Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jul 1.
Published in final edited form as: Cancer Res. 2019 Oct 1;80(1):5–16. doi: 10.1158/0008-5472.CAN-19-1169

The Role of Metabolic Plasticity in Blood and Brain Stem Cell Pathophysiology

Catherine J Libby 1, Jonathan McConathy 2, Victor Darley-Usmar 3, Anita B Hjelmeland 1
PMCID: PMC7153784  NIHMSID: NIHMS1540615  PMID: 31575548

Abstract

Our understanding of intratumoral heterogeneity in cancer continues to evolve, with current models incorporating single cell signatures to explore cell-cell interactions and differentiation state. The transition between stem and differentiation states in non-neoplastic cells requires metabolic plasticity, and this plasticity is increasingly recognized to play a central role in cancer biology. The insights from hematopoietic and neural stem cell differentiation pathways were used to identify cancer stem cells in leukemia and gliomas. Similarly, defining metabolic heterogeneity and fuel switching signals in non-neoplastic stem cells may also give important insights into the corresponding molecular mechanisms controlling metabolic plasticity in cancer. These advances are important, because metabolic adaptation to anti-cancer therapeutics is rooted in this inherent metabolic plasticity and is a therapeutic challenge to be overcome.

Introduction

The initiation and progression of cancer requires the dysregulation of normal physiologic cell signals, metabolism, and biological processes, particularly those that are involved in human development. Human developmental and metabolic programs have evolved to respond to environmental changes. Cancer cells have accelerated this adaptability to respond to changing requirements for tumor growth, metastasis, and therapeutic resistance. Cancer cells select for the best adapted metabolic and growth programs encoded by mutations allowing the fittest cells to adjust and survive in their changing microenvironments. Because biological fitness reflects energy investment in progeny, it is not surprising that metabolic plasticity is one of the key hallmarks of cancer (1).

The metabolic plasticity of cancer indicates the ability of and need for cancer cells to adapt to intrinsic and extrinsic pressures to survive. To achieve this goal, cancer cells take advantage of the complete set of existing metabolic pathways, utilizing those most beneficial in the current environmental conditions to promote and maintain their growth. This notion of metabolic adaptation in cancer is enshrined in the Warburg hypothesis, which has evolved over the last few decades of research (2). An important feature of the aerobic glycolysis characteristic of the Warburg effect is that it is not simply about producing energy; intermediates used for the synthesis of biomolecules are also generated by glucose-dependent pathways including the pentose phosphate pathway. These building blocks are required for rapid cell growth, providing a fundamental benefit for aerobic glycolysis in tumor cells. To obtain the additional glucose required to fuel these pathways, cancer cells upregulate glucose transporters (GLUTs), including the ubiquitously expressed GLUT1 and the more selectively expressed GLUT3. The metabolic differences in glucose utilization between normal and tumor tissue are exploited clinically for the detection of primary tumors and metastasis, monitoring response to therapy, and detecting recurrent neoplasms by using the glucose analog 2-deoxy-2-[18F]fluoro-D-glucose (FDG) in conjunction with positron emission tomography (PET).

A number of excellent articles describe the importance of metabolic plasticity in a broad range of cancers (3-6). From these and other studies, we now understand that cancer cells are metabolically heterogeneous and metabolic adaptation to the changing environments a cancer cell experiences through its lifetime requires an intrinsic plasticity. However, metabolic plasticity, particularly partitioning between glycolysis and mitochondrial oxidative phosphorylation and fuel selection, is not unique to tumor cells. For example, macrophage phenotypes exhibit plasticity between glycolysis and oxidative phosphorylation as they adapt to the different stages of inflammation (7,8). Similarly, T-cells adopt an aerobic glycolysis program as they become activated (9,10). During development, metabolic plasticity is critical for the regulation of cell fate, as shown by the activation of glycolysis during induced pluripotent stem cell reprogramming (11-13) which is partially characterized by teratoma formation. These data suggest that understanding mechanisms of metabolic plasticity in non-neoplastic cells may inform metabolic plasticity in cancers.

Stem cells can self-renew to regenerate themselves or differentiate into defined lineages during development and for tissue maintenance, including after injury. Hematopoietic stem cells differentiate into well-characterized hematopoietic lineages yielding distinct cell populations with unique marker profiles (including erythrocytes, granulocytes, lymphocytes, monocytes, and thrombocytes). Similarly, neural stem cells differentiate into brain lineages (neurons, astrocytes, and oligodendrocytes) with stem cell and differentiation states that can be distinguished based on distinct markers. Building on these hierarchies, cancer cells with characteristics similar to stem cells were identified, first in leukemias (14) and subsequently in solid tumors, including gliomas (15,16). These cancer stem cells, or tumor initiating cells, were identified partly on the basis of the expression of cell surface stem cell markers, which permitted segregation of subsets of tumor cells via flow cytometry. For example, leukemia stem cells were CD34hiCD38 similar to hematopoietic stem cells, and glioblastoma stem cells were CD133+ similar to neural stem cells (14-16). In comparison with non-stem (marker-negative) cancer cells, which typically constitute the majority of the cancer, cancer stem cells isolated from the same patient had enhanced ability to propagate the disease in immunocompromised mice. Cancer stem cells underwent self-renewal or differentiation into lineages bearing the same genetic mutations as in the original cancer. The functional similarities between cancer stem cells and non-neoplastic stem cells were partially attributed to the utilization of common stem cell signaling pathways that are activated/elevated in cancer stem cells when compared to non-stem cancer cells. Increasing evidence suggests similarities in metabolism and metabolic plasticity between normal and neoplastic stem cells that also contribute to common phenotypes. As the cancer stem cell hypothesis states that a subset of cancer cells exhibits similarities to non-neoplastic stem cells, it does not directly address the cell of origin for a cancer (17). However, multiple studies demonstrate that the acquisition of mutations by hematopoietic stem cells or neural stem cells leads to blood or brain tumors (14,18-23). These data indicate the importance of the perturbation of developmental programs for the initiation and maintenance of cancers. They also suggest that cancer cell metabolic plasticity is another instance of cancer cell acquisition or reactivation of existing developmental programs to promote cancer growth.

In both normal and neoplastic stem cells, it is now becoming clear that metabolic plasticity is important for self-renewal and differentiation programs, linking bioenergetics to cell proliferation and cell state (13,24-28). We highlight here similarities in metabolic reprogramming in stem cells and cancer to understand how developmental cellular bioenergetics contributes to tumor initiation and growth. We will focus on examples of pathways regulating metabolic plasticity in hematopoietic and neural stem cells and their potential relationships to cancers of the blood and brain. We will highlight two important pathways, one regulating glycolysis and one regulating fatty acid oxidation, which impact normal and neoplastic blood and brain cell fate to exemplify how stem cell metabolic plasticity may inform cancer. We additionally provide examples of therapeutic interventions at both the preclinical and clinical stage. These studies on understanding metabolic pathways in cancer and their impacts on metabolic plasticity may inform new metabolotherapies.

Glycolytic Metabolism in Non-neoplastic and Neoplastic Blood and Brain Stem Cells Regulated by Oxygen Dependent Plasticity and HIFs

Hypoxia, or low oxygen tension, is known to promote glycolysis through stabilization of hypoxia inducible transcription factor 1α and 2α (HIF1α and HIF2α), which can transcriptionally upregulate multiple members of the glycolysis pathway and facilitate increased GLUT-dependent glucose uptake (Figure 1A). While HIF1α and HIF2α have many overlapping target genes, there are unique targets including the HIF2α-dependent, stem cell transcription factor Oct4 (29). The importance of modeling hypoxia, and in vivo microenvironments in general, is still relatively underappreciated considering that the majority of experiments continue to be performed at atmospheric oxygen tensions (21% O2), estimated to be 17-20% O2 with 5% CO2 supplementation in standard cell culture incubators in buffered, high glucose media. Physiological oxygen tensions in the brain and bone marrow are approximately 3-7%, with some regions less than 1.5% O2 (30), and even short-term exposure to atmospheric oxygen had deleterious effects on hematopoietic stem cell self-renewal and engraftment (31). In solid tumors, including gliomas, some tissue portions approach anoxia. However, fluctuations in oxygen tension can quickly occur due to blood vessel formation during angiogenesis or rapid tumor growth, contributing to the requirement for metabolic plasticity as the cancer cell inhabits these changing tumor microenvironments.

Figure 1. Pathways for Fuel Switching in Normal and Neoplastic Brain and Bone Stem Cells.

Figure 1.

(A) Low oxygen tension is a physiologic environment in the brain and bone marrow impacting glycolytic metabolism with hypoxia stabilization of HIFs leading to increased expression of genes regulating glycolysis and stem cell maintenance. While tissue culture is typically performed under atmospheric oxygen (21% O2), lower oxygen levels (1.5-7% O2) are common in the normal brain and bone marrow with nearly anoxic conditions in portions of solid tumors. All enzymes and/or genes shown in green are induced by hypoxia/HIF with the impact of increasing glycolysis and decreasing oxidative phosphorylation. (B) Perturbation of FOXO3 levels in normal and neoplastic neural (and hematopoietic) stem cells demonstrates a role in the regulation of self-renewal, ROS, and glycolysis due in part to regulation of HIF stability or activity. (C) Normal hematopoietic and neural stem cell signaling indicate the importance of PPARγ transcriptional regulation of CPT1 for fatty acid oxidation. Results in leukemia and brain tumors confirm the importance of fatty acid oxidation as a mechanism for energy production, particularly when glucose levels are low.

As our understanding of the impact of physiological levels of oxygen tension has improved, a number of similarities between normal and neoplastic stem cell biology have been uncovered. Many of these findings have been recently reviewed (32-35); therefore, we provided here a summary of the commonalities between hematopoietic and neural stem cell responses to changes in oxygen tension that inform stem cell fate and metabolism. In both hematopoietic stem cells and neural stem cells, hypoxia facilitated growth and survival (36-38) via a glycolytic shift (39,40). During reprogramming, induced pluripotent stem cells also undergo a glycolytic shift that is HIF1α and HIF2α dependent (41,42). While both HIF1α and HIF2α were important for hematopoietic stem cell maintenance in hypoxia, HIF2α was also critical for hematopoietic stem cell colony forming ability in normoxia where loss of HIF1α had no effect (43).

Similar results have been reported with leukemia and glioma stem cells as culture in hypoxia promoted growth, stem cell percentages and/or marker expression, and therapeutic resistance (44,45). In glioma stem cells both HIF1α and HIF2α were important for glioma stem cell maintenance, but HIF2α was expressed at higher oxygen tensions than HIF1α and was particularly elevated in the glioma stem cell fraction (46). In leukemia, a majority of the research has focused on HIF1α as it was associated with poorer disease prognosis and maintenance of leukemia stem cells (47), whereas HIF2α was not. However, HIF2α has been shown to protect both hematopoietic and leukemia stem cells from apoptosis in response to endoplasmic reticulum stress (43). As such, an area of increasing interest for cancer therapy has been the development of HIF inhibitors (48). HIF1α inhibitors may have promise for leukemia treatment as HIF1α targeting inhibits leukemia stem cell (49), but not hematopoietic stem cell (50), self-renewal. HIF2α inhibitors have been of particular interest for the treatment of glioma (51) with two HIF2α inhibitors, PT2385 and PT2977, in clinical trial for glioma after successful preclinical development (52-54). These data suggest distinct roles for HIF1α and HIF2α that still need to be further delineated while affirming a critical role for hypoxia/HIF in the regulation of normal and neoplastic hematopoietic and neural stem cell maintenance and metabolism.

Integrating Mitochondrial Respiration, ROS, and HIF Regulation of Glycolysis: FOXO3 Importance in Non-neoplastic and Neoplastic Stem Cells of the Blood and Brain

While it is common to consider distinct shifts in metabolism (i.e. glycolysis or oxidative phosphorylation), there is considerable interplay between metabolic states and gradients of metabolic functions that regulate cell fate. As an example, we highlight roles of the Forkhead box O3 (FOXO3) transcription factor in developmental programs and metabolism in the blood and brain. FOXO3 is a transcription factor that is inhibited by growth factor mediated, AKT phosphorylation-induced translocation from the nucleus, but oxidative and nutrient stress increase FOXO3 mediated-transcription (55-57). Thus, environment mediated shifts in FOXO3 activity may explain why FOXO family members are generally considered to be tumor suppressors even though some data indicate a pro-tumorigenic role for FOXO3.

