 2463: Enhancing the antileukemic activity of venetoclax against leukemia stem cells by targeting oxidative phosphorylation through dual inhibition of PI3K and HDAC

Author(s):  
Katie Hege ◽  
Hasini Kalpage ◽  
Maik Hüttemann ◽  
Holly Pitman ◽  
Jeffrey W. Taub ◽  
...  
Leukemia ◽  
2017 ◽  
Vol 32 (4) ◽  
pp. 855-864 ◽  
Author(s):  
S Fujita ◽  
D Honma ◽  
N Adachi ◽  
K Araki ◽  
E Takamatsu ◽  
...  

2017 ◽  
Vol 23 (10) ◽  
pp. 1234-1240 ◽  
Author(s):  
Elodie M Kuntz ◽  
Pablo Baquero ◽  
Alison M Michie ◽  
Karen Dunn ◽  
Saverio Tardito ◽  
...  

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5257-5257
Author(s):  
Wenbin Zhong ◽  
Vesa Olkkonen ◽  
Xu Bing ◽  
Biying Zhu ◽  
Guoping Li ◽  
...  

Abstract Acute myelogenous leukemia (AML) is one of the deadliest hematological malignancies and there is at present no efficient strategy to defeat it. New detailed insight into AML leukemia stem cells (LSCs) survival will facilitate the identification of targets for the development of new therapeutic approaches. Recent work has provided evidence that LSCs are defective in their ability to employ glycolysis, but are highly reliant on oxidative phosphorylation, and the maintenance of mitochondrial function is essential for LSCs survival. It is increasingly clear that Ca2+ released constitutively from endoplasmic reticulum (ER) is taken up by mitochondria to sustain optimal bioenergetics and cell survival. Here we report three striking findings: 1) oxysterol-binding protein (OSBP)-related protein 4 (ORP4L) is expressed in LSCs but not in normal hematopoietic stem cells (HSCs). 2) ORP4L is essential for LSC bioenergetics; It forms a complex with PLCβ3 and IP3 receptor 1 (ITPR1) to control Ca2+ release from the ER and subsequent cytosolic and mitochondrial parallel Ca2+ spike oscillations that sustain pyruvate dehydrogenase (PDH) activation and oxidative phosphorylation. 3) ORP4L inhibition eradicates LSCs in vitro and in vivo through impairment of Ca2+-dependent bioenergetics. These results suggest a novel role of ORP4L in governing Ca2+ release to sustain mitochondrial function and survival of LSCs and identify ORP4L as a putative new oncoprotein and therapeutic target for LSCs elimination. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (4) ◽  
pp. 389-394 ◽  
Author(s):  
Courtney L. Jones ◽  
Brett M. Stevens ◽  
Angelo D’Alessandro ◽  
Rachel Culp-Hill ◽  
Julie A. Reisz ◽  
...  

Abstract We have previously demonstrated that oxidative phosphorylation is required for the survival of human leukemia stem cells (LSCs) from patients with acute myeloid leukemia (AML). More recently, we demonstrated that LSCs in patients with de novo AML rely on amino acid metabolism to drive oxidative phosphorylation. Notably, although overall levels of amino acids contribute to LSC energy metabolism, our current findings suggest that cysteine may be of particular importance for LSC survival. We demonstrate that exogenous cysteine is metabolized exclusively to glutathione. Upon cysteine depletion, glutathione synthesis is impaired, leading to reduced glutathionylation of succinate dehydrogenase A (SDHA), a key component of electron transport chain complex (ETC) II. Loss of SDHA glutathionylation impairs ETC II activity, thereby inhibiting oxidative phosphorylation, reducing production of ATP, and leading to LSC death. Given the role of cysteine in driving LSC energy production, we tested cysteine depletion as a potential therapeutic strategy. Using a novel cysteine-degrading enzyme, we demonstrate selective eradication of LSCs, with no detectable effect on normal hematopoietic stem/progenitor cells. Together, these findings indicate that LSCs are aberrantly reliant on cysteine to sustain energy metabolism, and that targeting this axis may represent a useful therapeutic strategy.


2012 ◽  
Vol 53 (12) ◽  
pp. 2474-2478 ◽  
Author(s):  
Krzysztof Flis ◽  
David Irvine ◽  
Mhairi Copland ◽  
Ravi Bhatia ◽  
Tomasz Skorski

2013 ◽  
Vol 12 (3) ◽  
pp. 329-341 ◽  
Author(s):  
Eleni D. Lagadinou ◽  
Alexander Sach ◽  
Kevin Callahan ◽  
Randall M. Rossi ◽  
Sarah J. Neering ◽  
...  

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 932-932
Author(s):  
Ajay Abraham ◽  
Shaowei Qiu ◽  
Balu K Chacko ◽  
Hui Li ◽  
Andrew J Paterson ◽  
...  