FOXO3 is a critical regulator of stem cell maintenance that is linked to metabolism (Figure 1B). Studies using cells cultured from FOXO3 knockout mice revealed roles in both hematopoietic stem cell and neural stem cell self-renewal and differentiation (58-63). Loss of FOXO3 in hematopoietic and neural stem cells decreased stem cell characteristics and resulted in lineage shifts, with gene signatures associated with changes in HIF targets (58-63). Interestingly, metabolic profiling of FOXO3 knockout cells demonstrated reduced glycolysis, glutaminolysis, and utilization of the pentose phosphate pathway along with elevations in reactive oxygen species (ROS) (64,65). Multiple studies suggest FOXO3 is critical for reducing levels of hypoxia-induced ROS and HIF-1 stabilization/activity, although FOXO3 may maintain mitochondrial metabolic function independent of regulation of redox homeostasis (66). These data provide a strong rationale for investigating the role of FOXO3 in neoplastic stem cells of the brain and blood as they link mitochondria and ROS to HIF and glycolysis.

Similar to hematopoietic stem cells, FOXO3 was critical to maintain the long-term malignant potential of leukemia stem cells (67) as nuclear FOXO3 was elevated in the leukemia stem cell population (68). However, consequences of FOXO3 manipulation in glioma stem cells have been mixed. FOXO3 targeting in gliomas decreased glucose uptake (69) and increased ROS (Figure 1B), and FOXO3 was elevated in chemotherapy (temozolomide) resistant glioblastoma cells (70) that have reduced ROS in association with changes in oxidative phosphorylation (71,72). FOXO3 was important for glioma stem cell maintenance post-irradiation (73), but other results indicated that elevated FOXO3 levels or activity increased glioma stem cell differentiation (74). These differences may be explained by evidence that FOXO3 effects are p53 dependent in gliomas (73), but the signaling pathways downstream of FOXO3 or its transcriptional targets may also mediate context dependent phenotypes that are likely to complicate treatment strategies.

The data summarized here present an intriguing case in which one protein is able to coordinate multiple responses to integrate metabolic signals and provide evidence that two cancers have co-opted this to promote growth and survival. These studies provide links between FOXO3 and both stem cell states and metabolic shifts in brain and blood cancers, while also demonstrating crosstalk between pathways that are important for regulating metabolic plasticity.

Adaptive metabolism in Hematopoietic, Neural, and Neoplastic Stem Cells

Fuel switching is an important aspect of metabolic plasticity and is also a feature of cancer stem cells. Several studies demonstrate that fatty acid oxidation is important for both stem and cancer cell growth (Figure 1C) (75). For example, neural progenitor cell oxygen consumption was maintained when cells were cultured without glucose and mitochondrial function was fatty acid dependent, with linoleic acid promoting oxidative phosphorylation (76). One pathway found to be critical for the regulation of hematopoietic stem cell self-renewal through a lipid metabolism dependent mechanism involves promyelocytic leukemia protein (PML). This protein is important for the formation of nuclear bodies that regulate cell growth, survival, and genomic stability. Loss of PML in hematopoietic and neural stem cells resulted in increased proliferation and loss of quiescence followed by exhaustion (77-80) or decreased neurogenesis (81). Effects of loss of PML in hematopoietic stem cells were associated with decreased peroxisome proliferator-activated receptor γ (PPARγ) (78), which regulates fatty acid oxidation due, in part, to transcriptional upregulation of carnitine palmitoyltransferase 1 (CPT1). CPT1 transports fatty acids into the mitochondria for fatty acid oxidation (Figure 1C) (82,83). Changes in CPT1 expression and fatty acid utilization to generate energy promote the survival of thymocytes in mice with a hematopoietic cell oxidative phosphorylation defect (84), directly linking metabolic shifts to cell survival in hematopoietic progenitor subsets. Elevation of CPT1 and fatty acid oxidation was also important for neural stem cell quiescence and cell fate as loss of CPT1A decreased neural stem cell percentages in vivo (85,86). These data suggested a link between stem cell maintenance and lipid metabolism, which was confirmed when the CPT1 and fatty acid oxidation inhibitor etomoxir was determined to decrease neural stem cells (76,86) and promote hematopoietic stem cell exhaustion (82).

Regulation of the fatty acid oxidation pathway by a PML/CPT1 cascade is also suggested to be important in cancer, particularly in the cancer stem cell fraction. Leukemia stem cells were found to reside in an adipose tissue niche where the breakdown of lipids could facilitate fatty acid oxidation (87). Indeed, addition of fatty acids increased glioma cell proliferation (88) and oxygen consumption (89). PML targeting in leukemia stem cells or glioma stem cells reduced tumor initiating capacity in animal models (77,90), and degradation of PML mediated sensitivity to several chemo- and small molecule inhibitor therapies (90-92). While conflicting data suggested that PML is lower in glioblastoma cells (93,94) and could be elevated to increase cell death (95), CPT1 was elevated (96), supporting the notion that fatty acid oxidation is important for glioma growth. Together, the data suggest that cancer stem cells can reside in a lipid-rich niche and readily utilize fatty acids as a fuel source.

The utilization of fatty acid oxidation can be suppressed when glucose is available, but some cancers downregulate pathways that suppress fatty acid oxidation. With the loss of suppression, fatty acid oxidation can be active in cancers even if glucose is available. For example, levels of prolyl hydroxylase 3 (PHD3), which reduces fatty acid oxidation and promotes fatty acid synthesis when nutrients are high, are decreased in acute myeloid leukemia and a subset of gliomas (97,98). PHD3 hydroxylates the proline residues of the rate limiting enzyme for fatty acid synthesis, acetyl-coA carboxylase 2 (ACACB or ACC2), which results in increased conversion of acetyl-CoA to malonyl-CoA. Malonyl-CoA is important for fatty acid synthesis, but can also repress the activity of CPT1, decreasing transport of fatty acids into the mitochondria (99). Loss of PHD3 increased the growth of glioma cells, including in hypoxia, and PHD3 is a hypoxia response gene (98,100) that may repress glycolysis under hypoxia (101). These data suggest the possibility that PHD3 could mediate hypoxia dependent fatty acid oxidation suppression and that both glycolysis and fatty acid oxidation can be activated in hypoxia when PDH3 is lost, further promoting metabolic plasticity and fuel switching in cancers. Furthermore, elevation of fatty acid binding proteins by hypoxia in glioma cells can increase fatty acid uptake and formation of lipid droplets that can be used for energy generation upon reoxygenation (102). These data indicate an alternative method for metabolic shifts toward fatty acid oxidation that could be targeted for patient treatments, either by direct inhibition of fatty acid uptake or inhibition of hypoxia signaling.

In addition to providing leukemic stem cells with byproducts for fatty acid oxidation, adipocytes release glutamine into the microenvironment. Glutamine is able to support cellular metabolism as a carbon source and aiding in redox homeostasis and has been reviewed as an important metabolic factor in both leukemias and gliomas previously (103-106). Glutamine protects leukemia cells from L-asparaginase, a first-line therapy for acute lymphoblastic leukemia (107). However, leukemia cell lines exhibit varying responses to glutamine withdraw conditions, indicating heterogeneity in metabolic programs and suggesting that glutaminase inhibition may be a viable therapeutic option for a subset of leukemias (108,109). Glutamine also plays an important role in cell fate determination for hematopoietic stem cells. Erythropoiesis is reliant on glutamine with differentiating cells consuming more glutamine and showing decreased levels of glycolysis (110); however, when glutamine metabolism is inhibited, the cells instead develop toward a myelocytic lineage (111,112). In neural stem cells, glutamine is able to enhance survival following perinatal hypoxia in the subventricular zone (113), and multiple studies indicate elevated glutamine utilization in gliomas with the 18F-labeled amino acids [18F]fluciclovine (FDA approved for detection and localization of biochemically recurrent prostate cancer) and (2S,4R)-4-[18F]fluoroglutamine (4-FGln) being tested as PET imaging agents for glioma detection and characterization (114). Together, the data strongly indicate that similarities in metabolic developmental programs and cell signaling, particularly with regards to fuel switching required for transitioning cell state, can be used to understand the underlying mechanisms of cancer cell pathways and identify potential vulnerabilities for therapeutic targeting.

Cancer cell metabolism as a diagnostic tool and therapeutic target

Non-invasive monitoring of cancer progression and therapeutic response is a major benefit of cancer imaging. Many current cancer imaging techniques, as well as those under development, were based on determinations of fundamental differences in metabolism between cancer and non-cancer cells and/or solid tumors. For example, the glycolytic shift and increased uptake of glucose in gliomas and other solid tumors provided the basis for the use of the glucose analog FDG for PET imaging. However, the relatively high consumption of glucose by the normal brain as well as the elevated levels of FDG in regions of inflammation are substantial limitations of this approach (115). Studies suggest that aerobic glycolysis can be estimated in gliomas using [18F]FDG in combination with [15O]oxygen, but larger studies are needed to establish the utility and clinical significance of this approach (116-118).

The leading PET imaging agents for glioma target system L amino acid transport, allowing them to cross the blood-brain barrier and accumulate preferentially in glioma cells due to upregulation of specific transporter proteins (particularly LAT1/SLCA5) (115,119,120). This class of PET tracers, including L-[11C]methionine (MET), 3,4-dihydroxy-6-[18F]fluoro-L-phenylalanine (FDOPA), and O-(2-[18F]fluoro)-L-tyrosine (FET), is becoming standard of care in some countries and is entering international society guidelines for glioma imaging (121,122). The PET tracer [18F]FDOPA is currently under review by the Food and Drug Administration (FDA) in the United States (US) for congenital hyperinsulinism, which may make this agent more widely available for other applications including neuro-oncology in the US.

Other PET tracers have been investigated for glioma imaging include amino acids targeting glutamine transport and metabolism such as 4-[18F]FGln and [18F]fluciclovine as mentioned above (114,123-127). At the time of publication of this manuscript, [18F]fluciclovine has been submitted for approval to the FDA for a new indication in glioma. Other tracers that have been investigated in glioma include the thymidine analogue 2’-deoxy-2’-[18F]fluorothymidine (FLT) for proliferation imaging, 1-[18F]fluoranyl-3-(2-nitroimidazol-1-yl)propan-2-ol (FMISO) for hypoxia imaging (128-132), [11C]choline targeting membrane synthesis (133-135), and [11C]acetate targeting oxidative phosphorylation and fatty acid synthesis (136,137). There has been less application of metabolic PET tracers for leukemias, although [18F]FDG and [18F]FLT are the PET tracers that have been most studied in leukemias. [18F]FDG can play a role in detecting extramedullary sites of leukemia (138-140), and recent studies suggest that [18F]FLT-PET can serve as an early indicator of response to therapy in acute myelogenous leukemia (141,142).

Another approach for measuring tumor metabolism in both clinical and research settings uses the inherent nuclear spin properties of endogenous stable nuclides including protons, carbon atoms, and phosphorous atoms using single-voxel magnetic resonance (MR) spectroscopy (MRS) or multi-voxel magnetic resonance spectroscopic imaging (MRSI) (143). The inherent low signal in MR can be overcome using hyperpolarization techniques to generate short-lived small molecules with orders of magnitude greater signal than conventional MR. Hyperpolarized MRS using 13C has been utilized to evaluate the conversion of pyruvate to lactate as well as the production of bicarbonate to determine glioma metabolism and growth (144,145). Intriguingly, MRS can also be used to detect 2-hydroxyglutarate, the novel metabolite resulting from isocitrate dehydrogenase mutations, which are found in gliomas and leukemias (143). These data demonstrate that cancer imaging could provide information about tumor genetics without biopsy. Monitoring immediate changes in metabolism post-treatment may also be used to rapidly determine therapy responders and non-responders, permitting personalized medicine approaches in real time.

Beyond tumor imaging, understanding the metabolic alterations of cancer could provide opportunities for therapeutic intervention. Direct inhibition of glucose uptake and utilization is one commonly pursued approach, with a number of groups developing inhibitors to components of the glycolysis pathway, including via targeting GLUTs. Some GLUT inhibitors have shown promise in vitro for targeting a wide range of solid tumors, including gliomas, with limited toxicities observed in vitro (146-149). However, GLUTs are widely expressed with GLUT1 at the blood brain barrier and GLUT3 important for neurons, complicating treatment options for brain tumors. Nevertheless, there is hope that in combining GLUT inhibitors and the ketogenic diet, another way to decrease glucose availability to the tumor while providing ketones for an alternative fuel, could ameliorate potential side effects (150-152). While the ketogenic diet was originally developed to treat epilepsy, it has been of interest for cancer therapy (152-158), with multiple trials currently listed on clinicaltrials.gov as recruiting patients. Unfortunately, it is difficult for patients to maintain the ketogenic diet and success has been limited (152-158), but this relatively simple intervention will permit evaluation of impacts when used in combination with current standard of care.