Abstract Despite the success of BCR-ABL tyrosine kinase inhibitors (TKIs) in inducing remission and prolonging survival of CML patients, cures remain elusive because of persistence of primitive leukemia stem cells (LSC). We have reported that SIRT1, a NAD+ dependent deacetylase, is overexpressed in CML LSC and contributes to their maintenance and TKI resistance, related at least in part to increased p53 acetylation and transcriptional activity (Li et.al; Cancer Cell 2012). However, these studies using RNAi and pharmacological inhibitors were limited by possible off-target effects and limited duration of exposure. Therefore, to definitively delineate the role of SIRT1 in CML development, we developed a conditional SIRT1 knockout mouse model by crossing SIRT1 floxed mice (SIRT1fl/fl) with the Mx1-Cre strain to delete SIRT1 in hematopoietic stem cells (HSC), and crossed with transgenic BCR-ABL mice (BA Mx1-Cre SIRT1fl/fl). SIRT1 deletion in normal HSC did not inhibit normal hematopoiesis. In contrast, SIRT1 deletion in LSC profoundly inhibited the development of CML, and SIRT1-deleted CML mice did not develop evidence of morbidity and demonstrated significantly lower leukocytosis, neutrophilia and splenomegaly, and markedly improved survival, compared to controls (Fig-1). CML development in the SCL-tTA/BCR-ABL mouse model is associated with reduction in BM stem cells, and increase in stem/progenitor cell populations in the spleen. Splenic stem and progenitor populations were significantly decreased in SIRT1-deleted mice, whereas BM LTHSC were increased, indicating reversal of alterations associated with CML. SIRT1 deletion was associated with a significant decrease in LTHSC in G0 phase and increase in LTHSC in G1 phase of cell cycle. SIRT1-deleted LTHSCs generated significantly lower WBC counts, donor cell and donor LTHSC engraftment following transplantation into wild-type recipients compared to controls, indicating reduced in vivo repopulating ability. Treatment with the BCR-ABL TKI Nilotinib (50mg/kg/day) or vehicle for 4 weeks resulted in additional suppression of CML hematopoiesis in SIRT1-deleted mice. Gene expression analysis identified significant inhibition of mitochondria and oxidative phosphorylation related genes in SIRT1-deleted CML stem cells compared to controls. We performed extracellular flux analysis of oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) using the Seahorse XF analyzer to evaluate mitochondrial respiration and glycolysis in c-Kit+ stem/progenitor cells from SIRT1-deleted and control CML and normal mice. CML LSC demonstrated significantly increased ATP-linked respiration, maximal mitochondrial respiration and mitochondrial reserve capacity compared to normal HSC. SIRT1-deletion resulted in significant reduction in basal and maximal mitochondrial respiration, and in mitochondrial reserve capacity, in CML LSC (Fig-2). In contrast, SIRT1-deletion did not result in significant change in mitochondrial respiration in normal HSC, suggesting that effects of SIRT1 deletion on mitochondrial respiration were specific to CML cells. The SIRT1 substrate PGC-1α is a master regulator of mitochondrial biogenesis and activity. Treatment of CML mice with the PGC-1α inhibitor SR-18292 significantly reduced basal, ATP-linked and maximal mitochondrial respiration. Consistent with results obtained with murine LSC, SIRT1 inhibition with TV39OH resulted in significant reduction in mitochondrial respiration in CML CD34+ cells compared to vehicle controls. Treatment of CML mice with Nilotinib did not significantly alter mitochondrial respiration in LSC. Similarly TKI treatment did not affect mitochondrial respiration in CML CD34+ cells compared to vehicle controls. In conclusion, our studies definitively demonstrate that SIRT1 is required for leukemia development, and reveal its critical role in increased mitochondrial respiration in CML LSC. Increased mitochondrial metabolism is kinase-independent, and is mediated by PGC-1a. SIRT1 inhibition results in enhanced inhibition of TKI-treated LSC. Our studies demonstrate an essential role for SIRT1 in altered mitochondrial metabolism in CML LSC, and support further exploration of SIRT1 pathway inhibition for LSC targeting. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 206-206 ◽  
Author(s):  
Eleni D Lagadinou ◽  
Alexander Sach ◽  
Kevin P Callahan ◽  
Randall M. Rossi ◽  
Sarah Neering ◽  
...  