Glucose metabolism can also be inhibited by the non-metabolizable analog of glucose, 2-deoxy glucose (2-DG). 2-DG can reduce glucose uptake and tumor growth in animal models, but is not-specifically imported by tumor cells (159). However, one clinical trial indicated 2-DG was well tolerated in patients with glioma (160). A recent study showed that the ketogenic diet was able to increase the maximum tolerated dose of 2-DG and reduce its side effects in non-tumor bearing mice (161). 2-DG has also been reported to reduce leukemia cell viability at low doses(162), and in combination with the pentose phosphate pathway inhibitor dehydroepiandrosterone was effective at lower doses (163). 2-DG, however, has not been fully explored due to concerns regarding toxicities (159,164). These data lead to the potential of combining metabolic therapies at lower doses to potentiate possible toxicities as a potential therapeutic option for glioma in particular.

Dichloroacetate is a clinically utilized inhibitor of glycolysis via targeting of pyruvate dehydrogenase, but it is also controversial as it is an environmental toxin with the potential to induce liver toxicities and neoplasia (165-167). However, as dichloroacetate is blood brain barrier penetrant, many studies have tested its efficacy against gliomas, with significant antitumor effects shown, depending on the dosage and schedule (168,169). Two completed clinical trials demonstrated that dichloroacetate was safe, and Michelakis and colleagues. indicated dichloroacetate may provide a therapeutic benefit for some patients with glioma (170,171). In leukemia, dichloroacetate was mainly studied in the context of B-chronic lymphocytic leukemia where it decreased viability of p53 wildtype cells and also synergized with the p53 activator, Nutlin-3 (172-174). Dichloroacetate was also reported to work in concert with metformin, another agent that targets metabolism as described below, to induce B-chronic lymphocytic leukemia cell death (175). Again, these data suggest that combinations of metabolic therapies have the potential to target both glioma and leukemia.

Metformin is more commonly known as an anti-diabetic therapy, however, it has become of interest as a cancer therapeutic because it targets mitochondrial metabolism. There are over 250 clinical trials currently active or complete. In both glioma and leukemia, metformin has shown anti-cancer efficacy alone and in combination with chemotherapies (176-179) (Supplemental Table 1). Metformin or its more potent analog phenformin have been reported to activate adenosine monophosphate-activated protein kinase (AMPK), leading to glioma or leukemia cell death (180,181), but the clinical benefit is not yet clear and development of lactic acidosis is a risk. Additional preclinical studies have investigated the combination of metformin and arsenic trioxide in glioma and observed a decrease in proliferation and elevated autophagy and apoptosis (182). Both of these agents lead to the dysregulation of mitochondrial metabolism and may have potential benefits in various combinations. Arsenic trioxide and its various derivatives have shown much promise in vitro with dramatic reductions in tumor growth through inhibition of hexokinase 2 or activation of AMPK (183-185). In vivo studies have been inconclusive as subcutaneous studies showed tumor regression (184), but intracranial studies indicated that insufficient drug was able to cross the blood brain barrier to promote survival (186). Nonetheless, over 75 studies have investigated arsenic trioxide compounds for the treatment of glioma or leukemia in the clinic. Unfortunately, one of the most efficacious compounds in vitro, 4-(N-(S-penicillaminylacetyl)amino) phenylarsonous acid, did not increase progression free survival time in a clinical trial, although it was shown to be safe (187). Overall, many new approaches to targeting glucose and mitochondrial metabolism are beginning to be investigated in the clinic, and the results of these ongoing studies will be of much interest as they are reported.

On the basis of the utilization of fatty acids in cancers, including the therapy resistant cancer stem cells, inhibition of fatty acid oxidation with etomoxir has been explored as a potential therapeutic strategy. Etomoxir treatment decreased the percentage of leukemia stem cells, sensitized them to apoptosis (188,189) and even resensitized tyrosine kinase inhibitor ibrutinib resistant cells by targeting the metabolic shift to fatty acid oxidation in those cells (190). However, it is important to note that, in addition to inhibiting CPT1 as mentioned above, etomoxir also inhibits diglyceride aceyltransferase (regulating triacylglycerol production and retinoid signaling) and has the ability to inhibit coenzyme-A at high concentrations (191). In human glioblastoma, including glioma stem cells, etomoxir decreased cell proliferation in multiple assays (88,192) and improved survival of orthotopic tumor bearing mice with etomoxir delivered via osmotic pumps (192). Alternative methods to target fatty acid oxidation with the lipid avocatin B (193), perhexiline (194), or ST1326 (195) also increased leukemia cell death. Linking these growth inhibitory data to cellular bioenergetics, adenosine triphosphate (ATP) levels, and oxygen consumption rates in both glioblastoma and leukemia cells were decreased with etomoxir treatment (88,189,192). While these data suggest the potential of targeting of fatty acid oxidation as an anti-glioma therapy, hepatotoxicity and variability in glioma stem cell responses to etomoxir/fatty acid oxidation inhibitors due, in part to metabolic heterogeneity and plasticity will complicate patient treatments. Metabolic profiling of glioma stem cell lines indicated a subset with elevated lipid signaling as well as cells with increased lipid droplet formation after treatment with the ATP synthase inhibitor oligomycin. However, these cells did not all respond similarly to etomoxir treatment. Cells either increased or decreased number in response to etomoxir (196) indicating that more research will be needed to fully understand the context dependent signaling at play, likely due to further metabolic plasticity.

In addition, direct targeting of fatty acid synthesis via pharmacologic or genetic targeting of ACC1/2 has also been shown to decrease the growth of glioblastoma cells in association with reduced ATP production (197). However, long-term treatment of cells with an ACC1/2 inhibitor resulted in increased extracellular acidification in some glioblastoma cells, suggesting the potential for shifts toward glycolysis to compensate for loss of fatty acid synthesis (197). In addition, statins, also known as 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase inhibitors, showed efficacy in vitro via inhibiting cholesterol production in glioma and leukemia, leading to decreased proliferation and increased apoptosis (198-202). Statins are particularly attractive as they have a long clinical history, limited toxicities, and may reduce graft-vs-host disease in patients with acute myeloid leukemia, although this was not seen across all leukemia subtypes (203). A combination of idarubicin and cytarabine with pravastatin was shown to both promote regression or have no benefit, indicating that further studies are warranted to investigate the benefit of using such a widely available drug as an adjuvant therapy (204,205) as well as the possibility that other combinations may prove more beneficial (Supplemental Table 1). While in vitro studies were encouraging (206), clinical evidence for the benefit of statins against gliomas is lacking, although there are a number of ongoing trials further investigating their role (207-210) (Supplemental Table 1). When considered more broadly, the data suggest there must be additional investigations to determine the best strategies for the application of metabolic inhibitors for anti-cancer therapies.

Conclusions

We summarize evidence that pathways common between normal and neoplastic stem cells, which regulate cell fate and survival mediate changes in metabolism, need to be more closely considered for developing novel therapeutics (Figure 2A). The data suggest that the metabolic plasticity intrinsic to cancer biology is itself a metabolic target. Given the inherent plasticity of the cancer cell, this will likely require a strategy that develops a multi-pronged approach to prevent the evolutionary adaptations for cell survival from thriving in cancer cells (Figure 2B). In limited studies where strategies to target both glycolysis and fatty acid oxidation have been tested, some benefits of the combinations have been demonstrated. Additional studies have indicated that combinatorial therapies may provide for a lower effective dose and thereby limit toxicities in non-malignant tissues that do not rely on multiple highly functioning metabolic pathways. Thus, there may be benefits from combinatorial metabolism targeting approaches that have not yet been identified.

Figure 2. Summary of Metabolic Plasticity in the Context of Stem Cell State and Pathobiology and Potential Therapeutic Interventions.

Figure 2.

(A) Tumor environments, importantly oxygen tension as well as nutrient availability, potently impact cellular metabolism which is critical for cell state transitions. Cancer cell activation of developmental programs permits metabolic plasticity critical for adaptations necessary for cell survival. (B) Multiple strategies and specific inhibitors are available to target glycolysis, oxidative phosphorylation, and fatty acid oxidation in cancer cells, but specificity to the tumor is likely to be a concern. By preventing alternative fuel sources and/or maintenance of a quiescent population, combinatorial approaches may sensitize tumor cells to chemo- and radiotherapy.

Supplementary Material

1

Acknowledgements

The authors would like to thank Dr. Burt Nabors and Dr. Ravi Bhatia for their reading of the manuscript. This work was supported by National Institutes of Health grant 1R01NS104339 to ABH, F31NS10545801A1 to CJL and UAB Nathan Shock Center P30AG050886 to VDU.

Abbreviations:

2-DG

2-deoxy-D-glucose

ACC

acetyl-coA carboxylase

AMPK

adenosine monophosphate-activated protein kinase

ATP

adenosine triphosphate

CPT1

carnitine palmitoyltransferase 1

FDG

2-deoxy-2-[18F]fluoro-D-glucose

FDOPA

3,4-dihydroxy-6-[18F]fluoro-L-phenylalanine

FLT

2’-deoxy-2’-[18F]fluorothymidine

GLUT

glucose transporter

HIF

hypoxia inducible transcription factor

MR

magnetic resonance

MRS

MR spectroscopy

PET

positron emission tomography

PHD3

prolyl hydroxylase 3

PML

promyelocytic leukemia protein

PPARγ

peroxisome proliferator-activated receptor γ

ROS

reactive oxygen species

Footnotes

Potential Conflicts of Interest: The authors declare no conflicts of interest.

References

  • 1.Hanahan D, and Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144, 646–674 [DOI] [PubMed] [Google Scholar]
  • 2.Koppenol WH, Bounds PL, and Dang CV (2011) Otto Warburg’s contributions to current concepts of cancer metabolism. Nat Rev Cancer 11, 325–337 [DOI] [PubMed] [Google Scholar]
  • 3.Luo M, and Wicha MS (2015) Metabolic plasticity of cancer stem cells. Oncotarget 6, 35141–35142 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Jia D, Lu M, Jung KH, Park JH, Yu L, Onuchic JN, Kaipparettu BA, and Levine H (2019) Elucidating cancer metabolic plasticity by coupling gene regulation with metabolic pathways. Proc Natl Acad Sci U S A 116, 3909–3918 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Levine AJ, and Puzio-Kuter AM (2010) The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes. Science 330, 1340–1344 [DOI] [PubMed] [Google Scholar]
  • 6.Al Tameemi W, Dale TP, Al-Jumaily RMK, and Forsyth NR (2019) Hypoxia-Modified Cancer Cell Metabolism. Front Cell Dev Biol 7, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Noe JT, and Mitchell RA (2019) Tricarboxylic acid cycle metabolites in the control of macrophage activation and effector phenotypes. J Leukoc Biol [DOI] [PubMed] [Google Scholar]
  • 8.Saha S, Shalova IN, and Biswas SK (2017) Metabolic regulation of macrophage phenotype and function. Immunol Rev 280, 102–111 [DOI] [PubMed] [Google Scholar]
  • 9.van der Windt GJ, O’Sullivan D, Everts B, Huang SC, Buck MD, Curtis JD, Chang CH, Smith AM, Ai T, Faubert B, Jones RG, Pearce EJ, and Pearce EL (2013) CD8 memory T cells have a bioenergetic advantage that underlies their rapid recall ability. Proceedings of the National Academy of Sciences of the United States of America 110, 14336–14341 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Pearce EL, Poffenberger MC, Chang CH, and Jones RG (2013) Fueling immunity: insights into metabolism and lymphocyte function. Science 342, 1242454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Zhang H, Badur MG, Divakaruni AS, Parker SJ, Jager C, Hiller K, Murphy AN, and Metallo CM (2016) Distinct Metabolic States Can Support Self-Renewal and Lipogenesis in Human Pluripotent Stem Cells under Different Culture Conditions. Cell Rep 16, 1536–1547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gu W, Gaeta X, Sahakyan A, Chan AB, Hong CS, Kim R, Braas D, Plath K, Lowry WE, and Christofk HR (2016) Glycolytic Metabolism Plays a Functional Role in Regulating Human Pluripotent Stem Cell State. Cell Stem Cell 19, 476–490 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Folmes CD, Dzeja PP, Nelson TJ, and Terzic A (2012) Metabolic plasticity in stem cell homeostasis and differentiation. Cell Stem Cell 11, 596–606 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Bonnet D, and Dick JE (1997) Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 3, 730–737 [DOI] [PubMed] [Google Scholar]
  • 15.Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, and Dirks PB (2004) Identification of human brain tumour initiating cells. Nature 432, 396–401 [DOI] [PubMed] [Google Scholar]
  • 16.Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, De Vitis S, Fiocco R, Foroni C, Dimeco F, and Vescovi A (2004) Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res 64, 7011–7021 [DOI] [PubMed] [Google Scholar]
  • 17.Lathia JD, Mack SC, Mulkearns-Hubert EE, Valentim CL, and Rich JN (2015) Cancer stem cells in glioblastoma. Genes Dev 29, 1203–1217 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Siebzehnrubl FA, Reynolds BA, Vescovi A, Steindler DA, and Deleyrolle LP (2011) The origins of glioma: E Pluribus Unum? Glia 59, 1135–1147 [DOI] [PubMed] [Google Scholar]
  • 19.Visvader JE (2011) Cells of origin in cancer. Nature 469, 314. [DOI] [PubMed] [Google Scholar]
  • 20.Itzykson R, and Solary E (2013) An evolutionary perspective on chronic myelomonocytic leukemia. Leukemia 27, 1441. [DOI] [PubMed] [Google Scholar]
  • 21.Jan M, and Majeti R (2012) Clonal evolution of acute leukemia genomes. Oncogene 32, 135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Jan M, Snyder TM, Corces-Zimmerman MR, Vyas P, Weissman IL, Quake SR, and Majeti R (2012) Clonal Evolution of Preleukemic Hematopoietic Stem Cells Precedes Human Acute Myeloid Leukemia. Science Translational Medicine 4, 149ra118–149ra118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Singh SK, Clarke ID, Hide T, and Dirks PB (2004) Cancer stem cells in nervous system tumors. Oncogene 23, 7267–7273 [DOI] [PubMed] [Google Scholar]
  • 24.Sancho P, Barneda D, and Heeschen C (2016) Hallmarks of cancer stem cell metabolism. Br J Cancer 114, 1305–1312 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wu J, Ocampo A, and Belmonte JCI (2016) Cellular Metabolism and Induced Pluripotency. Cell 166, 1371–1385 [DOI] [PubMed] [Google Scholar]
  • 26.Jia D, Park JH, Jung KH, Levine H, and Kaipparettu BA (2018) Elucidating the Metabolic Plasticity of Cancer: Mitochondrial Reprogramming and Hybrid Metabolic States. Cells 7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Menendez JA (2015) Metabolic control of cancer cell stemness: Lessons from iPS cells. Cell Cycle 14, 3801–3811 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lehuede C, Dupuy F, Rabinovitch R, Jones RG, and Siegel PM (2016) Metabolic Plasticity as a Determinant of Tumor Growth and Metastasis. Cancer Res 76, 5201–5208 [DOI] [PubMed] [Google Scholar]
  • 29.Covello KL, Kehler J, Yu H, Gordan JD, Arsham AM, Hu CJ, Labosky PA, Simon MC, and Keith B (2006) HIF-2alpha regulates Oct-4: effects of hypoxia on stem cell function, embryonic development, and tumor growth. Genes Dev 20, 557–570 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Spencer JA, Ferraro F, Roussakis E, Klein A, Wu J, Runnels JM, Zaher W, Mortensen LJ, Alt C, Turcotte R, Yusuf R, Cote D, Vinogradov SA, Scadden DT, and Lin CP (2014) Direct measurement of local oxygen concentration in the bone marrow of live animals. Nature 508, 269–273 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Mantel CR, O’Leary HA, Chitteti BR, Huang X, Cooper S, Hangoc G, Brustovetsky N, Srour EF, Lee MR, Messina-Graham S, Haas DM, Falah N, Kapur R, Pelus LM, Bardeesy N, Fitamant J, Ivan M, Kim KS, and Broxmeyer HE (2015) Enhancing Hematopoietic Stem Cell Transplantation Efficacy by Mitigating Oxygen Shock. Cell 161, 1553–1565 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wielockx B, Grinenko T, Mirtschink P, and Chavakis T (2019) Hypoxia Pathway Proteins in Normal and Malignant Hematopoiesis. Cells 8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Karigane D, and Takubo K (2017) Metabolic regulation of hematopoietic and leukemic stem/progenitor cells under homeostatic and stress conditions. Int J Hematol 106, 18–26 [DOI] [PubMed] [Google Scholar]
  • 34.Semenza GL (2017) Hypoxia-inducible factors: coupling glucose metabolism and redox regulation with induction of the breast cancer stem cell phenotype. EMBO J 36, 252–259 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Saito S, Lin YC, Tsai MH, Lin CS, Murayama Y, Sato R, and Yokoyama KK (2015) Emerging roles of hypoxia-inducible factors and reactive oxygen species in cancer and pluripotent stem cells. Kaohsiung J Med Sci 31, 279–286 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Morrison SJ, Csete M, Groves AK, Melega W, Wold B, and Anderson DJ (2000) Culture in reduced levels of oxygen promotes clonogenic sympathoadrenal differentiation by isolated neural crest stem cells. J Neurosci 20, 7370–7376 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Studer L, Csete M, Lee SH, Kabbani N, Walikonis J, Wold B, and McKay R (2000) Enhanced proliferation, survival, and dopaminergic differentiation of CNS precursors in lowered oxygen. J Neurosci 20, 7377–7383 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Simsek T, Kocabas F, Zheng J, Deberardinis RJ, Mahmoud AI, Olson EN, Schneider JW, Zhang CC, and Sadek HA (2010) The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell 7, 380–390 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Candelario KM, Shuttleworth CW, and Cunningham LA (2013) Neural stem/progenitor cells display a low requirement for oxidative metabolism independent of hypoxia inducible factor-1alpha expression. J Neurochem 125, 420–429 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Zhu LL, Zhao T, huang X, Liu ZH, Wu LY, Wu KW, and Fan M (2011) Gene expression profiles and metabolic changes in embryonic neural progenitor cells under low oxygen. Cell Reprogram 13, 113–120 [DOI] [PubMed] [Google Scholar]
  • 41.Mathieu J, Zhou W, Xing Y, Sperber H, Ferreccio A, Agoston Z, Kuppusamy KT, Moon RT, and Ruohola-Baker H (2014) Hypoxia-inducible factors have distinct and stage-specific roles during reprogramming of human cells to pluripotency. Cell Stem Cell 14, 592–605 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Nagao A, Kobayashi M, Koyasu S, Chow CCT, and Harada H (2019) HIF-1-Dependent Reprogramming of Glucose Metabolic Pathway of Cancer Cells and Its Therapeutic Significance. Int J Mol Sci 20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Rouault-Pierre K, Lopez-Onieva L, Foster K, Anjos-Afonso F, Lamrissi-Garcia I, Serrano-Sanchez M, Mitter R, Ivanovic Z, de Verneuil H, Gribben J, Taussig D, Rezvani HR, Mazurier F, and Bonnet D (2013) HIF-2alpha protects human hematopoietic stem/progenitors and acute myeloid leukemic cells from apoptosis induced by endoplasmic reticulum stress. Cell Stem Cell 13, 549–563 [DOI] [PubMed] [Google Scholar]
  • 44.Takeuchi M, Kimura S, Kuroda J, Ashihara E, Kawatani M, Osada H, Umezawa K, Yasui E, Imoto M, Tsuruo T, Yokota A, Tanaka R, Nagao R, Nakahata T, Fujiyama Y, and Maekawa T (2010) Glyoxalase-I is a novel target against Bcr-Abl+ leukemic cells acquiring stem-like characteristics in a hypoxic environment. Cell Death Differ 17, 1211–1220 [DOI] [PubMed] [Google Scholar]
  • 45.Shibao S, Minami N, Koike N, Fukui N, Yoshida K, Saya H, and Sampetrean O (2018) Metabolic heterogeneity and plasticity of glioma stem cells in a mouse glioblastoma model. Neuro Oncol 20, 343–354 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Li Z, Bao S, Wu Q, Wang H, Eyler C, Sathornsumetee S, Shi Q, Cao Y, Lathia J, McLendon RE, Hjelmeland AB, and Rich JN (2009) Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer cell 15, 501–513 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Giambra V, Jenkins CE, Lam SH, Hoofd C, Belmonte M, Wang X, Gusscott S, Gracias D, and Weng AP (2015) Leukemia stem cells in T-ALL require active Hif1alpha and Wnt signaling. Blood 125, 3917–3927 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Fallah J, and Rini BI (2019) HIF Inhibitors: Status of Current Clinical Development. Curr Oncol Rep 21, 6. [DOI] [PubMed] [Google Scholar]
  • 49.Magliulo D, and Bernardi R (2018) HIF-alpha factors as potential therapeutic targets in leukemia. Expert Opin Ther Targets 22, 917–928 [DOI] [PubMed] [Google Scholar]