Abstract Abstract 206 Targeting leukemia stem cells (LSCs) is a priority for the development of improved therapeutic regimens. However, the intrinsic heterogeneity of malignant populations in acute myelogenous leukemia (AML) has made it challenging to identify biological properties appropriately conserved amongst primitive cell types. To better characterize physiological features of LSCs related to growth and survival, we previously investigated oxidative state and demonstrated that the majority of functionally-defined LSCs are characterized by relatively low levels of reactive oxygen species (termed “ROS-low”)(Lagadinou et al, abstract 639 ASH 2011). Based on these findings herein we have used primary AML specimens and flow cytometric sorting for endogenous ROS levels so as to enrich for ROS-low LSCs, and we have characterized mechanisms controlling LSC energy production and redox state. We report here that LSC-enriched ROS-low cells are metabolically dormant tumor populations characterized by low levels of oxygen–dependent mitochondrial respiration (OXPHOS), low rates of anaerobic glycolysis, and a low overall cellular ATP content. These properties are unique for LSCs, as bulk leukemic cells and non-tumorigenic ROS-high cells were found to be significantly more metabolically active with regard to both aerobic and anaerobic types of energy production. Intriguingly, we further demonstrate that in contrast to bulk leukemic cells, ROS-low subsets are deficient in their ability to utilize glycolysis when mitochondrial respiration is pharmacologically blocked, indicating a paradoxical dependence of LSCs on mitochondrial energy production. To investigate the mechanisms that underlie the distinct metabolic properties of ROS-low cells, we performed gene expression studies using RNA-seq based methods. In agreement with an important role of mitochondrial metabolism in LSCs we found several mitochondrial-related genes up-regulated in ROS-low cells. Importantly, we found that ROS-low cells express significantly higher levels of bcl-2 both at the mRNA and protein level. To determine if bcl-2 up-regulation relates to the metabolic status of ROS-low cells, we evaluated the bio-energetic profile of bulk AML cells and isolated ROS-low subsets +/− in vitro treatment with the bcl-2 pharmacologic inhibitor ABT-263 and the closely related compound ABT-737. We found that functional inhibition of bcl-2 by this class of drugs results in severe OXPHOS blockage both in total AML cells and ROS-low subsets, indicating a novel non-canonical activity of bcl-2 in promoting AML cell mitochondrial bioenergetics. In unfractionated total AML cells, the bcl-2-inhibitor initiated impairment of OXPHOS was associated with a robust induction of glycolysis and variable toxicity, indicating glycolysis as a compensatory protective response of leukemic cells in this class of drugs. In contrast, bcl-2 inhibition and OXPHOS impairment in ROS-low cells was not compensated by glycolysis, and resulted in depletion of cellular ATP levels, elimination of cellular glutathione pool, oxidation and profound toxicity in the LSC-enriched ROS-low compartment in vitro. Taken together, these studies indicated ABT-263 as an approach to eradicate LSCs by impairing fundamental aspects of LSC metabolism. To more directly investigate this issue we performed xenograft analyses. We first treated ROS-low AML populations in vitro with ABT-263 concentrations equal to the IC50 concentration of the total AML cells for each sample, and then transplanted treated vs. vehicle control cells into immune deficient NSG mice. We found that ABT-263 reduced LSC potential in all AML specimens evaluated by this functional assay. Next, we treated mice engrafted with primary human AML cells with ABT-737 in vivo (50mg/kg IP for 15d), and then performed serial transplantation analyses with the engrafted cells from treated and control mice. These functional studies showed that ABT-737 clearly reduced leukemia burden in the treated primary recipients, and also significantly reduced the capacity of engrafted leukemia cells to establish AML in secondary recipients. Based on these findings, our studies propose a model wherein the unique physiology of ROS-low LSCs provides an opportunity for selective targeting via disruption of Bcl-2-dependent oxidative phosphorylation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Author(s):  
Maria Amaya ◽  
Anagha Inguva ◽  
Shanshan Pei ◽  
Courtney L Jones ◽  
Anna Krug ◽  
...  

AML is characterized by the presence of leukemia stem cells (LSCs), and failure to fully eradicate this population contributes to disease persistence/relapse. Prior studies have characterized metabolic vulnerabilities of LSCs, which demonstrate preferential reliance on oxidative phosphorylation (OXPHOS) for energy metabolism and survival. In the present study, using both genetic and pharmacologic strategies in primary human AML specimens, we show that signal transducer and activator of transcription 3 (STAT3) mediates OXPHOS in LSCs. STAT3 regulates AML-specific expression of MYC, which in turn controls transcription of the neutral amino acid transporter SLC1A5. We show that genetic inhibition of MYC or SLC1A5 acts to phenocopy the impairment of OXPHOS observed with STAT3 inhibition, thereby establishing this axis as a regulatory mechanism linking STAT3 to energy metabolism. Inhibition of SLC1A5 reduces intracellular levels of glutamine, glutathione and multiple TCA metabolites, leading to reduced TCA cycle activity and inhibition of OXPHOS. Based on these findings, we used a novel small molecule STAT3 inhibitor, that binds STAT3 and disrupts STAT3-DNA, to evaluate the biological role of STAT3. We show that STAT3 inhibition selectively leads to cell death in AML stem and progenitor cells derived from newly diagnosed and relapsed patients, while sparing normal hematopoietic cells. Together, these findings establish a STAT3-mediated mechanism that controls energy metabolism and survival in primitive AML cells.


Sign in / Sign up

Export Citation Format

Share Document