  • 50.Vukovic M, Sepulveda C, Subramani C, Guitart AV, Mohr J, Allen L, Panagopoulou TI, Paris J, Lawson H, Villacreces A, Armesilla-Diaz A, Gezer D, Holyoake TL, Ratcliffe PJ, and Kranc KR (2016) Adult hematopoietic stem cells lacking Hif-1alpha self-renew normally. Blood 127, 2841–2846 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Renfrow JJ, Soike MH, Debinski W, Ramkissoon SH, Mott RT, Frenkel MB, Sarkaria JN, Lesser GJ, and Strowd RE (2018) Hypoxia-inducible factor 2alpha: a novel target in gliomas. Future Med Chem 10, 2227–2236 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Xie C, Gao X, Sun D, Zhang Y, Krausz KW, Qin X, and Gonzalez FJ (2018) Metabolic Profiling of the Novel Hypoxia-Inducible Factor 2alpha Inhibitor PT2385 In Vivo and In Vitro. Drug Metab Dispos 46, 336–345 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Murugesan T, Rajajeyabalachandran G, Kumar S, Nagaraju S, and Jegatheesan SK (2018) Targeting HIF-2alpha as therapy for advanced cancers. Drug Discov Today 23, 1444–1451 [DOI] [PubMed] [Google Scholar]
  • 54.Courtney KD, Infante JR, Lam ET, Figlin RA, Rini BI, Brugarolas J, Zojwalla NJ, Lowe AM, Wang K, Wallace EM, Josey JA, and Choueiri TK (2018) Phase I Dose-Escalation Trial of PT2385, a First-in-Class Hypoxia-Inducible Factor-2alpha Antagonist in Patients With Previously Treated Advanced Clear Cell Renal Cell Carcinoma. J Clin Oncol 36, 867–874 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Liang R, and Ghaffari S (2017) Mitochondria and FOXO3 in stem cell homeostasis, a window into hematopoietic stem cell fate determination. J Bioenerg Biomembr 49, 343–346 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Santo EE, and Paik J (2018) FOXO in Neural Cells and Diseases of the Nervous System. Curr Top Dev Biol 127, 105–118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Ferber EC, Peck B, Delpuech O, Bell GP, East P, and Schulze A (2012) FOXO3a regulates reactive oxygen metabolism by inhibiting mitochondrial gene expression. Cell Death Differ 19, 968–979 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Miyamoto K, Araki KY, Naka K, Arai F, Takubo K, Yamazaki S, Matsuoka S, Miyamoto T, Ito K, Ohmura M, Chen C, Hosokawa K, Nakauchi H, Nakayama K, Nakayama KI, Harada M, Motoyama N, Suda T, and Hirao A (2007) Foxo3a is essential for maintenance of the hematopoietic stem cell pool. Cell Stem Cell 1, 101–112 [DOI] [PubMed] [Google Scholar]
  • 59.Miyamoto K, Miyamoto T, Kato R, Yoshimura A, Motoyama N, and Suda T (2008) FoxO3a regulates hematopoietic homeostasis through a negative feedback pathway in conditions of stress or aging. Blood 112, 4485–4493 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hinman RM, Nichols WA, Diaz TM, Gallardo TD, Castrillon DH, and Satterthwaite AB (2009) Foxo3−/− mice demonstrate reduced numbers of pre-B and recirculating B cells but normal splenic B cell sub-population distribution. Int Immunol 21, 831–842 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Zhang X, Camprecios G, Rimmele P, Liang R, Yalcin S, Mungamuri SK, Barminko J, D’Escamard V, Baron MH, Brugnara C, Papatsenko D, Rivella S, and Ghaffari S (2014) FOXO3-mTOR metabolic cooperation in the regulation of erythroid cell maturation and homeostasis. Am J Hematol 89, 954–963 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Renault VM, Rafalski VA, Morgan AA, Salih DA, Brett JO, Webb AE, Villeda SA, Thekkat PU, Guillerey C, Denko NC, Palmer TD, Butte AJ, and Brunet A (2009) FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell 5, 527–539 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Schmidt-Strassburger U, Schips TG, Maier HJ, Kloiber K, Mannella F, Braunstein KE, Holzmann K, Ushmorov A, Liebau S, Boeckers TM, and Wirth T (2012) Expression of constitutively active FoxO3 in murine forebrain leads to a loss of neural progenitors. FASEB J 26, 4990–5001 [DOI] [PubMed] [Google Scholar]
  • 64.Yeo H, Lyssiotis CA, Zhang Y, Ying H, Asara JM, Cantley LC, and Paik JH (2013) FoxO3 coordinates metabolic pathways to maintain redox balance in neural stem cells. EMBO J 32, 2589–2602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Yalcin S, Zhang X, Luciano JP, Mungamuri SK, Marinkovic D, Vercherat C, Sarkar A, Grisotto M, Taneja R, and Ghaffari S (2008) Foxo3 is essential for the regulation of ataxia telangiectasia mutated and oxidative stress-mediated homeostasis of hematopoietic stem cells. J Biol Chem 283, 25692–25705 [DOI] [PubMed] [Google Scholar]
  • 66.Rimmele P, Liang R, Bigarella CL, Kocabas F, Xie J, Serasinghe MN, Chipuk J, Sadek H, Zhang CC, and Ghaffari S (2015) Mitochondrial metabolism in hematopoietic stem cells requires functional FOXO3. EMBO Rep 16, 1164–1176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Naka K, Hoshii T, and Hirao A (2010) Novel therapeutic approach to eradicate tyrosine kinase inhibitor resistant chronic myeloid leukemia stem cells. Cancer Sci 101, 1577–1581 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Naka K, Hoshii T, Muraguchi T, Tadokoro Y, Ooshio T, Kondo Y, Nakao S, Motoyama N, and Hirao A (2010) TGF-beta-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia. Nature 463, 676–680 [DOI] [PubMed] [Google Scholar]
  • 69.Brucker DP, Maurer GD, Harter PN, Rieger J, and Steinbach JP (2016) FOXO3a orchestrates glioma cell responses to starvation conditions and promotes hypoxia-induced cell death. Int J Oncol 49, 2399–2410 [DOI] [PubMed] [Google Scholar]
  • 70.Xu K, Zhang Z, Pei H, Wang H, Li L, and Xia Q (2017) FoxO3a induces temozolomide resistance in glioblastoma cells via the regulation of beta-catenin nuclear accumulation. Oncol Rep 37, 2391–2397 [DOI] [PubMed] [Google Scholar]
  • 71.Oliva CR, Moellering DR, Gillespie GY, and Griguer CE (2011) Acquisition of chemoresistance in gliomas is associated with increased mitochondrial coupling and decreased ROS production. PLoS One 6, e24665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Oliva CR, Nozell SE, Diers A, McClugage SG 3rd, Sarkaria JN, Markert JM, Darley-Usmar VM, Bailey SM, Gillespie GY, Landar A, and Griguer CE (2010) Acquisition of temozolomide chemoresistance in gliomas leads to remodeling of mitochondrial electron transport chain. J Biol Chem 285, 39759–39767 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Firat E, and Niedermann G (2016) FoxO proteins or loss of functional p53 maintain stemness of glioblastoma stem cells and survival after ionizing radiation plus PI3K/mTOR inhibition. Oncotarget 7, 54883–54896 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Sunayama J, Sato A, Matsuda K, Tachibana K, Watanabe E, Seino S, Suzuki K, Narita Y, Shibui S, Sakurada K, Kayama T, Tomiyama A, and Kitanaka C (2011) FoxO3a functions as a key integrator of cellular signals that control glioblastoma stem-like cell differentiation and tumorigenicity. Stem Cells 29, 1327–1337 [DOI] [PubMed] [Google Scholar]
  • 75.Carracedo A, Cantley LC, and Pandolfi PP (2013) Cancer metabolism: fatty acid oxidation in the limelight. Nat Rev Cancer 13, 227–232 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Stoll EA, Makin R, Sweet IR, Trevelyan AJ, Miwa S, Horner PJ, and Turnbull DM (2015) Neural Stem Cells in the Adult Subventricular Zone Oxidize Fatty Acids to Produce Energy and Support Neurogenic Activity. Stem Cells 33, 2306–2319 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ito K, Bernardi R, Morotti A, Matsuoka S, Saglio G, Ikeda Y, Rosenblatt J, Avigan DE, Teruya-Feldstein J, and Pandolfi PP (2008) PML targeting eradicates quiescent leukaemia-initiating cells. Nature 453, 1072–1078 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Ito K, Carracedo A, Weiss D, Arai F, Ala U, Avigan DE, Schafer ZT, Evans RM, Suda T, Lee CH, and Pandolfi PP (2012) A PML-PPAR-delta pathway for fatty acid oxidation regulates hematopoietic stem cell maintenance. Nat Med 18, 1350–1358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lallemand-Breitenbach V, and de The H (2012) Hematopoietic stem cells burn fat to prevent exhaustion. Cell Stem Cell 11, 447–449 [DOI] [PubMed] [Google Scholar]
  • 80.Yusuf RZ, and Scadden DT (2012) Fate through fat: lipid metabolism determines stem cell division outcome. Cell Metab 16, 411–413 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Regad T, Bellodi C, Nicotera P, and Salomoni P (2009) The tumor suppressor Pml regulates cell fate in the developing neocortex. Nat Neurosci 12, 132–140 [DOI] [PubMed] [Google Scholar]
  • 82.Reilly SM, and Lee CH (2008) PPAR delta as a therapeutic target in metabolic disease. FEBS Lett 582, 26–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Melone MAB, Valentino A, Margarucci S, Galderisi U, Giordano A, and Peluso G (2018) The carnitine system and cancer metabolic plasticity. Cell Death Dis 9, 228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Cabon L, Bertaux A, Brunelle-Navas MN, Nemazanyy I, Scourzic L, Delavallee L, Vela L, Baritaud M, Bouchet S, Lopez C, Quang Van V, Garbin K, Chateau D, Gilard F, Sarfati M, Mercher T, Bernard OA, and Susin SA (2018) AIF loss deregulates hematopoiesis and reveals different adaptive metabolic responses in bone marrow cells and thymocytes. Cell Death Differ 25, 983–1001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Knobloch M, Pilz GA, Ghesquiere B, Kovacs WJ, Wegleiter T, Moore DL, Hruzova M, Zamboni N, Carmeliet P, and Jessberger S (2017) A Fatty Acid Oxidation-Dependent Metabolic Shift Regulates Adult Neural Stem Cell Activity. Cell Rep 20, 2144–2155 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Xie Z, Jones A, Deeney JT, Hur SK, and Bankaitis VA (2016) Inborn Errors of Long-Chain Fatty Acid beta-Oxidation Link Neural Stem Cell Self-Renewal to Autism. Cell Rep 14, 991–999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ye H, Adane B, Khan N, Sullivan T, Minhajuddin M, Gasparetto M, Stevens B, Pei S, Balys M, Ashton JM, Klemm DJ, Woolthuis CM, Stranahan AW, Park CY, and Jordan CT (2016) Leukemic Stem Cells Evade Chemotherapy by Metabolic Adaptation to an Adipose Tissue Niche. Cell Stem Cell 19, 23–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Pike LS, Smift AL, Croteau NJ, Ferrick DA, and Wu M (2011) Inhibition of fatty acid oxidation by etomoxir impairs NADPH production and increases reactive oxygen species resulting in ATP depletion and cell death in human glioblastoma cells. Biochim Biophys Acta 1807, 726–734 [DOI] [PubMed] [Google Scholar]
  • 89.Pike Winer LS, and Wu M (2014) Rapid analysis of glycolytic and oxidative substrate flux of cancer cells in a microplate. PLoS One 9, e109916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Zhou W, Cheng L, Shi Y, Ke SQ, Huang Z, Fang X, Chu CW, Xie Q, Bian XW, Rich JN, and Bao S (2015) Arsenic trioxide disrupts glioma stem cells via promoting PML degradation to inhibit tumor growth. Oncotarget 6, 37300–37315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Amodeo V, A D, Betts J, Bartesaghi S, Zhang Y, Richard-Londt A, Ellis M, R Roshani, M Vouri, Galavotti S, Oberndorfer S, Leite AP, Mackay A, Lampada A, Stratford EW, Li N, Dinsdale D, Grimwade D, Jones C, Nicotera P, Michod D, Brandner S, and Salomoni P. (2017) A PML/Slit Axis Controls Physiological Cell Migration and Cancer Invasion in the CNS. Cell Rep 20, 411–426 [DOI] [PubMed] [Google Scholar]
  • 92.Iwanami A, Gini B, Zanca C, Matsutani T, Assuncao A, Nael A, Dang J, Yang H, Zhu S, Kohyama J, Kitabayashi I, Cavenee WK, Cloughesy TF, Furnari FB, Nakamura M, Toyama Y, Okano H, and Mischel PS (2013) PML mediates glioblastoma resistance to mammalian target of rapamycin (mTOR)-targeted therapies. Proc Natl Acad Sci U S A 110, 4339–4344 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Wu HC, Lin YC, Liu CH, Chung HC, Wang YT, Lin YW, Ma HI, Tu PH, Lawler SE, and Chen RH (2014) USP11 regulates PML stability to control Notch-induced malignancy in brain tumours. Nat Commun 5, 3214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kuwayama K, Matsuzaki K, Mizobuchi Y, Mure H, Kitazato KT, Kageji T, Nakao M, and Nagahiro S (2009) Promyelocytic leukemia protein induces apoptosis due to caspase-8 activation via the repression of NFkappaB activation in glioblastoma. Neuro Oncol 11, 132–141 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Okazaki T, Kageji T, Kuwayama K, Kitazato KT, Mure H, Hara K, Morigaki R, Mizobuchi Y, Matsuzaki K, and Nagahiro S (2012) Up-regulation of endogenous PML induced by a combination of interferon-beta and temozolomide enhances p73/YAP-mediated apoptosis in glioblastoma. Cancer Lett 323, 199–207 [DOI] [PubMed] [Google Scholar]
  • 96.Wakamiya T, Suzuki SO, Hamasaki H, Honda H, Mizoguchi M, Yoshimoto K, and Iwaki T (2014) Elevated expression of fatty acid synthase and nuclear localization of carnitine palmitoyltransferase 1C are common among human gliomas. Neuropathology 34, 465–474 [DOI] [PubMed] [Google Scholar]
  • 97.German NJ, Yoon H, Yusuf RZ, Murphy JP, Finley LW, Laurent G, Haas W, Satterstrom FK, Guarnerio J, Zaganjor E, Santos D, Pandolfi PP, Beck AH, Gygi SP, Scadden DT, Kaelin WG Jr., and Haigis MC (2016) PHD3 Loss in Cancer Enables Metabolic Reliance on Fatty Acid Oxidation via Deactivation of ACC2. Mol Cell 63, 1006–1020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Henze AT, Garvalov BK, Seidel S, Cuesta AM, Ritter M, Filatova A, Foss F, Dopeso H, Essmann CL, Maxwell PH, Reifenberger G, Carmeliet P, Acker-Palmer A, and Acker T (2014) Loss of PHD3 allows tumours to overcome hypoxic growth inhibition and sustain proliferation through EGFR. Nat Commun 5, 5582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.McGarry JD (1995) Malonyl-CoA and carnitine palmitoyltransferase I: an expanding partnership. Biochem Soc Trans 23, 481–485 [DOI] [PubMed] [Google Scholar]
  • 100.Henze AT, Riedel J, Diem T, Wenner J, Flamme I, Pouyseggur J, Plate KH, and Acker T (2010) Prolyl hydroxylases 2 and 3 act in gliomas as protective negative feedback regulators of hypoxia-inducible factors. Cancer Res 70, 357–366 [DOI] [PubMed] [Google Scholar]
  • 101.Chen N, Rinner O, Czernik D, Nytko KJ, Zheng D, Stiehl DP, Zamboni N, Gstaiger M, and Frei C (2011) The oxygen sensor PHD3 limits glycolysis under hypoxia via direct binding to pyruvate kinase. Cell Res 21, 983–986 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Bensaad K, Favaro E, Lewis CA, Peck B, Lord S, Collins JM, Pinnick KE, Wigfield S, Buffa FM, Li JL, Zhang Q, Wakelam MJO, Karpe F, Schulze A, and Harris AL (2014) Fatty acid uptake and lipid storage induced by HIF-1alpha contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep 9, 349–365 [DOI] [PubMed] [Google Scholar]
  • 103.Tohyama S, Fujita J, Hishiki T, Matsuura T, Hattori F, Ohno R, Kanazawa H, Seki T, Nakajima K, Kishino Y, Okada M, Hirano A, Kuroda T, Yasuda S, Sato Y, Yuasa S, Sano M, Suematsu M, and Fukuda K (2016) Glutamine Oxidation Is Indispensable for Survival of Human Pluripotent Stem Cells. Cell Metab 23, 663–674 [DOI] [PubMed] [Google Scholar]
  • 104.Altman BJ, Stine ZE, and Dang CV (2016) From Krebs to clinic: glutamine metabolism to cancer therapy. Nat Rev Cancer 16, 773. [DOI] [PubMed] [Google Scholar]
  • 105.Scalise M, Pochini L, Galluccio M, Console L, and Indiveri C (2017) Glutamine Transport and Mitochondrial Metabolism in Cancer Cell Growth. Front Oncol 7, 306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Li T, and Le A (2018) Glutamine Metabolism in Cancer. Adv Exp Med Biol 1063, 13–32 [DOI] [PubMed] [Google Scholar]
  • 107.Ehsanipour EA, Sheng X, Behan JW, Wang X, Butturini A, Avramis VI, and Mittelman SD (2013) Adipocytes cause leukemia cell resistance to L-asparaginase via release of glutamine. Cancer Res 73, 2998–3006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Goto M, Miwa H, Shikami M, Tsunekawa-Imai N, Suganuma K, Mizuno S, Takahashi M, Mizutani M, Hanamura I, and Nitta M (2014) Importance of glutamine metabolism in leukemia cells by energy production through TCA cycle and by redox homeostasis. Cancer Invest 32, 241–247 [DOI] [PubMed] [Google Scholar]
  • 109.Matre P, Velez J, Jacamo R, Qi Y, Su X, Cai T, Chan SM, Lodi A, Sweeney SR, Ma H, Davis RE, Baran N, Haferlach T, Su X, Flores ER, Gonzalez D, Konoplev S, Samudio I, DiNardo C, Majeti R, Schimmer AD, Li W, Wang T, Tiziani S, and Konopleva M (2016) Inhibiting glutaminase in acute myeloid leukemia: metabolic dependency of selected AML subtypes. Oncotarget 7, 79722–79735 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Daud H, Browne S, Al-Majmaie R, Murphy W, and Al-Rubeai M (2016) Metabolic profiling of hematopoietic stem and progenitor cells during proliferation and differentiation into red blood cells. N Biotechnol 33, 179–186 [DOI] [PubMed] [Google Scholar]
  • 111.Oburoglu L, Romano M, Taylor N, and Kinet S (2016) Metabolic regulation of hematopoietic stem cell commitment and erythroid differentiation. Curr Opin Hematol 23, 198–205 [DOI] [PubMed] [Google Scholar]
  • 112.Oburoglu L, Tardito S, Fritz V, de Barros SC, Merida P, Craveiro M, Mamede J, Cretenet G, Mongellaz C, An X, Klysz D, Touhami J, Boyer-Clavel M, Battini JL, Dardalhon V, Zimmermann VS, Mohandas N, Gottlieb E, Sitbon M, Kinet S, and Taylor N (2014) Glucose and glutamine metabolism regulate human hematopoietic stem cell lineage specification. Cell Stem Cell 15, 169–184 [DOI] [PubMed] [Google Scholar]
  • 113.Brazel CY, Nunez JL, Yang Z, and Levison SW (2005) Glutamate enhances survival and proliferation of neural progenitors derived from the subventricular zone. Neuroscience 131, 55–65 [DOI] [PubMed] [Google Scholar]
  • 114.Dunphy MPS, Harding JJ, Venneti S, Zhang H, Burnazi EM, Bromberg J, Omuro AM, Hsieh JJ, Mellinghoff IK, Staton K, Pressl C, Beattie BJ, Zanzonico PB, Gerecitano JF, Kelsen DP, Weber W, Lyashchenko SK, Kung HF, and Lewis JS (2018) In Vivo PET Assay of Tumor Glutamine Flux and Metabolism: In-Human Trial of (18)F-(2S,4R)-4-Fluoroglutamine. Radiology 287, 667–675 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kim MM, Parolia A, Dunphy MP, and Venneti S (2016) Non-invasive metabolic imaging of brain tumours in the era of precision medicine. Nat Rev Clin Oncol 13, 725–739 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Vlassenko AG, McConathy J, Couture LE, Su Y, Massoumzadeh P, Leeds HS, Chicoine MR, Tran DD, Huang J, Dahiya S, Marcus DS, Fouke SJ, Rich KM, Raichle ME, and Benzinger TL (2015) Aerobic Glycolysis as a Marker of Tumor Aggressiveness: Preliminary Data in High Grade Human Brain Tumors. Dis Markers 2015, 874904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Rhodes CG, Wise RJ, Gibbs JM, Frackowiak RS, Hatazawa J, Palmer AJ, Thomas DG, and Jones T (1983) In vivo disturbance of the oxidative metabolism of glucose in human cerebral gliomas. Ann Neurol 14, 614–626 [DOI] [PubMed] [Google Scholar]
  • 118.Mineura K, Yasuda T, Kowada M, Shishido F, Ogawa T, and Uemura K (1986) Positron emission tomographic evaluation of histological malignancy in gliomas using oxygen-15 and fluorine-18-fluorodeoxyglucose. Neurol Res 8, 164–168 [DOI] [PubMed] [Google Scholar]
  • 119.Choudhary G, Langen KJ, Galldiks N, and McConathy J (2018) Investigational PET tracers for high-grade gliomas. Q J Nucl Med Mol Imaging 62, 281–294 [DOI] [PubMed] [Google Scholar]
  • 120.Galldiks N, and Langen KJ (2017) Amino acid PET in neuro-oncology: applications in the clinic. Expert Rev Anticancer Ther 17, 395–397 [DOI] [PubMed] [Google Scholar]
  • 121.Law I, Albert NL, Arbizu J, Boellaard R, Drzezga A, Galldiks N, la Fougere C, Langen KJ, Lopci E, Lowe V, McConathy J, Quick HH, Sattler B, Schuster DM, Tonn JC, and Weller M (2019) Joint EANM/EANO/RANO practice guidelines/SNMMI procedure standards for imaging of gliomas using PET with radiolabelled amino acids and [(18)F]FDG: version 1.0. Eur J Nucl Med Mol Imaging 46, 540–557 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Albert NL, Weller M, Suchorska B, Galldiks N, Soffietti R, Kim MM, la Fougere C, Pope W, Law I, Arbizu J, Chamberlain MC, Vogelbaum M, Ellingson BM, and Tonn JC (2016) Response Assessment in Neuro-Oncology working group and European Association for Neuro-Oncology recommendations for the clinical use of PET imaging in gliomas. Neuro Oncol [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Zhu L, Ploessl K, Zhou R, Mankoff D, and Kung HF (2017) Metabolic Imaging of Glutamine in Cancer. J Nucl Med 58, 533–537 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Venneti S, Dunphy MP, Zhang H, Pitter KL, Zanzonico P, Campos C, Carlin SD, La Rocca G, Lyashchenko S, Ploessl K, Rohle D, Omuro AM, Cross JR, Brennan CW, Weber WA, Holland EC, Mellinghoff IK, Kung HF, Lewis JS, and Thompson CB (2015) Glutamine-based PET imaging facilitates enhanced metabolic evaluation of gliomas in vivo. Sci Transl Med 7, 274ra217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Wakabayashi T, Iuchi T, Tsuyuguchi N, Nishikawa R, Arakawa Y, Sasayama T, Miyake K, Nariai T, Narita Y, Hashimoto N, Okuda O, Matsuda H, Kubota K, Ito K, Nakazato Y, and Kubomura K (2017) Diagnostic Performance and Safety of Positron Emission Tomography Using 18F-Fluciclovine in Patients with Clinically Suspected High- or Low-grade Gliomas: A Multicenter Phase IIb Trial. Asia Ocean J Nucl Med Biol 5, 10–21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Tsuyuguchi N, Terakawa Y, Uda T, Nakajo K, and Kanemura Y (2017) Diagnosis of Brain Tumors Using Amino Acid Transport PET Imaging with 18F-fluciclovine: A Comparative Study with L-methyl-11C-methionine PET Imaging. Asia Ocean J Nucl Med Biol 5, 85–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Kondo A, Ishii H, Aoki S, Suzuki M, Nagasawa H, Kubota K, Minamimoto R, Arakawa A, Tominaga M, and Arai H (2016) Phase IIa clinical study of [18F]fluciclovine: efficacy and safety of a new PET tracer for brain tumors. Ann Nucl Med 30, 608–618 [DOI] [PubMed] [Google Scholar]
  • 128.Yamaguchi S, Hirata K, Toyonaga T, Kobayashi K, Ishi Y, Motegi H, Kobayashi H, Shiga T, Tamaki N, Terasaka S, and Houkin K (2016) Change in 18F-Fluoromisonidazole PET Is an Early Predictor of the Prognosis in the Patients with Recurrent High-Grade Glioma Receiving Bevacizumab Treatment. PLoS One 11, e0167917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Bell C, Dowson N, Fay M, Thomas P, Puttick S, Gal Y, and Rose S (2015) Hypoxia imaging in gliomas with 18F-fluoromisonidazole PET: toward clinical translation. Semin Nucl Med 45, 136–150 [DOI] [PubMed] [Google Scholar]
  • 130.Kobayashi H, Hirata K, Yamaguchi S, Terasaka S, Shiga T, and Houkin K (2013) Usefulness of FMISO-PET for glioma analysis. Neurol Med Chir (Tokyo) 53, 773–778 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Hirata K, Terasaka S, Shiga T, Hattori N, Magota K, Kobayashi H, Yamaguchi S, Houkin K, Tanaka S, Kuge Y, and Tamaki N (2012) 18F-Fluoromisonidazole positron emission tomography may differentiate glioblastoma multiforme from less malignant gliomas. Eur J Nucl Med Mol Imaging 39, 760–770 [DOI] [PubMed] [Google Scholar]
  • 132.Toyonaga T, Yamaguchi S, Hirata K, Kobayashi K, Manabe O, Watanabe S, Terasaka S, Kobayashi H, Hattori N, Shiga T, Kuge Y, Tanaka S, Ito YM, and Tamaki N (2017) Hypoxic glucose metabolism in glioblastoma as a potential prognostic factor. Eur J Nucl Med Mol Imaging 44, 611–619 [DOI] [PubMed] [Google Scholar]
  • 133.Takenaka S, Asano Y, Shinoda J, Nomura Y, Yonezawa S, Miwa K, Yano H, and Iwama T (2014) Comparison of 11C-methionine, 11C-choline, and 18F-fluorodeoxyglucose-PET for distinguishing glioma recurrence from radiation necrosis. Neurol Med Chir (Tokyo) 54, 280–289 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Li W, Ma L, Wang X, Sun J, Wang S, and Hu X (2014) 11C-choline PET/CT tumor recurrence detection and survival prediction in post-treatment patients with high-grade gliomas. Tumour Biol 35, 12353–12360 [DOI] [PubMed] [Google Scholar]
  • 135.Kato T, Shinoda J, Nakayama N, Miwa K, Okumura A, Yano H, Yoshimura S, Maruyama T, Muragaki Y, and Iwama T (2008) Metabolic assessment of gliomas using 11C-methionine, [18F] fluorodeoxyglucose, and 11C-choline positron-emission tomography. AJNR Am J Neuroradiol 29, 1176–1182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Tsuchida T, Takeuchi H, Okazawa H, Tsujikawa T, and Fujibayashi Y (2008) Grading of brain glioma with 1-11C-acetate PET: comparison with 18F-FDG PET. Nucl Med Biol 35, 171–176 [DOI] [PubMed] [Google Scholar]
  • 137.Yamamoto Y, Nishiyama Y, Kimura N, Kameyama R, Kawai N, Hatakeyama T, Kaji M, and Ohkawa M (2008) 11C-acetate PET in the evaluation of brain glioma: comparison with 11C-methionine and 18F-FDG-PET. Mol Imaging Biol 10, 281–287 [DOI] [PubMed] [Google Scholar]
  • 138.Cribe AS, Steenhof M, Marcher CW, Petersen H, Frederiksen H, and Friis LS (2013) Extramedullary disease in patients with acute myeloid leukemia assessed by 18F-FDG PET. Eur J Haematol 90, 273–278 [DOI] [PubMed] [Google Scholar]
  • 139.Zhou WL, Wu HB, Wang LJ, Tian Y, Dong Y, and Wang QS (2016) Usefulness and pitfalls of F-18-FDG PET/CT for diagnosing extramedullary acute leukemia. Eur J Radiol 85, 205–210 [DOI] [PubMed] [Google Scholar]
  • 140.Zhang S, Wang W, Kan Y, Liu J, and Yang J (2018) Extramedullary Infiltration of Acute Lymphoblastic Leukemia in Multiple Organs on FDG PET/CT. Clin Nucl Med 43, 217–219 [DOI] [PubMed] [Google Scholar]
  • 141.Vanderhoek M, Juckett MB, Perlman SB, Nickles RJ, and Jeraj R (2011) Early assessment of treatment response in patients with AML using [(18)F]FLT PET imaging. Leuk Res 35, 310–316 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Han EJ, Lee BH, Kim JA, Park YH, and Choi WH (2017) Early assessment of response to induction therapy in acute myeloid leukemia using (18)F-FLT PET/CT. EJNMMI Res 7, 75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Li Y, Park I, and Nelson SJ (2015) Imaging tumor metabolism using in vivo magnetic resonance spectroscopy. Cancer J 21, 123–128 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Gordon JW, Chen HY, Autry A, Park I, Van Criekinge M, Mammoli D, Milshteyn E, Bok R, Xu D, Li Y, Aggarwal R, Chang S, Slater JB, Ferrone M, Nelson S, Kurhanewicz J, Larson PEZ, and Vigneron DB (2019) Translation of Carbon-13 EPI for hyperpolarized MR molecular imaging of prostate and brain cancer patients. Magn Reson Med 81, 2702–2709 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Park I, Larson PEZ, Gordon JW, Carvajal L, Chen HY, Bok R, Van Criekinge M, Ferrone M, Slater JB, Xu D, Kurhanewicz J, Vigneron DB, Chang S, and Nelson SJ (2018) Development of methods and feasibility of using hyperpolarized carbon-13 imaging data for evaluating brain metabolism in patient studies. Magn Reson Med 80, 864–873 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Libby CJ, Zhang S, Benavides GA, Scott SE, Li Y, Redmann M, Tran AN, Otamias A, Darley-Usmar V, Napierala M, Zhang J, Augelli-Szafran CE, Zhang W, and Hjelmeland AB (2018) Identification of Compounds That Decrease Glioblastoma Growth and Glucose Uptake in Vitro. ACS Chem Biol [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Ojelabi OA, Lloyd KP, Simon AH, De Zutter JK, and Carruthers A (2016) WZB117 (2-Fluoro-6-(m-hydroxybenzoyloxy) Phenyl m-Hydroxybenzoate) Inhibits GLUT1-mediated Sugar Transport by Binding Reversibly at the Exofacial Sugar Binding Site. J Biol Chem 291, 26762–26772 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Siebeneicher H, Bauser M, Buchmann B, Heisler I, Muller T, Neuhaus R, Rehwinkel H, Telser J, and Zorn L (2016) Identification of novel GLUT inhibitors. Bioorg Med Chem Lett 26, 1732–1737 [DOI] [PubMed] [Google Scholar]
  • 149.Siebeneicher H, Cleve A, Rehwinkel H, Neuhaus R, Heisler I, Muller T, Bauser M, and Buchmann B (2016) Identification and Optimization of the First Highly Selective GLUT1 Inhibitor BAY-876. ChemMedChem 11, 2261–2271 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Flavahan WA, Wu Q, Hitomi M, Rahim N, Kim Y, Sloan AE, Weil RJ, Nakano I, Sarkaria JN, Stringer BW, Day BW, Li M, Lathia JD, Rich JN, and Hjelmeland AB (2013) Brain tumor initiating cells adapt to restricted nutrition through preferential glucose uptake. Nat Neurosci 16, 1373–1382 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Dai Y, Zhao Y, Tomi M, Shin BC, Thamotharan S, Mazarati A, Sankar R, Wang EA, Cepeda C, Levine MS, Zhang J, Frew A, Alger JR, Clark PM, Sondhi M, Kositamongkol S, Leibovitch L, and Devaskar SU (2017) Sex-Specific Life Course Changes in the Neuro-Metabolic Phenotype of Glut3 Null Heterozygous Mice: Ketogenic Diet Ameliorates Electroencephalographic Seizures and Improves Sociability. Endocrinology 158, 936–949 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Seyfried TN, Kiebish MA, Marsh J, Shelton LM, Huysentruyt LC, and Mukherjee P (2011) Metabolic management of brain cancer. Biochim Biophys Acta 1807, 577–594 [DOI] [PubMed] [Google Scholar]
  • 153.Champ CE, Palmer JD, Volek JS, Werner-Wasik M, Andrews DW, Evans JJ, Glass J, Kim L, and Shi W (2014) Targeting metabolism with a ketogenic diet during the treatment of glioblastoma multiforme. J Neurooncol 117, 125–131 [DOI] [PubMed] [Google Scholar]
  • 154.Nebeling LC, Miraldi F, Shurin SB, and Lerner E (1995) Effects of a ketogenic diet on tumor metabolism and nutritional status in pediatric oncology patients: two case reports. J Am Coll Nutr 14, 202–208 [DOI] [PubMed] [Google Scholar]
  • 155.Rieger J, Bahr O, Maurer GD, Hattingen E, Franz K, Brucker D, Walenta S, Kammerer U, Coy JF, Weller M, and Steinbach JP (2014) ERGO: a pilot study of ketogenic diet in recurrent glioblastoma. Int J Oncol 44, 1843–1852 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Schwartz K, Chang HT, Nikolai M, Pernicone J, Rhee S, Olson K, Kurniali PC, Hord NG, and Noel M (2015) Treatment of glioma patients with ketogenic diets: report of two cases treated with an IRB-approved energy-restricted ketogenic diet protocol and review of the literature. Cancer Metab 3, 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Woolf EC, Curley KL, Liu Q, Turner GH, Charlton JA, Preul MC, and Scheck AC (2015) The Ketogenic Diet Alters the Hypoxic Response and Affects Expression of Proteins Associated with Angiogenesis, Invasive Potential and Vascular Permeability in a Mouse Glioma Model. PLoS One 10, e0130357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Zuccoli G, Marcello N, Pisanello A, Servadei F, Vaccaro S, Mukherjee P, and Seyfried TN (2010) Metabolic management of glioblastoma multiforme using standard therapy together with a restricted ketogenic diet: Case Report. Nutr Metab (Lond) 7, 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Labak CM, Wang PY, Arora R, Guda MR, Asuthkar S, Tsung AJ, and Velpula KK (2016) Glucose transport: meeting the metabolic demands of cancer, and applications in glioblastoma treatment. Am J Cancer Res 6, 1599–1608 [PMC free article] [PubMed] [Google Scholar]
  • 160.Mohanti BK, Rath GK, Anantha N, Kannan V, Das BS, Chandramouli BA, Banerjee AK, Das S, Jena A, Ravichandran R, Sahi UP, Kumar R, Kapoor N, Kalia VK, Dwarakanath BS, and Jain V (1996) Improving cancer radiotherapy with 2-deoxy-D-glucose: phase I/II clinical trials on human cerebral gliomas. Int J Radiat Oncol Biol Phys 35, 103–111 [DOI] [PubMed] [Google Scholar]
  • 161.Voss M, Lorenz NI, Luger AL, Steinbach JP, Rieger J, and Ronellenfitsch MW (2018) Rescue of 2-Deoxyglucose Side Effects by Ketogenic Diet. Int J Mol Sci 19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Gu L, Yi Z, Zhang Y, Ma Z, Zhu Y, and Gao J (2017) Low dose of 2-deoxy-D-glucose kills acute lymphoblastic leukemia cells and reverses glucocorticoid resistance via N-linked glycosylation inhibition under normoxia. Oncotarget 8, 30978–30991 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Miwa H, Shikami M, Goto M, Mizuno S, Takahashi M, Tsunekawa-Imai N, Ishikawa T, Mizutani M, Horio T, Gotou M, Yamamoto H, Wakabayashi M, Watarai M, Hanamura I, Imamura A, Mihara H, and Nitta M (2013) Leukemia cells demonstrate a different metabolic perturbation provoked by 2-deoxyglucose. Oncol Rep 29, 2053–2057 [DOI] [PubMed] [Google Scholar]
  • 164.Libby CJ, Tran AN, Scott SE, Griguer C, and Hjelmeland AB (2018) The pro-tumorigenic effects of metabolic alterations in glioblastoma including brain tumor initiating cells. Biochim Biophys Acta 1869, 175–188 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Papandreou I, Goliasova T, and Denko NC (2011) Anticancer drugs that target metabolism: Is dichloroacetate the new paradigm? Int J Cancer 128, 1001–1008 [DOI] [PubMed] [Google Scholar]
  • 166.Stacpoole PW, Henderson GN, Yan Z, and James MO (1998) Clinical pharmacology and toxicology of dichloroacetate. Environ Health Perspect 106 Suppl 4, 989–994 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Stacpoole PW (2011) The dichloroacetate dilemma: environmental hazard versus therapeutic goldmine--both or neither? Environ Health Perspect 119, 155–158 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Fedorchuk AG, Pyaskovskaya ON, Gorbik GV, Prokhorova IV, Kolesnik DL, and Solyanik GI (2016) Effectiveness of sodium dichloroacetate against glioma C6 depends on administration schedule and dosage. Exp Oncol 38, 80–83 [PubMed] [Google Scholar]
  • 169.Duan Y, Zhao X, Ren W, Wang X, Yu KF, Li D, Zhang X, and Zhang Q (2013) Antitumor activity of dichloroacetate on C6 glioma cell: in vitro and in vivo evaluation. Onco Targets Ther 6, 189–198 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Michelakis ED, Sutendra G, Dromparis P, Webster L, Haromy A, Niven E, Maguire C, Gammer TL, Mackey JR, Fulton D, Abdulkarim B, McMurtry MS, and Petruk KC (2010) Metabolic modulation of glioblastoma with dichloroacetate. Sci Transl Med 2, 31ra34. [DOI] [PubMed] [Google Scholar]
  • 171.Dunbar EM, Coats BS, Shroads AL, Langaee T, Lew A, Forder JR, Shuster JJ, Wagner DA, and Stacpoole PW (2014) Phase 1 trial of dichloroacetate (DCA) in adults with recurrent malignant brain tumors. Invest New Drugs 32, 452–464 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Abramek J, Bogucki J, Ziaja-Soltys M, Stepniewski A, and Bogucka-Kocka A (2019) Effect of sodium dichloroacetate on apoptotic gene expression in human leukemia cell lines. Pharmacol Rep 71, 248–256 [DOI] [PubMed] [Google Scholar]
  • 173.Agnoletto C, Brunelli L, Melloni E, Pastorelli R, Casciano F, Rimondi E, Rigolin GM, Cuneo A, Secchiero P, and Zauli G (2015) The anti-leukemic activity of sodium dichloroacetate in p53mutated/null cells is mediated by a p53-independent ILF3/p21 pathway. Oncotarget 6, 2385–2396 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Agnoletto C, Melloni E, Casciano F, Rigolin GM, Rimondi E, Celeghini C, Brunelli L, Cuneo A, Secchiero P, and Zauli G (2014) Sodium dichloroacetate exhibits anti-leukemic activity in B-chronic lymphocytic leukemia (B-CLL) and synergizes with the p53 activator Nutlin-3. Oncotarget 5, 4347–4360 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Voltan R, Rimondi E, Melloni E, Gilli P, Bertolasi V, Casciano F, Rigolin GM, Zauli G, and Secchiero P (2016) Metformin combined with sodium dichloroacetate promotes B leukemic cell death by suppressing anti-apoptotic protein Mcl-1. Oncotarget 7, 18965–18977 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Molenaar RJ, Coelen RJS, Khurshed M, Roos E, Caan MWA, van Linde ME, Kouwenhoven M, Bramer JAM, Bovee J, Mathot RA, Klumpen HJ, van Laarhoven HWM, van Noorden CJF, Vandertop WP, Gelderblom H, van Gulik TM, and Wilmink JW (2017) Study protocol of a phase IB/II clinical trial of metformin and chloroquine in patients with IDH1-mutated or IDH2-mutated solid tumours. BMJ Open 7, e014961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Molenaar RJ, van de Venne T, Weterman MJ, Mathot RA, Klumpen HJ, Richel DJ, and Wilmink JW (2018) A phase Ib study of everolimus combined with metformin for patients with advanced cancer. Invest New Drugs 36, 53–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Ramos-Penafiel CO, Martinez-Murillo C, Santoyo-Sanchez A, Jimenez-Ponce F, Rozen-Fuller E, Collazo-Jaloma J, Olarte-Carrillo I, and Martinez-Tovar A (2014) [Effect of metformin addition to an acute lymphoblastic leukemia chemotherapy treatment]. Rev Med Inst Mex Seguro Soc 52, 270–275 [PubMed] [Google Scholar]
  • 179.Trucco M, Barredo JC, Goldberg J, Leclerc GM, Hale GA, Gill J, Setty B, Smith T, Lush R, Lee JK, and Reed DR (2018) A phase I window, dose escalating and safety trial of metformin in combination with induction chemotherapy in relapsed refractory acute lymphoblastic leukemia: Metformin with induction chemotherapy of vincristine, dexamethasone, PEG-asparaginase, and doxorubicin. Pediatr Blood Cancer 65, e27224. [DOI] [PubMed] [Google Scholar]
  • 180.Sato A, Sunayama J, Okada M, Watanabe E, Seino S, Shibuya K, Suzuki K, Narita Y, Shibui S, Kayama T, and Kitanaka C (2012) Glioma-initiating cell elimination by metformin activation of FOXO3 via AMPK. Stem Cells Transl Med 1, 811–824 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Vara-Ciruelos D, Dandapani M, Russell FM, Grzes KM, Atrih A, Foretz M, Viollet B, Lamont DJ, Cantrell DA, and Hardie DG (2019) Phenformin, But Not Metformin, Delays Development of T Cell Acute Lymphoblastic Leukemia/Lymphoma via Cell-Autonomous AMPK Activation. Cell Rep 27, 690–698 e694 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Carmignani M, Volpe AR, Aldea M, Soritau O, Irimie A, Florian IS, Tomuleasa C, Baritchii A, Petrushev B, Crisan G, and Valle G (2014) Glioblastoma stem cells: a new target for metformin and arsenic trioxide. J Biol Regul Homeost Agents 28, 1–15 [PubMed] [Google Scholar]
  • 183.Tsoli M, Chang C, Shen H, Liu J, Chintranjan A, Franshaw L, Dilda P, Carcaboso AM, Hogg P, and Ziegler D (2016) HG-19: COMBINED TARGETING OF MITOCHONDRIAL FUNCTION AND mTOR IS A POTENT NOVEL THERAPEUTIC APPROACH FOR DIFFUSE INTRINSIC PONTINE GLIOMA. Neuro-Oncology 18, iii51–iii51 [Google Scholar]
  • 184.Luk PP, Chung SA, Shen H, Decollogne S, Dilda PJ, McDonald KL, and Hogg PJ (2012) Abstract 1131: Blocking ATP delivery to hexokinase II in glioblastoma is a promising therapeutic strategy. Cancer Research 72, 1131–1131 [Google Scholar]
  • 185.Dilda PJ, Perrone GG, Philp A, Lock RB, Dawes IW, and Hogg PJ (2008) Insight into the selectivity of arsenic trioxide for acute promyelocytic leukemia cells by characterizing Saccharomyces cerevisiae deletion strains that are sensitive or resistant to the metalloid. Int J Biochem Cell Biol 40, 1016–1029 [DOI] [PubMed] [Google Scholar]
  • 186.Jue TJ, Chung SA, Rapkins RW, Dilda P, Hogg P, and McDonald KL (2017) P08.22 Targeting glioblastoma mitochondrial metabolism to inhibit cell proliferation & tumor growth. Neuro-Oncology 19, iii59–iii59 [Google Scholar]
  • 187.Tran B, Hamilton AL, Horvath L, Lam M, Savas PS, Grimison PS, Whittle JR, Kuo JC-Y, Signal N, Edmonds D, Hogg PJ, Rischin D, and Desai J (2016) First-in-man trial of 4-(N-(S-penicillaminylacetyl)amino) phenylarsonous acid (PENAO) as a continuous intravenous infusion (CIVI), in patients (pt) with advanced solid tumours. Journal of Clinical Oncology 34, e14025–e14025 [Google Scholar]
  • 188.Samudio I, Harmancey R, Fiegl M, Kantarjian H, Konopleva M, Korchin B, Kaluarachchi K, Bornmann W, Duvvuri S, Taegtmeyer H, and Andreeff M (2010) Pharmacologic inhibition of fatty acid oxidation sensitizes human leukemia cells to apoptosis induction. J Clin Invest 120, 142–156 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Estan MC, Calvino E, Calvo S, Guillen-Guio B, Boyano-Adanez Mdel C, de Blas E, Rial E, and Aller P (2014) Apoptotic efficacy of etomoxir in human acute myeloid leukemia cells. Cooperation with arsenic trioxide and glycolytic inhibitors, and regulation by oxidative stress and protein kinase activities. PLoS One 9, e115250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Galicia-Vazquez G, and Aloyz R (2018) Ibrutinib Resistance Is Reduced by an Inhibitor of Fatty Acid Oxidation in Primary CLL Lymphocytes. Front Oncol 8, 411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Divakaruni AS, Hsieh WY, Minarrieta L, Duong TN, Kim KKO, Desousa BR, Andreyev AY, Bowman CE, Caradonna K, Dranka BP, Ferrick DA, Liesa M, Stiles L, Rogers GW, Braas D, Ciaraldi TP, Wolfgang MJ, Sparwasser T, Berod L, Bensinger SJ, and Murphy AN (2018) Etomoxir Inhibits Macrophage Polarization by Disrupting CoA Homeostasis. Cell Metab 28, 490–503 e497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Lin H, Patel S, Affleck VS, Wilson I, Turnbull DM, Joshi AR, Maxwell R, and Stoll EA (2017) Fatty acid oxidation is required for the respiration and proliferation of malignant glioma cells. Neuro Oncol 19, 43–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Lee EA, Angka L, Rota SG, Hanlon T, Mitchell A, Hurren R, Wang XM, Gronda M, Boyaci E, Bojko B, Minden M, Sriskanthadevan S, Datti A, Wrana JL, Edginton A, Pawliszyn J, Joseph JW, Quadrilatero J, Schimmer AD, and Spagnuolo PA (2015) Targeting Mitochondria with Avocatin B Induces Selective Leukemia Cell Death. Cancer Res 75, 2478–2488 [DOI] [PubMed] [Google Scholar]
  • 194.Liu PP, Liu J, Jiang WQ, Carew JS, Ogasawara MA, Pelicano H, Croce CM, Estrov Z, Xu RH, Keating MJ, and Huang P (2016) Elimination of chronic lymphocytic leukemia cells in stromal microenvironment by targeting CPT with an antiangina drug perhexiline. Oncogene 35, 5663–5673 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Gugiatti E, Tenca C, Ravera S, Fabbi M, Ghiotto F, Mazzarello AN, Bagnara D, Reverberi D, Zarcone D, Cutrona G, Ibatici A, Ciccone E, Darzynkiewicz Z, Fais F, and Bruno S (2018) A reversible carnitine palmitoyltransferase (CPT1) inhibitor offsets the proliferation of chronic lymphocytic leukemia cells. Haematologica 103, e531–e536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Grande S, Palma A, Ricci-Vitiani L, Luciani AM, Buccarelli M, Biffoni M, Molinari A, Calcabrini A, D’Amore E, Guidoni L, Pallini R, Viti V, and Rosi A (2018) Metabolic Heterogeneity Evidenced by MRS among Patient-Derived Glioblastoma Multiforme Stem-Like Cells Accounts for Cell Clustering and Different Responses to Drugs. Stem Cells Int 2018, 3292704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Jones JE, Esler WP, Patel R, Lanba A, Vera NB, Pfefferkorn JA, and Vernochet C (2017) Inhibition of Acetyl-CoA Carboxylase 1 (ACC1) and 2 (ACC2) Reduces Proliferation and De Novo Lipogenesis of EGFRvIII Human Glioblastoma Cells. PLoS One 12, e0169566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Jones KD, Couldwell WT, Hinton DR, Su Y, He S, Anker L, and Law RE (1994) Lovastatin induces growth inhibition and apoptosis in human malignant glioma cells. Biochem Biophys Res Commun 205, 1681–1687 [DOI] [PubMed] [Google Scholar]
  • 199.Kikuchi T, Nagata Y, and Abe T (1997) In vitro and in vivo antiproliferative effects of simvastatin, an HMG-CoA reductase inhibitor, on human glioma cells. J Neurooncol 34, 233–239 [DOI] [PubMed] [Google Scholar]
  • 200.Wong WW, Dimitroulakos J, Minden MD, and Penn LZ (2002) HMG-CoA reductase inhibitors and the malignant cell: the statin family of drugs as triggers of tumor-specific apoptosis. Leukemia 16, 508–519 [DOI] [PubMed] [Google Scholar]
  • 201.Vitols S, Angelin B, and Juliusson G (1997) Simvastatin impairs mitogen-induced proliferation of malignant B-lymphocytes from humans--in vitro and in vivo studies. Lipids 32, 255–262 [DOI] [PubMed] [Google Scholar]
  • 202.Wang X, Huang Z, Wu Q, Prager BC, Mack SC, Yang K, Kim LJY, Gimple RC, Shi Y, Lai S, Xie Q, Miller TE, Hubert CG, Song A, Dong Z, Zhou W, Fang X, Zhu Z, Mahadev V, Bao S, and Rich JN (2017) MYC-Regulated Mevalonate Metabolism Maintains Brain Tumor-Initiating Cells. Cancer Res 77, 4947–4960 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Hamadani M, Awan FT, and Devine SM (2008) The impact of HMG-CoA reductase inhibition on the incidence and severity of graft-versus-host disease in patients with acute leukemia undergoing allogeneic transplantation. Blood 111, 3901–3902 [DOI] [PubMed] [Google Scholar]
  • 204.Advani AS, McDonough S, Copelan E, Willman C, Mulford DA, List AF, Sekeres MA, Othus M, and Appelbaum FR (2014) SWOG0919: a Phase 2 study of idarubicin and cytarabine in combination with pravastatin for relapsed acute myeloid leukaemia. Br J Haematol 167, 233–237 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Shadman M, Mawad R, Dean C, Chen TL, Shannon-Dorcy K, Sandhu V, Hendrie PC, Scott BL, Walter RB, Becker PS, Pagel JM, and Estey EH (2015) Idarubicin, cytarabine, and pravastatin as induction therapy for untreated acute myeloid leukemia and high-risk myelodysplastic syndrome. Am J Hematol 90, 483–486 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Tapia-Perez JH, Kirches E, Mawrin C, Firsching R, and Schneider T (2011) Cytotoxic effect of different statins and thiazolidinediones on malignant glioma cells. Cancer Chemother Pharmacol 67, 1193–1201 [DOI] [PubMed] [Google Scholar]
  • 207.Larner J, Jane J, Laws E, Packer R, Myers C, and Shaffrey M (1998) A phase I-II trial of lovastatin for anaplastic astrocytoma and glioblastoma multiforme. Am J Clin Oncol 21, 579–583 [DOI] [PubMed] [Google Scholar]
  • 208.Thibault A, Samid D, Tompkins AC, Figg WD, Cooper MR, Hohl RJ, Trepel J, Liang B, Patronas N, Venzon DJ, Reed E, and Myers CE (1996) Phase I study of lovastatin, an inhibitor of the mevalonate pathway, in patients with cancer. Clin Cancer Res 2, 483–491 [PubMed] [Google Scholar]
  • 209.Altwairgi AK, Raja S, Manzoor M, Aldandan S, Alsaeed E, Balbaid A, Alhussain H, Orz Y, Lary A, and Alsharm AA (2017) Management and treatment recommendations for World Health Organization Grade III and IV gliomas. Int J Health Sci (Qassim) 11, 54–62 [PMC free article] [PubMed] [Google Scholar]
  • 210.Happold C, Gorlia T, Nabors LB, Erridge SC, Reardon DA, Hicking C, Picard M, Stupp R, Weller M,Group, E. B. T, on behalf of the, C., and Groups, C. C. T. (2018) Do statins, ACE inhibitors or sartans improve outcome in primary glioblastoma? J Neurooncol 138, 163–171 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES