Next Article in Journal
Machine-Learning-Based Survival Prediction in Castration-Resistant Prostate Cancer: A Multi-Model Analysis Using a Comprehensive Clinical Dataset
Previous Article in Journal
The CORTEX Project: A Pre–Post Randomized Controlled Feasibility Trial Evaluating the Efficacy of a Computerized Cognitive Remediation Therapy Program for Adult Inpatients with Anorexia Nervosa
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Metabolic Signature of FLT3-Mutated AML: Clinical and Therapeutic Implications

by
Cristina Banella
1,
Gianfranco Catalano
2,3,
Maura Calvani
1,
Eleonora Candi
4,
Nelida Ines Noguera
2,3,* and
Serena Travaglini
4,*
1
Department of Pediatric Hematology-Oncology, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy
2
Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
3
Santa Lucia Foundation, I.R.C.C.S., Neuro-Oncohematology, 00143 Rome, Italy
4
Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
*
Authors to whom correspondence should be addressed.
J. Pers. Med. 2025, 15(9), 431; https://doi.org/10.3390/jpm15090431
Submission received: 26 June 2025 / Revised: 9 August 2025 / Accepted: 29 August 2025 / Published: 8 September 2025
(This article belongs to the Special Issue Acute Myeloid Leukemia: Current Progress and Future Directions)

Abstract

Acute Myeloid Leukemia (AML) is a genetically and clinically heterogeneous malignancy marked by poor prognosis and limited therapeutic options, especially in older patients. While conventional treatments such as the “7 + 3” chemotherapy regimen and allogeneic stem cell transplantation remain standard care options, the advent of next-generation sequencing (NGS) has transformed our understanding of AML’s molecular complexity. Among the emerging hallmarks of AML, metabolic reprogramming has gained increasing attention for its role in supporting leukemic cell proliferation, survival, and therapy resistance. Distinct AML subtypes—shaped by specific genetic alterations, including FLT3, NPM1, and IDH mutations—exhibit unique metabolic phenotypes that reflect their underlying molecular landscapes. Notably, FLT3-ITD mutations are associated with enhanced reactive oxygen species (ROS) production and altered energy metabolism, contributing to disease aggressiveness and poor clinical outcomes. This review highlights the interplay between metabolic plasticity and genetic heterogeneity in AML, with a particular focus on FLT3-driven metabolic rewiring. We discuss recent insights into how these metabolic dependencies may be exploited therapeutically, offering a rationale for the development of metabolism-targeted strategies in the treatment of FLT3-mutated AML.

1. Introduction

Acute Myeloid Leukemia (AML) is a highly heterogeneous and aggressive hematologic malignancy that primarily affects older adults [1,2]. Despite recent advances in treatment, AML remains a challenging disease with a poor prognosis, particularly in older adults, with a five-year survival rate below 30% [3]. For many years, the “7 + 3” chemotherapy regimen, consisting of cytarabine and anthracycline, followed by allogeneic stem cell transplantation, has been the standard of care for high-risk AML patients, irrespective of their clinical and molecular characteristics [4]. Recent advancements in next-generation sequencing (NGS) technologies have significantly enhanced our understanding of the molecular landscape of AML [5]. These breakthroughs have facilitated the identification of novel genetic aberrations and altered signaling pathways which hold promise as potential prognostic biomarkers and therapeutic targets. In light of this, significant progress has been made toward achieving higher complete remission (CR) rates, longer relapse-free survival (RFS), and better overall survival (OS) in AML [6].
Among emerging biomarkers, metabolic reprogramming represents one of the major hallmarks of cancer, playing a crucial role in cancer cell growth, migration, and metastatic progression [7]. Since Otto Warburg’s publication in 1920s proposing that cancer cells preferentially use glucose over mitochondrial respiration to generate ATP regardless of oxygen conditions [8,9], numerous studies have revealed the dynamic nature of cancer cell metabolism [10]. These investigations have identified a wide spectrum of metabolic alterations, each associated with distinct cancer types and disease stages. This metabolic diversity underscores the complexity of cancer reprograming, highlighting how these shifts might contribute not only to tumor growth but also to the heterogeneity observed among different tumors and their therapeutic responses [11,12,13].
Metabolic plasticity also plays a pivotal role in leukemia initiation and progression [14]. In AML, genetic and molecular heterogeneity is reflected in the preferential metabolic pathways used by different AML subtypes, shaped by their distinct molecular backgrounds. Indeed, numerous studies have shown that specific mutations and molecular signatures can activate oncogenic signaling pathways, directly contributing to these metabolic adaptations, which are crucial for meeting the energy demands and supporting the proliferation and survival of leukemic cells. Notably, an increased production of reactive oxygen species (ROS) has been linked to the aggressive behavior and poor prognosis of FLT3-ITD-mutated AML [15]. Beyond FLT3 mutations, other recurrent genetic alterations, such as NPM1, IDH1, and IDH2 and balanced translocation [16,17,18], including t(8;21)(q22;q22), t(15;17)(q24;q21), and t(16;16)(p13;q24) [19,20,21,22,23], profoundly influence the metabolic profile of leukemic cells. Notably, IDH mutations provide evidence of the strong connection between leukemogenesis and metabolism, playing a crucial role in the reprogramming of epigenetic, transcriptional, and biochemical profiles of AML cells [18,24]. This metabolic heterogeneity poses a significant challenge in the context of AML therapy, highlighting the necessity of gaining deeper insight into cancer metabolism for the development of treatment strategies directed toward specific metabolic pathways.
Herein, we provide an in-depth exploration of the role of cellular metabolism in AML, with a particular focus on recent discoveries regarding metabolic alterations associated with FLT3 mutations. We extensively discuss how these mutations impact cellular energy metabolism, highlighting their influence on leukemic cell survival and proliferation. By examining the latest advances in this field, we aim to elucidate the therapeutic implications of targeting distinct metabolic vulnerabilities associated with FLT3-mutated AML. A comprehensive understanding of the metabolic preferences driven by FLT3 mutations may uncover critical dependencies, paving the way for the development of more effective and subtype-specific treatment strategies [25,26,27,28,29,30].

2. Clinical and Molecular Features of FLT3-Mutated AML

AML is a complex and rapidly progressing malignancy, where the prognosis and treatment outcome are significantly influenced by the underlying genetic and molecular features. Recent advances in molecular biology, with the widespread use of NGS technologies, have uncovered crucial genetic alterations, allowing for an integrated approach that takes into account clinical and laboratory characteristics. This integrated approach has led to improved prognostic stratification and treatment personalization.
Among the most frequently mutated genes in AML, somatic alterations of the FLT3 gene are recognized as a distinct nosological entity, identified in approximately 25–30% of de novo AML cases. Accordingly, the European LeukemiaNET (ELN) guidelines recommend FLT3 testing at the time of initial AML diagnosis to guide treatment decisions and assess patient eligibility for allogeneic hematopoietic stem cell transplantation (allo-HSCT). According to the latest update of the ELN 2022 recommendations, two different types of FLT3 mutations have been considered clinically relevant: internal tandem duplications (ITD) in the juxtamembrane domain (JM) of the gene and point mutations within the tyrosine kinase domain (TKD) [31,32]. In addition, rare deletions and point mutations affecting the JM domain have also been described, but the biological and prognostic significance of these alterations is still unclear [33,34].
FLT3 abnormalities, particularly the ITD variant, are associated with a high incidence of relapse and aggressive disease progression, making it a critical target for therapeutic intervention in AML management [35,36,37].
Amidst the ongoing advances in the molecular characterization and prognostic stratification of AML patients, the metabolic profile of leukemic cells remains largely overlooked. While the emerging role of metabolism in tumor initiation, progression, and response to therapy has been increasingly recognized, the precise contribution of these cellular metabolic networks to key cancer cell functions is far from being fully understood.

3. Metabolic Dependencies in FLT3-Mutated AML

Hematopoietic stem cells (HSCs), referred to as ‘dormant’ (dHSCs), exhibit low metabolic demands within the bone marrow niche. In their dormant state, HSCs primarily rely on aerobic glycolysis (AG) to maintain low levels of ROS and energy production. However, upon activation, they shift toward oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO) to meet increased energy demands [38,39,40]. Similarly, AML cells, particularly those harboring FLT3 mutations, modulate their metabolic pathways dynamically in response to nutrient availability, microenvironmental stress, and therapeutic pressure (Figure 1) [41,42]. This metabolic flexibility allows cancer cells to sustain proliferation and survival under fluctuating conditions, such as hypoxia or drug exposure, by modulating glycolysis, mitochondrial respiration, and lipid metabolism accordingly [43,44,45,46,47,48]. Therefore, the metabolic features described should be considered adaptable responses rather than fixed characteristics, playing a key role in disease progression and resistance to treatment.

3.1. Oxidative Phosphorylation (OXPHOS)

Mitochondria serve as cellular metabolic hubs within leukemic cells, where carbohydrate, amino acid, and fatty acid pathways converge in the tricarboxylic acid (TCA) cycle. (Figure 2) [49]. The key metabolic processes orchestrated by these organelles, including TCA cycle activity, glutaminolysis, oxidative phosphorylation, and fatty acid oxidation, are profoundly altered in AML [50].
Normal hematopoietic stem cells (HSCs) primarily rely on glycolysis for energy production, whereas leukemia stem cells (LSCs) depend heavily on oxidative phosphorylation (OXPHOS) for their biosynthetic processes and survival [51,52]. This distinction highlights OXPHOS as a potential novel target in AML. In particular, FLT3-mutated AML cells exhibit unique metabolic flexibility by rapidly activating OXPHOS upon oxygen availability, which is crucial for their survival in a fluctuating hypoxic and normoxic environment within the bone marrow [53,54,55]. The metabolic adaptability of leukemic cells, as they switch from anaerobic glycolysis to OXPHOS, enables efficient energy production and helps maintain mitochondrial outer membrane impermeability, which is essential for cell survival [56]. Such metabolic plasticity helps to develop resistance to chemotherapy. On the other end, these cells demonstrate a low spare respiratory capacity compared to their normal counterpart, suggesting that targeting the OXPHOS chain could be a promising therapeutic strategy for this specific patient subset [13,14,57]. Notably, OXPHOS and purine synthesis are central metabolic pathways targeted by different synthetic lethal treatments to resensitize leukemic cells to TKI treatment [58]. Indeed, recent studies in sorafenib-resistant FLT3-ITD cell lines have revealed that resistant leukemic cells undergo metabolic reprogramming characterized by impaired mitochondrial OXPHOS and a compensatory upregulation of glycolysis. This metabolic shift supports cell survival under sustained TKI pressure despite mitochondrial dysfunction [59,60]. Importantly, these resistant cells demonstrate collateral sensitivity to glycolytic inhibitors such as 2-deoxyglucose (2-DG) and 3-bromopyruvate propylester (3-BrOP), which target key glycolytic enzymes including hexokinase 2, highly expressed in the mitochondrial fraction of resistant cells [60,61,62].
Exploiting this metabolic vulnerability through synthetic lethal strategies, combining FLT3 tyrosine kinase inhibitors with glycolytic blockade, has been shown to resensitize resistant leukemic cells to TKI treatment, overcoming resistance mechanisms and promoting apoptotic cell death more effectively. [61,63,64] Furthermore, targeting mitochondrial oxidative phosphorylation (OXPHOS) and purine synthesis pathways alongside FLT3 inhibition has emerged as a promising complementary approach to enhance therapeutic efficacy [59]. Collectively, these findings underscore the critical role of metabolic rewiring in TKI resistance and provide a strong rationale for combined therapeutic strategies targeting both oncogenic signaling and metabolic dependencies to achieve durable responses in FLT3-mutated AML.

3.2. Aerobic Glycolysis

Beyond bioenergetics, glucose metabolism encompasses multiple pathways, including the pentose phosphate pathway, serine biosynthesis, and one-carbon metabolism, collectively generating essential molecular precursors [65]. AML cells demonstrate elevated GLUT1 expression, correlating with increased glucose uptake, while heightened pyruvate and lactate concentrations in patient sera are associated with unfavorable prognosis [66]. Over a decade ago, in mouse models, Ying-Hua Wang and colleagues established that genetic ablation of glycolytic enzymes PKM2 or LDHA disrupts aerobic glycolytic flux and attenuates leukemic expansion, whereas normal hematopoietic stem cells demonstrate tolerance to depletion of either enzyme. Paradoxically, normal cellular compartments showed increased proliferative capacity upon PKM2 or LDHA deletion [67]. Thus, the glycolytic pathway is important for leukemia maintenance and progression, and leukemic cells are more sensitive to the inhibition of AG than normal hematopoietic cells. AG remains fundamental in tumor proliferation, metastasis, and therapeutic resistance through multifaceted regulation of glycolytic enzymes, signaling cascades, non-coding RNAs, and bone marrow microenvironmental factors.
FLT3-mutated AMLs exhibit heightened glycolytic activity [68]. Despite increased lactate production, this does necessarily reduce mitochondrial pyruvate metabolism, which remains crucial for energy production in these cells [69,70]. FLT3-mutated AML cells display a unique metabolic profile characterized by low levels of pyruvate dehydrogenase kinase 1 (PDK1), which is involved in deactivating pyruvate dehydrogenase (PDH), a critical mitochondrial multienzyme complex responsible for catalyzing the oxidative decarboxylation of pyruvate, with this expression pattern correlating with heightened OXPHOS states. This metabolic profile is linked to a specific FLT3-mutated/PDP1 signaling axis that mediates pyruvate metabolism [71].
The increased glycolysis is regulated by key enzymes such as hexokinase 2 (HK2), which is involved in glucose phosphorylation and glycolytic pathway activation. FLT3 mutations lead to constitutive activation of glycolysis through the activation of MYC, enhancing FLT3 expression and inducing a positive feedback loop that further amplifies glycolytic metabolism [68]. In this scenario, the interaction between the first glycolytic enzyme HK2 and the voltage-dependent anion channel 1 (VDAC1) on the outer mitochondrial membrane facilitates the coupling of glycolysis and OXPHOS, significantly increasing ATP production and supporting the high energy demands of leukemic cells [72]. Targeting this glycolytic pathway may impair the metabolic function of FLT3-ITD AML cells and provide potential therapeutic avenue.
Emerging research has uncovered distinctive metabolic characteristics in FLT3-mutated AML, particularly their enhanced aerobic glycolytic metabolism compared to wild-type counterparts [73]. Targeted metabolic interventions reveal a remarkable therapeutic vulnerability: glycolysis inhibition demonstrates selective cytotoxicity against FLT3-ITD leukemic cells while sparing wild-type leukemia and normal cellular populations [74]. Pre-clinical investigations in a murine model explored the synergistic potential of combined therapeutic strategies, specifically integrating a glycolysis inhibitor (2-deoxy-D-glucose) with the multi-targeted tyrosine kinase inhibitor sorafenib. The combination therapy demonstrated significantly prolonged survival compared to monotherapy, with experimental mice experiencing extended survival from 32 to 41 days.

3.3. Glutaminolysis

Standard amino acids support a wide range of cellular functions critical for neoplastic proliferation, including protein synthesis, nucleotide production, lipid biosynthesis, and glutathione generation. They also play key roles in the regulation of epigenetic mechanisms and post-translational modifications [75]. Glutamine represents the predominant amino acid supporting AML proliferation and survival through dual functionality in α-ketoglutarate provision for TCA cycling and leucine importation, facilitating mTORC1-mediated protein synthesis. Glutamine enters AML cells via SLC1A5 transporters before conversion to glutamate and α-ketoglutarate through glutaminolytic pathways, particularly clear in FLT3-mutated AML. Comparative analyses demonstrate heightened glutamine dependency in leukemic blasts versus normal HSCs, evidenced by resistance of normal HSCs to glutamine deprivation-induced apoptosis, contrasting with significant cell death in primary AML samples following SLC1A5 knockdown [76]. Furthermore, glutamine limitation substantially reduces oxygen consumption rates, suggesting critical interdependence between glutaminolysis and oxidative phosphorylation in leukemic metabolism [77].
Notably, glutamine metabolism emerges as another critical metabolic process in FLT3-mutated AML. Disrupting glutamine uptake, such as through the inhibition of SLC1A5, a transporter crucial for glutamine uptake, prevents mTORC1 activation and induces apoptosis, sparing normal HSCs. Experimental studies using shRNA-mediated inhibition of SLC1A5 showed antileukemic effects in xenograft mouse models, inducing autophagy and significantly reducing tumor growth [78]. Glutamine oxidation is particularly important for cells resistant to treatments like quizartinib, as these cells rely on mitochondrial metabolism for survival.
Mitochondrial glutamine catabolism represents a fundamental metabolic vulnerability in FLT3-ITD AML cells that exhibit therapeutic resilience to quizartinib [79]. These treatment-refractory malignant clones demonstrate pronounced reliance on glutaminolytic pathways to sustain bioenergetic homeostasis and biosynthetic processes, underscoring their dependency on oxidative phosphorylation machinery [79]. Compelling evidence reveals remarkable therapeutic synergy when quizartinib is paired with L-asparaginase, an amidohydrolase that depletes circulating glutamine and asparagine pools. This combinatorial strategy substantially attenuates glutamine catabolism in surviving leukemic populations, diminishing their capacity for metabolic persistence following FLT3 pathway blockade. Mechanistically, FLT3-ITD oncogenic signaling orchestrates upregulation of pyruvate dehydrogenase phosphatase 1 (PDP1), a key regulatory component of the pyruvate dehydrogenase multienzyme complex, through RAS-mediated transcriptional cascades. Elevated PDP1 expression augments both glycolytic flux and glutamine utilization, conferring metabolic flexibility that enables resistance to FLT3 inhibition. Notably, genetic ablation of PDP1 restores cellular sensitivity to quizartinib even under oxygen-limited conditions, establishing PDP1 as a critical metabolic rheostat in FLT3-mutant leukemogenesis. The therapeutic rationale for quizartinib-L-asparaginase combination therapy extends beyond simple glutamine depletion, encompassing disruption of the enhanced oxidative metabolism that characterizes resistant cell populations. By simultaneously impairing mitochondrial amino acid oxidation and PDP1-driven metabolic reprogramming, this anti-metabolic intervention may eliminate minimal residual disease and augment treatment efficacy [80]. Indeed, the combination of quizartinib (AC220) with CB-839, a GLS inhibitor, leads to glutathione depletion, mitochondrial ROS accumulation, apoptotic cell death in vitro, and significantly extended survival in FLT3-ITD patient-derived xenograft models compared to FLT3 inhibition alone. CB-839 (telaglenastat), a highly selective and orally bioavailable GLS inhibitor, has shown promising tolerability in early-phase clinical trials across hematologic malignancies including AML, with mostly low-grade adverse events reported and an acceptable safety profile even in combination regimens [81]. By contrast, non-selective glutamine antagonists such as DON or acivicin historically induced dose-limiting gastrointestinal, neurological, and hematopoietic toxicities due to broad enzyme targeting [82,83]. Importantly, because immune effector cells (e.g., T cells, NK cells, macrophages) also rely on glutamine, GLS inhibition may carry the risk of immunosuppressive sequelae [82] (NCT02071927). Finally, the high metabolic plasticity of AML cells, encompassing upregulation of alternative substrate transporters or compensatory pathways like fatty acid oxidation or micropinocytosis, could limit long-term efficacy of glutaminase targeting, emphasizing the need for rationally designed combination regimens that balance metabolic targeting, antileukemic potency, and tolerability.

3.4. Fatty Acid Oxidation (FAO)

Lipids constitute an energetic substrate for neoplastic cells, not merely serving as energy reservoirs but providing essential components for membrane biogenesis and signaling pathway regulation [84,85]. AML blasts exhibit characteristic dysregulation of lipid metabolism compared with normal hematopoietic cells, presenting therapeutic targeting opportunities [86]. Comprehensive lipidomic profiling of plasma from AML patients at diagnosis revealed a striking pattern—there is a global reduction in total fatty acids and cholesterol, yet a selective elevation in certain free fatty acid species. These observations strongly imply a metabolic shift toward enhanced FAO as an adaptive response in AML metabolism [87]. In particular, the plasma of AML patients showed significantly increased levels of arachidonic acid (20:4 n − 6) and its precursors, such as gamma-linolenic acid (18:3 n − 6) and eicosatrienoic acid (20:3 n − 6), especially in individuals presenting with high bone marrow or peripheral blast counts and adverse prognostic risk [88]. These specific polyunsaturated fatty acids (PUFAs) are now recognized as metabolic biomarkers tightly associated with AML severity and metabolic reprogramming. Such selective enrichment of free fatty acids, despite overall plasma lipid depletion, underscores a proposed “futile cycle” in AML, whereby increased FAO provides acetyl-CoA to feed the TCA cycle and citrate pool, ultimately fueling de novo lipid synthesis required for rapid membrane biogenesis and proliferation. The upregulation of FAO-related transporters, such as carnitine transporter CT2 (SLC22A16), and enzymes like carnitine palmitoyltransferase I (CPT1a) further supports this metabolic rewiring as a pivotal survival strategy in AML cells [89]. This metabolic pathway supplies acetyl-CoA to the TCA cycle, subsequently increasing citrate production that initiates de novo fatty acid synthesis. FAO regulation plays a critical role in leukemic cell survival and quiescence, showing significant overexpression in AML cells compared to normal HSCs [90,91]. Quiescent HSCs maintain baseline FAO rates preserving dormancy, with metabolic status influencing symmetric versus asymmetric division outcomes and subsequent self-renewal capabilities [90,91].
It has been demonstrated that FLT3-mutated AML cells also showed increased fatty acid oxidation FAO rates, which play a pivotal role in supporting their metabolic needs [92]. Proteins such as CT2 and CPT1a are often upregulated, supporting this metabolic reliance. By promoting electron flux through the respiratory chain, agents like palmitate and dimethyl succinate can induce oxidative stress in FLT3-mutated cells, leading to cell death. This suggests that strategies promoting electron flux or targeting lipid metabolism, such as the PPARα agonist bezafibrate, known to increase mitochondrial mass and β-oxidation, may offer new therapeutic approaches for treating FLT3-mutated AML [93,94].

3.5. Sphingolipid Metabolism and Ceramide

Sphingolipids, including sphingomyelin (SM), ceramide (Cer), and glycosphingolipids, introduce another layer of metabolic complexity [95,96,97]. Sphingolipid formation and functionality depend upon oncogenic proteins, including sphingosine kinases and acid ceramidases, within AML cellular environments. Sphingosine-1-phosphate, generated through sphingosine kinase 1 activity, constitutively regulates AML cellular survival mechanisms [98]. Recent investigations demonstrate upregulation of S1PR3 (sphingosine-1-phosphate receptor 3) in both AML blasts and CD34+CD38- leukemic stem cells compared to normal hematopoietic stem cells. This receptor governs myeloid differentiation processes while activating inflammatory signaling cascades in primitive leukemic populations. Notably, S1PR3 activation in primary AML specimens promotes leukemic stem cell differentiation, potentially facilitating elimination of these therapy-resistant cellular reservoirs. Importantly, pharmacological activation of this receptor using the sphingosine-1-phosphate analog FTY720 (fingolimod) robustly reduced LSC frequency and leukemia burden in mice engrafted with patient-derived AML cells, including relapsed and chemoresistant samples. Strikingly, normal hematopoietic xenograft function was preserved, indicating a therapeutic window for selective targeting of LSCs. Therefore, harnessing S1PR3 signaling—either by genetic overexpression or via agonists such as FTY720—promises a strategy to deplete therapy-resistant leukemic reservoirs through enforced differentiation [99]. Moreover, FTY720 activates protein phosphatase 2A (PP2A) by disrupting SET–PP2A binding. In AML models, FTY720-mediated PP2A reactivation induces apoptosis and diminishes leukemic proliferation in a dose-dependent manner, with effects rescued by PP2A inhibition, indicating that the anticancer activity is PP2A-dependent [100,101]. Moreover, combining the CK2 inhibitor CX-4945 with FTY720 enhances antileukemic efficacy significantly. CX-4945 forces nuclear retention of SET, while FTY720 antagonizes SET–PP2A interaction in the cytoplasm, resulting in restored PP2A activity, reduced migration, and diminished cell viability and invasion in zebrafish AML xenografts—outperforming either agent alone [102].
Additionally, these lipid molecules not only just serve as structural components in cell membranes but also as potential regulators of cellular stress responses. Notably, Cer shows tumor-suppressing properties in many cancer types, and defects in Cer generation and clearance can lead to cancer cell survival and chemotherapy resistance [103,104]. The Cer transfer protein (CERT) transports Cer from the endoplasmic reticulum to the Golgi apparatus, playing a role in ceramide clearance and determining the ceramide-to-sphingomyelin ratio in cells. Inactivation of CERT has been shown to induce apoptosis in various cancer cell lines, including colon, breast, and lung carcinoma cells [105,106]. In FLT3-mutated AML, signaling pathways activated by FLT3 mutations suppress ceramide synthase 1 (CerS1) and Cer metabolism [107]. Pharmacological inhibition of sphingosine kinase 1 (SPHK1) using agents like MP-A08 restores Cer levels, triggering integrated stress responses and sensitizing FLT3-ITD AML cells to apoptosis—particularly when combined with venetoclax [108,109]. Equally compelling is the targeted inhibition of the ceramide transfer protein CERT. Both genetic knockdown and pharmacological inhibition with HPA-12 selectively induces Cer retention in FLT3-ITD AML cells, reducing viability and promoting apoptosis while sparing FLT3 wild-type cells [110]. Crucially, co-administration of HPA-12 with the FLT3 inhibitor crenolanib reveals a strong synergistic antileukemia effect, mediated through activation of the ER stress (GRP78/ATF6/CHOP axis) and induction of mitophagy [111,112]. This combination enhances Cer-driven apoptotic pathways and decreases leukemic stem cell reservoirs in both in vitro and in vivo models. Lipid accumulation within treated AML cells in different subcellular compartments suggests that lipids might induce other pro-cell death cascades, such as mitophagy [110]. Mitophagy has been reported to be involved in the inhibitory effect of crenolanib on FLT3-mutated AML cells. Another possibility is lipid stress-induced ferritin deficiency, particularly in FLT3-ITD AML. For instance, they can inhibit fatty acid uptake and oxidation, promoting intracellular lipid accumulation. In a recent study, fatty acid metabolism was inhibited through both genetic and pharmacological targeting of the FLT3–C/EBPα–SCD axis, including the use of APR-246 (eprenetapopt) to induce lipid peroxidation via glutathione depletion, and RSL3 to inhibit GPX4 and promote ferroptotic cell death. Although the specific SCD inhibitor was not disclosed, commonly used compounds such as A939572 or CAY10566 have been employed in similar contexts to suppress MUFA biosynthesis. Moreover, these interactions underscore the importance of ceramides as central nodes in lipid metabolism, affecting both the composition of cell membranes and cellular responses to variations in lipid levels [113]. These interactions underscore the importance of ceramides as central nodes in lipid metabolism, affecting both the composition of cell membranes and cellular responses to variations in lipid levels.

3.6. Anabolic Reprogramming in FLT3-ITD AML

Beyond conventional catabolic dependencies, FLT3-ITD-mutated AML demonstrates extensive metabolic reprogramming that encompasses amino acid utilization, one-carbon unit transfer reactions, and anabolic flux through the pentose phosphate pathway (PPP)—encompassing cellular processes fundamental to nucleotide biosynthesis, maintenance of redox homeostasis, and membrane component generation. Although specific investigations in FLT3-mutated disease remain sparse, comprehensive AML metabolomic analyses reveal enhanced expression of key enzymes including MTHFD2—a pivotal regulator of folate-mediated one-carbon transfer—alongside increased PPP activity, supporting cellular expansion under oncogenic conditions [114,115]. Additionally, l-type amino acid transporter 1 (LAT 1, SLC7A5) is frequently overexpressed, contributing to the uptake of branched-chain amino acids (BCAAs) and supporting oxidative respiration and anabolic growth. Notably, the selective LAT1 inhibitor JPH203 has demonstrated potent antileukemic activity in AML models, including samples resistant to venetoclax + azacitidine (Ven + Aza). Treatment with JPH203 impaired oxidative phosphorylation and viability in AML blasts, while sparing healthy hematopoietic cells; moreover, its combination with Ven + Aza produced synergistic eradication of leukemia activity in vitro [116].
The proliferative demands of AML cells necessitate sustained nucleotide and lipid production to fuel uncontrolled cellular division. These cells engage pentose phosphate pathway activation to produce ribose-5-phosphate precursors and generate NADPH, thereby enabling both de novo purine/pyrimidine synthesis and fatty acid production essential for membrane biogenesis. Recent evidence positions transketolase (TKT) as a central orchestrator of this metabolic rewiring: clinical specimens and established AML cell models exhibit substantially upregulated TKT levels, which facilitate cellular expansion, invasive capacity, and metabolic adaptation through transcriptional control of ribokinase (RBKS), creating a positive regulatory loop that amplifies non-oxidative pentose phosphate flux and promotes epithelial–mesenchymal transition phenotypes. In parallel, perturbation of guanine nucleotide biosynthesis, either through suppression of guanosine or pharmacological inhibition of Inosine Monophosphate Dehydrogenase 2 (IMPDH2), has been shown to induce myeloid differentiation and impair the oncogenic Lens Epithelium Derived Growth Factor (LEDGF)/menin/MLL-fusion complex in AML and MLL-rearranged leukemias. Notably, clinical translation of these insights includes the RNA polymerase I inhibitor CX-5461, which selectively downregulates menin and LEDGF, induces AML cell differentiation, and, critically, has demonstrated acceptable tolerability without neutropenia in early-phase hematologic trials [117].
Additionally, TKTL1, a transketolase paralog, mediates hypoxic adaptation in THP-1 leukemic cells through modulation of Glucose-6-Phosphate Dehydrogenase (G6PD) and Glyceraldehyde-3-Phosphate Dehydrogenase (GAPDH) enzymatic activity, maintaining both NADPH availability and glycolytic flux during environmental stress conditions [118]. Such anabolic dependencies present exploitable therapeutic windows: ribosomal assembly inhibitors and antifolate compounds that disrupt nucleotide availability have demonstrated promising activity in AML experimental systems and early clinical investigations.
Moreover, FLT3-ITD AML displays remarkable metabolic flexibility, deploying alternative nutrient acquisition mechanisms including bulk endocytosis (macropinocytosis), enhanced amino acid transporter expression, and metabolic shifts toward fatty acid oxidation when primary carbon sources become limiting [119]. This adaptive capacity undermines the sustained efficacy of single-pathway metabolic inhibitors. Notably, fatty acid oxidation-driven metabolic reprogramming contributes to resistance against venetoclax or FLT3-targeted agents, emphasizing the requirement for coordinated targeting of both catabolic and biosynthetic networks to prevent metabolic escape.
Collectively, therapeutic metabolic intervention in FLT3-ITD AML must extend beyond targeting glutamine catabolism or oxidative phosphorylation to encompass disruption of biosynthetic networks—encompassing nucleotide, membrane lipid, and amino acid production—through strategically designed combination approaches. This framework requires biomarker-guided patient selection, precision inhibitors targeting specific enzymes (such as MTHFD2, pentose phosphate regulators, or amino acid carriers), and optimized pharmacological delivery systems to achieve maximal antileukemic efficacy while preserving metabolic function in healthy proliferating tissues including immune effector cells.

3.7. ROS Dynamics in FLT3-Mutated AML

As discussed above, mitochondrial dependency recently emerged as a pivotal driver of energy in AML pathogenesis and progression. Notably, LSCs showed an increased mitochondrial mass and O2 consumption rate as compared to normal HSCs [120]. Indeed, OXPHOS dependency is closely linked to the increased production of mitochondrial reactive oxygen species (mtROS), which play a wide range of roles, including stimulating signaling pathways that promote tumorigenesis. Specifically, JNK, activated by ROS and endoplasmic reticulum stress (e.g., via IRE1α), is required for survival of FLT3-ITD leukemia cells, and its pharmacologic inhibition using SP600125 induces apoptosis even in TKI-resistant FLT3-ITD-TKD blasts [121]. While ERK is engaged simultaneously, ensuring complementary regulation of survival and proliferation pathways in FLT3-ITD AML, driven by RAS/MEK signaling downstream of FLT3-ITD, it promotes leukemic proliferation and metabolic remodeling and contributes to adaptive resistance following FLT3 inhibitor therapy [122]. Collectively, JNK/ERK, HIF1α, and associated regulatory networks orchestrate mitochondrial biogenesis and the regulation of protein function through various post-translational modifications [123]. During cellular metabolism, electrons derived from NADH and FADH2, which are products of substrate oxidation including glucose, glutamine, and fatty acids, are transferred through the ETC complexes, establishing a proton gradient essential for ATP synthesis [124,125]. During the process, however, either proton or electron leakage occurs. Proton and electron leak balance is intricately linked with superoxide production and cellular metabolism and resistance to stress. Electron leakage, predominantly occurring at Complex I (NADH dehydrogenase) [126] and Complex III (cytochrome bc1 complex), results in the partial reduction of molecular oxygen to form superoxide (O2) [127]. The generated superoxide is subsequently converted to hydrogen peroxide (H2O2) through the action of mitochondrial superoxide dismutase SOD1 in the intermembrane space and SOD2 in the matrix [128]. H2O2 can traverse membranes, functioning either as a signaling molecule or inducing oxidative damage to cellular components.
Previous investigations have established that FLT3 mutations contribute to oxidative damage through multiple mechanisms. One significant pathway involves increased intracellular reactive oxygen species (ROS) production in FLT3-mutated cells compared to their wild-type FLT3 counterparts, with NADPH oxidases (NOX family) localized in the endoplasmic reticulum and mitochondria serving as primary ROS sources [129]. While physiological ROS levels play crucial roles in cellular signaling and homeostasis, elevated concentrations rapidly react with biomolecules including proteins, lipids, carbohydrates, and nucleic acids, resulting in irreversible functional alterations or complete molecular destruction [130]. Additionally, FLT3 mutations hyperactivate the STAT5 and PI3K/AKT signaling cascades, thereby maintaining or enhancing the expression of p22phox and NOX proteins [131,132]. This upregulation promotes ROS production that subsequently diffuses into the nucleus, causing DNA damage—particularly double-strand breaks and mismatches. Additionally, the localization of FLT3 at the plasma membrane is essential for maintaining NOX protein levels and preventing the GSK3-β-mediated proteasomal degradation of p22phox. The functional mechanism involves NOX proteins associating with p22phox for membrane co-stabilization, followed by RAC1 binding and GDP-GTP exchange, which initiates oxygen conversion to superoxide [15].
Notably, FLT3-mutated cells exhibit increased RAC1-GTP binding to phosphorylated STAT5, resulting in enhanced recruitment of RAC1-GTP to NADPH oxidase complexes. Of particular significance is the nuclear membrane-bound NOX4D isoform, which is highly expressed in FLT3-ITD-positive patients and cell lines but nearly absent in wild-type FLT3-expressing counterparts. This isoform generates ROS that promote leukemic cell survival. A feedback loop exists between FLT3 signaling and ROS production, creating a self-reinforcing cycle. The oxidative environment enhances both wild-type FLT3 and FLT3-ITD signaling, possibly through oxidation of specific cysteine residues such as Cys790, which, in turn, further increases ROS production [133].
Recent research by Wu and colleagues demonstrated that in FLT3-mutated AML, FLT3 signaling upregulates key DNA damage response (DDR) factors through distinct pathways [134]. Through STAT5 activation, FLT3-ITD enhances expression of Wee1-like protein kinase (WEE1), checkpoint kinase 1 (CHK1), and proviral integration site for Moloney murine leukemia virus-1 (PIM-1). Concurrently, via ERK pathway activation, FLT3-ITD increases expression of radiation sensitive 51 (RAD51) and mismatch repair (MMR) factors including MutS homolog 2 (MSH2), MSH6, and MutL homolog 1 (MLH1). These adaptive responses promote AML cell survival under oxidative stress conditions and contribute to chemoresistance. Metabolically, FLT3-ITD-expressing cells exhibit distinctive characteristics, including elevated expression of succinate-CoA ligases and enhanced mitochondrial electron transport chain (ETC) complex II activity. This heightened respiratory capacity correlates with increased mitochondrial metabolism and consequent ROS production. Indeed, FLT3 mutations drive specific gene expression signatures [135], and FLT3-mutated AMLs typically display increased ROS levels, leading to enhanced DNA double-strand breaks [136]. Moreover, inhibition of FLT3-ITD creates a specific dependency on glutaminolysis for cellular survival [137].
Recent comprehensive multi-omics analyses by Erdem et al. have stratified AML based on pyruvate dehydrogenase kinase 1 (PDK1) expression levels [71]. PDK1 was identified as a critical determinant of distinct metabolic states in AML. PDK1-high AMLs exhibit reduced mitochondrial oxidative phosphorylation and frequently retain wild-type FLT3 and NPM1, also displaying enriched stemness signatures. Conversely, PDK1-low AMLs commonly harbor FLT3 mutations and are characterized by elevated cell cycle activity, enhanced oxidative phosphorylation, L-GMP (lymphoid-primed multipotent progenitor) signatures, and the highest oxygen consumption rates.
Kannan et al. proposed targeting the NRF2/HO-1 antioxidant pathway in FLT3-mutated AML to enhance therapeutic efficacy [138]. In non-FLT3 mutant AML, heme oxygenase-1 (HO-1) contributes to resistance against tumor necrosis factor (TNF)-induced apoptosis and epigenetically targeted agents [139]. The transcription factor NRF2, a major driver of HO-1 expression, has also been implicated in drug resistance in AML [140]. While numerous clinical studies have targeted NRF2 across various cancer types over the past three decades, none have specifically focused on FLT3-mutant AML [141,142], despite evidence suggesting that targeting NRF2 and antioxidant pathways may deplete leukemic stem cells (LSCs) [143,144]. Furthermore, there remains a need for reliable biomarkers of NRF2 inhibition across cancer types, with recent data suggesting that HO-1 expression or downstream redox parameters such as ROS levels or glutathione (GSH/GSSG) ratios may serve as useful indicators in FLT3-mutated AML.

3.8. Metabolic Targeting: Balancing Antileukemic Efficacy and Normal Cell Function

A critical challenge in metabolic targeting is posed by cancer stem cells (CSCs), a rare and intrinsically resistant subpopulation first identified in AML and later in various malignancies that sustains long-term tumor propagation through self-renewal and multilineage differentiation. While driving both drug resistance and therapy tolerance. These dual properties are driven by mechanisms such as quiescence-associated drug insensitivity, activation of survival pathways, and evasion of apoptosis [61,63,64,145,146]. Importantly, CSCs exhibit cell cycle heterogeneity and may persist as either resistant or tolerant clones under therapeutic pressure, complicating efforts to eradicate the disease through conventional or metabolic interventions.
This challenge is further deepened by the fact that canonical metabolic reprogramming events including enhanced glycolytic flux, augmented oxidative phosphorylation capacity, and glutamine catabolism are not restricted to malignant cells but represent conserved features of non-malignant proliferative cellular states, including activated T lymphocytes, tissue-resident macrophages, and other immune effector populations. Upon antigenic stimulation, T cells rapidly upregulate GLUT1-mediated glucose transport and ASCT2-dependent glutamine uptake, engage aerobic glycolysis concurrent with mitochondrial respiration, and activate mTORC1 signaling to satisfy biosynthetic and bioenergetic requirements—metabolic signatures that mirror those observed in transformed cells (including Warburg-type glycolysis and glutamine-fueled anaplerosis). Consequently, pharmacological disruption of glutamine metabolism or glycolytic pathways may compromise the activation kinetics, proliferative capacity, or effector functions of essential immune cell subsets, including CD8+ T cells, NK cells, B lymphocytes, and M1 macrophages, which exhibit obligate glutamine dependence for cytokine biosynthesis and antimicrobial responses [147,148,149].
From a translational perspective, broad-spectrum glutamine analogs such as 6-diazo-5-oxo-L-norleucine (DON) or L-(αS,5S)-α-amino-3-chloro-4,5-dihydro-5-isoxazoleacetic acid (acivicin)—which inhibit multiple glutamine-utilizing enzymes—have demonstrated dose-limiting toxicities including severe enterocolitis, peripheral neuropathy, and myelosuppression, reflecting their indiscriminate targeting of glutamine-dependent processes in rapidly dividing normal tissues [150,151]. While selective allosteric glutaminase (GLS1) inhibitors such as CB-839 (telaglenastat) and bis-2-(5-phenylacetamido-1,3,4-thiadiazol-2-yl)ethyl sulfide (BPTES) exhibit enhanced tumor selectivity and improved therapeutic windows, the metabolic glutamine addiction of immune effector cells poses inherent risks of treatment-related immunosuppression, particularly under conditions of prolonged GLS1 blockade [152,153]. Furthermore, the metabolic adaptability characteristic of AML blasts, including compensatory activation of alternative nutrient scavenging mechanisms (macropinocytosis, upregulation of amino acid transporters such as LAT1/SLC7A5) or metabolic rewiring toward fatty acid β-oxidation, presents significant challenges for durable therapeutic responses to single-agent metabolic inhibitors [154]. These considerations underscore the imperative for rational combination strategies or tumor-selective delivery platforms designed to maximize on-target antileukemic activity while preserving physiological metabolic functions in normal proliferative compartments.
In summary, effective metabolic intervention in FLT3-ITD AML necessitates comprehensive evaluation of potential off-target effects in normal proliferative tissues, particularly immune surveillance mechanisms, and implementation of precision medicine approaches incorporating biomarker-driven patient stratification, target-selective inhibition, pharmacokinetically optimized dosing regimens, and synergistic combination therapies that achieve maximal therapeutic efficacy while maintaining acceptable toxicity profiles.
Despite the challenges of balancing antileukemic efficacy with preservation of normal cellular metabolism, the identification of specific metabolic vulnerabilities of FLT3-mutated leukemia cells opens new avenues for targeted therapeutic strategies. In the following section, we will explore these distinct metabolic dependencies as promising targets for more selective and effective treatment.

4. Metabolic Vulnerabilities of FLT3-Mutated AML

With growing evidence on the dynamic nature of leukemic cell metabolism, the identification of specific metabolic dependencies paved the way for the design of metabolic-oriented treatments able of disarming leukemic cells and potentially overcoming challenges associated with drug resistance.
For many years, intensive induction chemotherapy has represented the standard of care for de novo AML, despite carrying high rates of early post-induction deaths and limited therapeutic benefit for elderly patients [155,156]. Accordingly, the exploration of less toxic treatment options, such as hypomethylating agents (HMAs), has significantly improved overall survival (OS) in patients not eligible for intensive induction therapy. Among the emerging trends in AML treatment, several targeted agents, including inhibitors of FLT3 (FLT3i) [36], Bcl-2 [35], and IDH1/2 [157,158], as well as gemtuzumab ozogamicin [159] and CPX351 [160], have radically changed treatment options for AML. The therapeutic landscape of FLT3-mutated AML has been significantly improved with the approval of tyrosine kinase inhibitors (TKIs), which, either as a single agent or in combination with other drugs, offer less aggressive options and, in some cases, avoid the need for cytotoxic chemotherapy [35]. However, despite these developments, many patients still develop resistance, highlighting the urgent need for innovative approaches.
In the last decade, increasing knowledge about the contribution of metabolic rewiring in AML pathogenesis and progression has led to the investigation of various metabolic interventions, with the aim to further improve treatment outcomes and address resistance mechanisms. Importantly, metabolic rewiring has been extensively observed across various AML subtypes, independent of the presence of FLT3 alterations [161]. Indeed, each AML subtype is associated with a distinct molecular profile and phenotypic landscape that shapes its metabolic dependencies. For instance, metabolomic analyses revealed that NPM1-mutated AML is characterized by high levels of free carnitine, whereas FLT3-ITD displays significant alterations in amino acid metabolism, particularly involving glutamic and aspartic acids, highlighting the key role of glutaminolysis and energy metabolism pathways in differentiating these subtypes [162]. Additionally, IDH1/2 mutations enhance mitochondrial respiration and fatty acid β-oxidation [163], while TP53 [164], DNMT3A [165], and ASXL1 mutations [166] drive distinct metabolic alterations, including enhanced glycolysis, TCA cycle remodeling, mitochondrial overactivation via the Akt/mTOR pathway, and increased oxidative stress, ultimately contributing to hematopoietic stem cell dysfunction and leukemogenesis.
These shared and subtype-specific metabolic vulnerabilities suggest that many metabolic-targeted therapies hold potential beyond FLT3-mutated AML. However, patient stratification integrating metabolic and genomic profiling could represent an excellent strategy to maximize therapeutic efficacy and to design approaches aimed at overcoming resistance mechanisms.
A number of metabolic-oriented drugs have been developed and tested in AML, either pre-clinically or in early-phase trials. These agents target key metabolic processes, offering potential to overcome treatment resistance and selectively eradicate LSCs (Table 1). Notably, treatment resistance in FLT3-mutated AML is closely related to dynamic metabolic adaptations that evolve over time. LSCs harbor metabolic plasticity, enabling them to shift between energy pathways to survive therapeutic pressure. Early in treatment, FLT3-mutated AML cells rely heavily on mitochondrial metabolism and OXPHOS. However, under prolonged FLT3i exposure, these cells adapt by increasing glycolysis and glutaminolysis to maintain energy production and redox balance, contributing to drug resistance [167,168,169].
A recent study by van Gils et al. categorizes metabolic dysregulation as one of six key mechanisms involved in therapy resistance in LSCs [180]. Notably, LSCs often reside in a quiescent state characterized by low ROS levels, favoring low-energy metabolic pathways that limit the efficacy of conventional chemotherapies, which target rapidly proliferating blasts. Additionally, metabolic heterogeneity exists among AML subsets, with some proliferative blasts maintaining mitochondrial activity but exhibiting reduced sensitivity to treatment, highlighting metabolic adaptation as a crucial factor in resistance [181]. These metabolic dynamics and their evolution over time emphasize the need for therapeutic strategies that target multiple metabolic pathways in FLT3-mutated AML to overcome resistance and improve patient outcomes.
Interestingly, bone marrow stromal cells showed the ability to transfer functional mitochondrial to AML cells through contact-dependent endocytic pathways, enhancing mitochondrial ATP production and survival under chemotherapy stress. This mitochondria-mediated metabolic support is particularly relevant for leukemia-initiating cells, contributing to treatment resistance and disease relapse [182].
Recent studies have clearly demonstrated that FLT3-ITD-mutated leukemic cells heavily rely on mitochondrial metabolism to maintain redox homeostasis and that FLT3 inhibition disrupts this balance by reducing OXPHOS activity, leading to decreased ATP production and an increased reliance on glycolysis, making these cells highly sensitive to oxidative stress and mitochondrial damage [183]. Additionally, FLT3i disrupt de novo purine synthesis, which is strictly dependent on mitochondrial function [58].
Several studies have highlighted the potential of combination treatments that target and disrupt metabolic pathways in AML treatment, as summarized in Table 2. These strategies focus on exploiting metabolic vulnerabilities in AML cells. One notable example of synthetic lethality involved the combination of the FLT3i Quizartinib (AC220) with glutaminase inhibitors, such as CB-839, and IACS-010759, a small-molecule inhibitor of complex I of the electron transport chain (ETC), offering a more effective and less toxic therapeutic approach [184,185]. Additionally, recent studies have shown that combining FLT3i with PDP1 inhibition reduces OXPHOS activity and enhances AML sensitivity to FLT3i, further supporting the promise of combination therapies targeting metabolic vulnerabilities in FLT3-mutated AML. Indeed, FLT3-ITD signaling upregulates PDP1 via RAS-MAPK, activating the pyruvate dehydrogenase complex and directing pyruvate toward mitochondrial oxidation. PDP1 depletion selectively impairs respiration and proliferation in FLT3-ITD cells while sparing wild-type counterparts. Crucially, PDP1 mediates quizartinib resistance by sustaining OXPHOS following FLT3 inhibition, and its knockdown restores drug sensitivity [80]. Moreover, FLT3 inhibition primarily disrupts glucose utilization and glycolytic pathways, while sparing glutamine metabolism, thereby inducing a significant dependence on glutaminolysis to sustain mitochondrial function and TCA cycle [186]. Accordingly, targeting glutaminolysis through GLS inhibitors emerges as a potential therapeutic strategy to overcome resistance to FLT3i. In line with these pre-clinical findings, several ongoing and completed clinical trials are investigating the efficacy of combining FLT3i with metabolic agents in relapsed/refractory AML. In particular, the FRIDA trial (NCT05546580) is evaluating the combination of Gilteritinib with ladademstat, an LSD1 inhibitor known to affect mitochondrial metabolism and epigenetic regulation. Additional studies are exploring regimens that include Venetoclax (NCT03625505, NCT04140487); Selinexor (NTC02530476), a nuclear export inhibitor that disrupts energy production and glycolysis; or hypomethylating agents, which interfere with mitochondrial metabolism and redox homeostasis.
Among metabolic-oriented drugs, Venetoclax in combination with azacitidine represents the standard of care for patients with newly diagnosed AML who are unfit for intensive chemotherapy.
Several studies have widely investigated the strong metabolic reprogramming induced by Venetoclax, through the inhibition of mitochondrial metabolism with an effect independent of Bcl-2 inhibition. Furthermore, combination with azacitidine disrupts energy metabolism by reducing glutathione levels, impairing OXPHOS and ultimately targeting leukemia stem cells (LSCs) [187]. The synergistic effect of Venetoclax and azacitidine further highlights the potential of mitochondrial metabolism inhibition in overcoming metabolic vulnerabilities in AML. Notably, a synergistic effect with FLT3i has been demonstrated through the suppression of MCL-1, a pro-survival protein with a key role in leukemia cell survival, especially in high-risk AML cases [195,196].
In light of the results obtained from targeting mitochondrial metabolism, further investigations have been conducted to test the in vitro and in vivo effects of mitochondrial ETC inhibitors, which have been revealed to be effective at reducing leukemia cell viability. Indeed, the high activity of Complex II led to the testing of the effect of genetic knockdown of the chaperone SDHAF1, which was able to delay the growth of AML cells. Consistent with this, pharmacological inhibition of Complex III using antimycin A resulted in a marked decrease in cell proliferation and triggered differentiation processes [188]. Lastly, the inhibition of mitochondrial ATP-synthase (complex V) with oligomycin A significantly improved the sensitivity of FLT3-mutated AML to FLT3i [189]. Overall, these findings may contribute to the identification of novel mitochondrial metabolic targets that could be exploited in combination with established therapies, such as venetoclax, to enhance treatment efficacy and potentially reduce toxicity.
A widely used drug with pleiotropic effects on metabolism is metformin, which has also been tested as an adjuvant treatment for leukemia. Metformin is able to inhibit OXPHOS in AML cells, also impairing glycolysis, and it induces apoptosis in AML cells without affecting normal hematopoietic stem cells [190]. Additionally, it enhances the effectiveness of FLT3i like sorafenib, potentially overcoming resistance by targeting the mTOR pathway, and the sensitivity to venetoclax, with a synergistic effect which results in greater antileukemia effects [191,197]. Moreover, FLT3-positive cells with acquired quizartinib resistance have been the subject of metabolic adaptation studies. Researchers in France discovered that these resistant leukemic cells become dependent on mitochondrial metabolism, with a specific reliance on glutamine oxidation pathways for their continued survival. Their investigation revealed a promising synergistic relationship between quizartinib and L-asparaginase that functions through complementary anti-metabolic mechanisms [79].
Additionally, Sphingolipids, including sphingomyelin (SM), ceramide (Cer), and glycosphingolipids, introduce another of metabolic target. In FLT3-mutated AML, signaling pathways activated by FLT3 mutation suppress ceramide synthase 1 (CerS1) and ceramide metabolism. Targeting FLT3-mutated AML with the FLT3 inhibitor crenolanib induced ceramide accumulation, leading to AML cell death. Additionally, targeting sphingosine kinase 1 (SPHK1) with its inhibitor MP-A08 induced ceramide accumulation, activated the downstream apoptotic integrated stress response, and sensitized AML cells to venetoclax.
Indeed, lipid accumulation within treated AML cells in different subcellular compartments suggests that lipids might induce other pro-cell death cascades, such as mitophagy. Mitophagy has been reported to be involved in the inhibitory effect of Crenolanib, an FLT3 inhibitor, inducing Cer accumulation and subsequently promoting cell death in FLT3-mutated AML cells [198,199].
The high dependency of FLT3-mutated cells on glycolysis has resulted in the identification of key glycolytic enzymes, including hexokinase 2 (HK2) and pyruvate kinase M2 (PKM2), which are often upregulated in this AML setting. Indeed, the use of glycolytic inhibitors, such as 2-Deoxy-d-Glucose (2-DG), significantly enhances the cytotoxic effects induced by TKIs [192,193]. Pharmacological inhibition of the de novo serine synthesis pathway with phosphoglycerate dehydrogenase (PHGDH) inhibitors, such as WQ-2101, significantly enhances the sensitivity of FLT3-mutated AMLs to standard chemotherapy [194]. In particular, PHGDH demonstrates marked overexpression in AML populations characterized by therapeutic refractoriness and adverse clinical outcomes. Genetic ablation or small-molecule inhibition of PHGDH selectively attenuates proliferative capacity and triggers programmed cell death in FLT3-ITD leukemic models both in vitro and within xenograft systems. Emerging evidence reveals that FLT3-ITD-positive patients exhibit systematic upregulation of serine metabolic machinery, encompassing downstream enzymes PSAT1 and PSPH, establishing a metabolic signature associated with enhanced serine flux. Notably, PHGDH inhibition demonstrates synergistic antileukemic activity when combined with established therapeutic agents, including Rylaze and the nucleoside analog cytarabine, suggesting that serine biosynthetic dependency represents an exploitable metabolic vulnerability in FLT3-mutant AML.
Overall, the growing understanding of metabolic rewiring and the identification of distinct metabolic vulnerabilities in FLT3-mutated AML have opened new avenues for novel therapeutic approaches, supporting the rationale for combining FLT3 inhibitors with agents targeting mitochondrial metabolism, glycolysis, glutaminolysis, and lipid pathways. While these insights underscore the potential of metabolic biomarkers in designing personalized treatment strategies, their clinical application still faces significant challenges. Metabolic plasticity, clonal heterogeneity, and the limited clinical validation of these biomarkers remain critical barriers to their effective translation into durable, patient-tailored therapies. Further exploration integrating genomic, metabolomic, and functional profiling will be essential to refine patient stratification and advance the development of metabolism-oriented therapeutic paradigms in AML.

5. Conclusions

The advances in understanding FLT3-positive clone metabolism underscore the importance of a translational approach that integrates metabolomics, genomics, and pharmacology to develop personalized therapies. Mitochondrial metabolism and its associated pathways are central therapeutic vulnerabilities in FLT3-mutated AML. The identification of metabolic dependencies has spurred the development of combinatorial approaches targeting OXPHOS, glycolysis, glutamine, and lipid metabolism, as well as ROS-regulated signaling. Notably, redox-sensitive markers such as HO-1 and PDK1 are emerging as potential biomarkers for patient stratification and treatment response. The integration of these metabolic insights into clinical strategies promises to enhance therapeutic efficacy, overcome drug resistance, and, ultimately, improve patient outcomes. Continued research into the metabolic landscape of FLT3-mutated AML will be essential to refine targeted therapies and to develop personalized, metabolism-oriented treatment paradigms.

Author Contributions

C.B., N.I.N. and S.T. composed, edited, and finalized the review. C.B., M.C. and E.C. designed the figures and reviewed the text. G.C. edited the clinical section and contributed to the overall revision of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

AIRC (AIRC Foundation for Cancer Research, Italy), ID. 30924; The research leading to these results received funding from AIRC under IG 2024—ID. 30924 project—P.I. (N.I.N). Financial support was also provided to N.I.N. via a project funded under the University Scientific Research Call RSA 2024, UTV (DR 3392, dated 23 October 2024), and further support was provided by the MUR-PNRR M4C2I1.3 PE6 project PE00000019 Heal Italia.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Döhner, H.; Weisdorf, D.J.; Bloomfield, C.D. Acute Myeloid Leukemia. N. Engl. J. Med. 2015, 373, 1136–1152. [Google Scholar] [CrossRef]
  2. Löwenberg, B.; Ossenkoppele, G.J.; Putten, W.V.; Schouten, H.C.; Graux, C.; Ferrant, A.; Sonneveld, P.; Maertens, J.; Jongen-Lavrencic, M.; von Lilienfeld-Toal, M.; et al. High-Dose Daunorubicin in Older Patients with Acute Myeloid Leukemia. N. Engl. J. Med. 2009, 361, 1235–1248. [Google Scholar] [CrossRef]
  3. Park, H.J.; Gregory, M.A. Acute Myeloid Leukemia in Elderly Patients: New Targets, New Therapies. Aging Cancer 2023, 4, 51–73. [Google Scholar] [CrossRef]
  4. Dombret, H.; Gardin, C. An Update of Current Treatments for Adult Acute Myeloid Leukemia. Blood 2016, 127, 53–61. [Google Scholar] [CrossRef]
  5. Papaemmanuil, E.; Gerstung, M.; Bullinger, L.; Gaidzik, V.I.; Paschka, P.; Roberts, N.D.; Potter, N.E.; Heuser, M.; Thol, F.; Bolli, N.; et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N. Engl. J. Med. 2016, 374, 2209–2221. [Google Scholar] [CrossRef]
  6. Shimony, S.; Stahl, M.; Stone, R.M. Acute Myeloid Leukemia: 2025 Update on Diagnosis, Risk-Stratification, and Management. Am. J. Hematol. 2025, 100, 860–891. [Google Scholar] [CrossRef] [PubMed]
  7. Jia, D.; Lu, M.; Jung, K.H.; Park, J.H.; Yu, L.; Onuchic, J.N.; Kaipparettu, B.A.; Levine, H. Elucidating Cancer Metabolic Plasticity by Coupling Gene Regulation with Metabolic Pathways. Proc. Natl. Acad. Sci. USA 2019, 116, 3909–3918. [Google Scholar] [CrossRef] [PubMed]
  8. Warburg, O. On the Origin of Cancer Cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef]
  9. Warburg, O.; Wind, F.; Negelein, E. The Metabolism of Tumors in the Body. J. Gen. Physiol. 1927, 8, 519–530. [Google Scholar] [CrossRef] [PubMed]
  10. Liu, H.; Wang, S.; Wang, J.; Guo, X.; Song, Y.; Fu, K.; Gao, Z.; Liu, D.; He, W.; Yang, L.-L. Energy Metabolism in Health and Diseases. Signal Transduct. Target. Ther. 2025, 10, 69. [Google Scholar] [CrossRef]
  11. Yoshida, G.J. Metabolic Reprogramming: The Emerging Concept and Associated Therapeutic Strategies. J. Exp. Clin. Cancer Res. 2015, 34, 111. [Google Scholar] [CrossRef] [PubMed]
  12. Takacova, M.; Kajanova, I.; Kolarcikova, M.; Lapinova, J.; Zatovicova, M.; Pastorekova, S. Understanding Metabolic Alterations and Heterogeneity in Cancer Progression through Validated Immunodetection of Key Molecular Com-ponents: A Case of Carbonic Anhydrase IX. Cancer Metastasis Rev. 2021, 40, 1035–1053. [Google Scholar] [CrossRef]
  13. Banella, C.; Catalano, G.; Travaglini, S.; Pelosi, E.; Ottone, T.; Zaza, A.; Guerrera, G.; Angelini, D.F.; Niscola, P.; Divona, M.; et al. Ascorbate Plus Buformin in AML: A Metabolic Targeted Treatment. Cancers 2022, 14, 2565. [Google Scholar] [CrossRef]
  14. Catalano, G.; Zaza, A.; Banella, C.; Pelosi, E.; Castelli, G.; Marinis, E.D.; Smigliani, A.; Travaglini, S.; Ottone, T.; Divona, M.; et al. MCL1 Regulates AML Cells Metabolism via Direct Interaction with HK2. Metabolic Signature at Onset Predicts Overall Survival in AMLs’ Patients. Leukemia 2023, 37, 1600–1610. [Google Scholar] [CrossRef]
  15. Moloney, J.N.; Stanicka, J.; Cotter, T.G. Subcellular Localization of the FLT3-ITD Oncogene Plays a Significant Role in the Production of NOX- and P22phox-Derived Reactive Oxygen Species in Acute Myeloid Leukemia. Leuk. Res. 2017, 52, 34–42. [Google Scholar] [CrossRef] [PubMed]
  16. Simonetti, G.; Mengucci, C.; Padella, A.; Fonzi, E.; Picone, G.; Delpino, C.; Nanni, J.; Tommaso, R.D.; Franchini, E.; Papayannidis, C.; et al. Integrated Genomic-Metabolic Classification of Acute Myeloid Leukemia Defines a Sub-group with NPM1 and Cohesin/DNA Damage Mutations. Leukemia 2021, 35, 2813–2826. [Google Scholar] [CrossRef] [PubMed]
  17. Parker, S.J.; Metallo, C.M. Metabolic Consequences of Oncogenic IDH Mutations. Pharmacol. Ther. 2015, 152, 54–62. [Google Scholar] [CrossRef]
  18. Nardozza, A.M.; Guarnera, L.; Travaglini, S.; Ottone, T.; Divona, M.; Bellis, E.D.; Savi, A.; Banella, C.; Noguera, N.I.; Di Fusco, D.; et al. Characterization of a Novel IDH2-R159H Mutation in Acute Myeloid Leukaemia: Effects on Cell Metabolism and Differentiation. Br. J. Haematol. 2024, 204, 719–723. [Google Scholar] [CrossRef]
  19. Banella, C.; Catalano, G.; Travaglini, S.; Divona, M.; Masciarelli, S.; Guerrera, G.; Fazi, F.; Coco, F.L.; Voso, M.T.; Noguera, N. PML/RARa Interferes with NRF2 Transcriptional Activity Increasing the Sensitivity to Ascorbate of Acute Promyelocytic Leukemia Cells. Cancers 2019, 12, 95. [Google Scholar] [CrossRef]
  20. Travaglini, S.; Silvestrini, G.; Attardi, E.; Fanciulli, M.; Scalera, S.; Antonelli, S.; Maurillo, L.; Palmieri, R.; Divona, M.; Ciuffreda, L.; et al. Evolution of Transcriptomic Profiles in Relapsed Inv(16) Acute Myeloid Leukemia. Leuk. Res. 2024, 145, 107568. [Google Scholar] [CrossRef]
  21. Bolkun, L.; Pienkowski, T.; Sieminska, J.; Godzien, J.; Pietrowska, K.; Kłoczko, J.; Wierzbowska, A.; Moniuszko, M.; Ratajczak, M.; Kretowski, A.; et al. Metabolomic Profile of Acute Myeloid Leukaemia Parallels of Prognosis and Response to Therapy. Sci. Rep. 2023, 23, 21809. [Google Scholar] [CrossRef]
  22. Sung, J.-Y.; Yun, W.; Kim, H.-Y.; Kim, H.-J.; Choi, J.R.; Kim, S.-H.; Jung, C.W.; Lee, S.-T. Metabolic Subtype Reveals Potential Therapeutic Vulnerability in Acute Promyelocytic Leukaemia. Clin. Transl. Med. 2022, 12, e964. [Google Scholar] [CrossRef]
  23. Balasundaram, N.; Ganesan, S.; Chendamarai, E.; Palani, H.K.; Venkatraman, A.; Alex, A.A.; David, S.; Kumar, S.P.; Radhakrishnan, N.R.; Yasar, M.; et al. Metabolic Adaptation Drives Arsenic Trioxide Resistance in Acute Promyelocytic Leukemia. Blood Adv. 2022, 6, 652–663. [Google Scholar] [CrossRef]
  24. Ferret, Y.; Boissel, N.; Helevaut, N.; Madic, J.; Nibourel, O.; Marceau-Renaut, A.; Bucci, M.; Geffroy, S.; Celli-Lebras, K.; Castaigne, S.; et al. Clinical Relevance of IDH1/2 Mutant Allele Burden during Follow-up in Acute Myeloid Leukemia. A Study by the French ALFA Group. Haematologica 2018, 103, 822–829. [Google Scholar] [CrossRef]
  25. Chen, W.-L.; Wang, J.-H.; Zhao, A.-H.; Xu, X.; Wang, Y.-H.; Chen, T.-L.; Li, J.-M.; Mi, J.-Q.; Zhu, Y.-M.; Liu, Y.-F.; et al. A Distinct Glucose Metabolism Signature of Acute Myeloid Leukemia with Prognostic Value. Blood 2014, 124, 1645–1654. [Google Scholar] [CrossRef] [PubMed]
  26. Larrue, C.; Saland, E.; Vergez, F.; Serhan, N.; Delabesse, E.; Mas, V.M.-D.; Hospital, M.-A.; Tamburini, J.; Manenti, S.; Sarry, J.E.; et al. Antileukemic Activity of 2-Deoxy-d-Glucose through Inhibition of N-Linked Glycosylation in Acute Myeloid Leukemia with FLT3-ITD or c-KIT Mutations. Mol. Cancer Ther. 2015, 14, 2364–2373. [Google Scholar] [CrossRef] [PubMed]
  27. Jacque, N.; Ronchetti, A.M.; Larrue, C.; Meunier, G.; Birsen, R.; Willems, L.; Saland, E.; Decroocq, J.; Maciel, T.T.; Lambert, M.; et al. Targeting Glutaminolysis Has Antileukemic Activity in Acute Myeloid Leukemia and Synergizes with BCL-2 Inhibition. Blood 2015, 126, 1346–1356. [Google Scholar] [CrossRef]
  28. Liyanage, S.U.; Hurren, R.; Voisin, V.; Bridon, G.; Wang, X.; Xu, C.; MacLean, N.; Siriwardena, T.P.; Gronda, M.; Yehudai, D.; et al. Leveraging Increased Cytoplasmic Nucleoside Kinase Activity to Target MtDNA and Oxidative Phosphorylation in AML. Blood 2017, 129, 2657–2666. [Google Scholar] [CrossRef] [PubMed]
  29. Ricciardi, M.R.; Mirabilii, S.; Allegretti, M.; Licchetta, R.; Calarco, A.; Torrisi, M.R.; Foà, R.; Nicolai, R.; Peluso, G.; Tafuri, A. Targeting the Leukemia Cell Metabolism by the CPT1a Inhibition: Functional Preclinical Effects in Leukemias. Blood 2015, 126, 1925–1929. [Google Scholar] [CrossRef]
  30. Cai, T.; Lorenzi, P.L.; Rakheja, D.; Pontikos, M.A.; Lodi, A.; Han, L.; Zhang, Q.; Ma, H.; Rahmani, M.; Bhagat, T.D.; et al. Gls Inhibitor CB-839 Modulates Cellular Metabolism in AML and Potently Suppresses AML Cell Growth When Combined with 5-Azacitidine. Blood 2016, 128, 4064. [Google Scholar] [CrossRef]
  31. Kiyoi, H.; Naoe, T. Biology, Clinical Relevance, and Molecularly Targeted Therapy in Acute Leukemia with FLT3 Mutation. Int. J. Hematol. 2006, 83, 301–308. [Google Scholar] [CrossRef]
  32. Döhner, H.; Wei, A.H.; Appelbaum, F.R.; Craddock, C.; DiNardo, C.D.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Godley, L.A.; Hasserjian, R.P.; et al. Diagnosis and Management of AML in Adults: 2022 Recommendations from an International Expert Panel on Behalf of the ELN. Blood 2022, 140, 1345–1377. [Google Scholar] [CrossRef]
  33. Bruno, S.; Bandini, L.; Patuelli, A.; Robustelli, V.; Venturi, C.; Mancini, M.; Forte, D.; Santis, S.D.; Monaldi, C.; Grassi, A.; et al. Case Report: A Novel Activating FLT3 Mutation in Acute Myeloid Leukemia. Front. Oncol. 2021, 11, 728613. [Google Scholar] [CrossRef]
  34. Travaglini, S.; Gurnari, C.; Antonelli, S.; Marchesi, F.; Angelis, G.D.; Ottone, T.; Divona, M.; Cristiano, A.; Hajrullaj, H.; Mengarelli, A.; et al. Functional Characterization and Response to FLT3 Inhibitors in Acute Myeloid Leukaemia with a Non-Canonical FLT3 Mutation: A Proof of Concept. Br. J. Haematol. 2023, 203, 327–330. [Google Scholar] [CrossRef]
  35. Kennedy, V.E.; Smith, C.C. FLT3 Targeting in the Modern Era: From Clonal Selection to Combination Therapies. Int. J. Hematol. 2023, 120, 528–540. [Google Scholar] [CrossRef]
  36. Ruglioni, M.; Crucitta, S.; Luculli, G.I.; Tancredi, G.; Del Giudice, M.L.; Mechelli, S.; Galimberti, S.; Danesi, R.; Del Re, M. Understanding Mechanisms of Resistance to FLT3 Inhibitors in Adult FLT3-Mutated Acute Myeloid Leukemia to Guide Treatment Strategy. Crit. Rev. Oncol. Hematol. 2024, 201, 104424. [Google Scholar] [CrossRef]
  37. Travaglini, S.; Gurnari, C.; Ottone, T.; Voso, M.T. Advances in the Pathogenesis of FLT3 -Mutated Acute Myeloid Leukemia and Targeted Treatments. Curr. Opin. Oncol. 2024, 36, 569–576. [Google Scholar] [CrossRef]
  38. Maryanovich, M.; Zaltsman, Y.; Ruggiero, A.; Goldman, A.; Shachnai, L.; Zaidman, S.L.; Porat, Z.; Golan, K.; Lapidot, T.; Gross, A. An MTCH2 Pathway Repressing Mitochondria Metabolism Regulates Haematopoietic Stem Cell Fate. Nat. Commun. 2015, 6, 7901. [Google Scholar] [CrossRef] [PubMed]
  39. Folmes, C.D.L.; Dzeja, P.P.; Nelson, T.J.; Terzic, A. Metabolic Plasticity in Stem Cell Homeostasis and Differentiation. Cell Stem Cell 2012, 11, 596–606. [Google Scholar] [CrossRef] [PubMed]
  40. Ito, K.; Suda, T. Metabolic Requirements for the Maintenance of Self-Renewing Stem Cells. Nat. Rev. Mol. Cell Biol. 2014, 15, 243–256. [Google Scholar] [CrossRef] [PubMed]
  41. Du, W.; Amarachintha, S.; Wilson, A.F.; Pang, Q. SCO2 Mediates Oxidative Stress-Induced Glycolysis to Oxidative Phosphorylation Switch in Hematopoietic Stem Cells. Stem Cells 2016, 34, 960–971. [Google Scholar] [CrossRef]
  42. Ito, K.; Bonora, M.; Ito, K. Metabolism as Master of Hematopoietic Stem Cell Fate. Int. J. Hematol. 2019, 109, 18–27. [Google Scholar] [CrossRef] [PubMed]
  43. Maiso, P.; Huynh, D.; Moschetta, M.; Sacco, A.; Aljawai, Y.; Mishima, Y.; Asara, J.M.; Roccaro, A.M.; Kimmelman, A.C.; Ghobrial, I.M. Metabolic Signature Identifies Novel Targets for Drug Resistance in Multiple Myeloma. Cancer Res. 2015, 75, 2071–2082. [Google Scholar] [CrossRef] [PubMed]
  44. Herst, P.M.; Howman, R.A.; Neeson, P.J.; Berridge, M.V.; Ritchie, D.S. The Level of Glycolytic Metabolism in Acute Myeloid Leukemia Blasts at Diagnosis Is Prognostic for Clinical Outcome. J. Leukoc. Biol. 2010, 89, 51–55. [Google Scholar] [CrossRef]
  45. Kominsky, D.J.; Klawitter, J.; Brown, J.L.; Boros, L.G.; Melo, J.V.; Eckhardt, S.G.; Serkova, N.J. Abnormalities in Glucose Uptake and Metabolism in Imatinib-Resistant Human BCR-ABL–Positive Cells. Clin. Cancer Res. 2009, 15, 3442–3450. [Google Scholar] [CrossRef] [PubMed]
  46. Lagadinou, E.D.; Sach, A.; Callahan, K.; Rossi, R.M.; Neering, S.J.; Minhajuddin, M.; Ashton, J.M.; Pei, S.; Grose, V.; O’Dwyer, K.M.; et al. BCL-2 Inhibition Targets Oxidative Phosphorylation and Selectively Eradicates Quiescent Human Leukemia Stem Cells. Cell Stem Cell 2013, 12, 329–341. [Google Scholar] [CrossRef] [PubMed]
  47. Zhong, W.; Yi, Q.; Xu, B.; Li, S.; Wang, T.; Liu, F.; Zhu, B.; Hoffmann, P.R.; Ji, G.; Lei, P.; et al. ORP4L Is Essential for T-Cell Acute Lymphoblastic Leukemia Cell Survival. Nat. Commun. 2016, 7, 12702. [Google Scholar] [CrossRef]
  48. Boag, J.M.; Beesley, A.H.; Firth, M.J.; Freitas, J.R.; Ford, J.; Hoffmann, K.; Cummings, A.J.; Klerk, N.H.D.; Kees, U.R. Altered Glucose Metabolism in Childhood Pre-B Acute Lymphoblastic Leukaemia. Leukemia 2006, 20, 1731–1737. [Google Scholar] [CrossRef]
  49. Spinelli, J.B.; Haigis, M.C. The Multifaceted Contributions of Mitochondria to Cellular Metabolism. Nat. Cell Biol. 2018, 20, 745–754. [Google Scholar] [CrossRef]
  50. Tjahjono, E.; Daneman, M.R.; Meika, B.; Revtovich, A.V.; Kirienko, N.V. Mitochondrial Abnormalities as a Target of Intervention in Acute Myeloid Leukemia. Front. Oncol. 2024, 14, 1532857. [Google Scholar] [CrossRef]
  51. Morganti, C.; Bonora, M.; Ito, K. Metabolism and HSC Fate: What NADPH Is Made For. Trends Cell Biol. 2024. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, Y.W.; Schönberger, K.; Cabezas-Wallscheid, N. Bidirectional Interplay between Metabolism and Epigenetics in Hematopoietic Stem Cells and Leukemia. EMBO J. 2023, 42, e112348. [Google Scholar] [CrossRef]
  53. Tavor, S.; Petit, I.; Porozov, S.; Avigdor, A.; Dar, A.; Leider-Trejo, L.; Shemtov, N.; Deutsch, V.; Naparstek, E.; Nagler, A.; et al. CXCR4 Regulates Migration and Development of Human Acute Myelogenous Leukemia Stem Cells in Transplanted NOD/SCID Mice. Cancer Res. 2004, 64, 2817–2824. [Google Scholar] [CrossRef] [PubMed]
  54. Möhle, R.; Bautz, F.; Rafii, S.; Moore, M.A.; Brugger, W.; Kanz, L. The Chemokine Receptor CXCR-4 Is Expressed on CD34+ Hematopoietic Progenitors and Leukemic Cells and Mediates Transendothelial Migration Induced by Stromal Cell-Derived Factor-1. Blood 1998, 91, 4523–4530. [Google Scholar] [CrossRef]
  55. Jin, L.; Hope, K.J.; Zhai, Q.; Smadja-Joffe, F.; Dick, J.E. Targeting of CD44 Eradicates Human Acute Myeloid Leukemic Stem Cells. Nat. Med. 2006, 12, 1167–1174. [Google Scholar] [CrossRef] [PubMed]
  56. Chipuk, J.E.; Bouchier-Hayes, L.; Green, D.R. Mitochondrial Outer Membrane Permeabilization during Apoptosis: The Innocent Bystander Scenario. Cell Death Differ. 2006, 13, 1396–1402. [Google Scholar] [CrossRef]
  57. Sriskanthadevan, S.; Jeyaraju, D.V.; Chung, T.E.; Prabha, S.; Xu, W.; Skrtic, M.; Jhas, B.; Hurren, R.; Gronda, M.; Wang, X.; et al. AML Cells Have Low Spare Reserve Capacity in Their Respiratory Chain That Renders Them Susceptible to Oxidative Metabolic Stress. Blood 2015, 125, 2120–2130. [Google Scholar] [CrossRef]
  58. Zhang, P.; Brinton, L.T.; Gharghabi, M.; Sher, S.; Williams, K.; Cannon, M.; Walker, J.S.; Canfield, D.; Beaver, L.; Cempre, C.B.; et al. Targeting OXPHOS de Novo Purine Synthesis as the Nexus of FLT3 Inhibitor-Mediated Synergistic Antileukemic Actions. Sci. Adv. 2022, 8, eabp9005. [Google Scholar] [CrossRef]
  59. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  60. Ganapathy-Kanniappan, S.; Kunjithapatham, R.; Geschwind, J.-F.H. Statins Impair Glucose Uptake in Tumor Cells. Cancer Biol. Ther. 2013, 14, 92–94. [Google Scholar] [CrossRef]
  61. Huang, I.N.; Melvin, J.N. Effects of Ratio Reinforcement Schedules on Choice Behavior. J. Gen. Psychol. 1990, 117, 99–106. [Google Scholar] [CrossRef]
  62. Tang, Z.; Yuan, S.; Hu, Y.; Zhang, H.; Wu, W.; Zeng, Z.; Yang, J.; Yun, J.; Xu, R.; Huang, P. Over-Expression of GAPDH in Human Colorectal Carcinoma as a Preferred Target of 3-Bromopyruvate Propyl Ester. J. Bioenerg. Biomembr. 2012, 44, 117–125. [Google Scholar] [CrossRef]
  63. Zhou, Y.; Zhou, Y.; Shingu, T.; Feng, L.; Chen, Z.; Ogasawara, M.; Keating, M.J.; Kondo, S.; Huang, P. Metabolic Alterations in Highly Tumorigenic Glioblastoma Cells: Preference for Hypoxia and High Dependency on Glycolysis. J. Biol. Chem. 2011, 286, 32843–32853. [Google Scholar] [CrossRef] [PubMed]
  64. Geschwind, J.-F.; Georgiades, C.S.; Ko, Y.H.; Pedersen, P.L. Recently Elucidated Energy Catabolism Pathways Provide Opportunities for Novel Treatments in Hepatocellular Carcinoma. Expert. Rev. Anticancer. Ther. 2004, 4, 449–457. [Google Scholar] [CrossRef] [PubMed]
  65. Liberti, M.V.; Locasale, J.W. The Warburg Effect: How Does It Benefit Cancer Cells? Trends Biochem. Sci. 2016, 41, 211–218. [Google Scholar] [CrossRef] [PubMed]
  66. Song, K.; Li, M.; Xu, X.; Xuan, L.; Huang, G.; Liu, Q. Resistance to Chemotherapy Is Associated with Altered Glucose Metabolism in Acute Myeloid Leukemia. Oncol. Lett. 2016, 12, 334–342. [Google Scholar] [CrossRef]
  67. Wang, Y.-H.; Israelsen, W.J.; Lee, D.; Yu, V.W.C.; Jeanson, N.T.; Clish, C.B.; Cantley, L.C.; Vander Heiden, M.G.; Scadden, D.T. Cell-State-Specific Metabolic Dependency in Hematopoiesis and Leukemogenesis. Cell 2014, 158, 1309–1323. [Google Scholar] [CrossRef]
  68. Ju, H.-Q.; Zhan, G.; Huang, A.; Sun, Y.; Wen, S.; Yang, J.; Lu, W.; Xu, R.; Li, J.; Li, Y.; et al. ITD Mutation in FLT3 Tyrosine Kinase Promotes Warburg Effect and Renders Therapeutic Sensitivity to Glycolytic Inhibition. Leukemia 2017, 31, 2143–2150. [Google Scholar] [CrossRef]
  69. Koppenol, W.H.; Bounds, P.L.; Dang, C. V Otto Warburg’s Contributions to Current Concepts of Cancer Metabolism. Nat. Rev. Cancer 2011, 11, 325–337. [Google Scholar] [CrossRef]
  70. DeBerardinis, R.J.; Chandel, N.S. We Need to Talk about the Warburg Effect. Nat. Metab. 2020, 2, 127–129. [Google Scholar] [CrossRef]
  71. Erdem, A.; Marin, S.; Pereira-Martins, D.A.; Cortés, R.; Cunningham, A.; Pruis, M.G.; Boer, B.D.; Heuvel, F.A.J.V.D.; Geugien, M.; Wierenga, A.T.J.; et al. The Glycolytic Gatekeeper PDK1 Defines Different Metabolic States between Genetically Distinct Subtypes of Human Acute Myeloid Leukemia. Nat. Commun. 2022, 13, 1105. [Google Scholar] [CrossRef]
  72. Peng, C.-J.; Fan, Z.; Luo, J.-S.; Wang, L.-N.; Li, Y.; Liang, C.; Zhang, X.-L.; Luo, X.-Q.; Huang, L.-B.; Tang, Y.-L. The Potential Transcriptomic and Metabolomic Mechanisms of ATO and ATRA in Treatment of FLT3-ITD Acute Myeloid Leukemia. Technol. Cancer Res. Treat. 2024, 23, 15330338231223080. [Google Scholar] [CrossRef]
  73. Huang, A.; Ju, H.-Q.; Liu, K.; Zhan, G.; Liu, D.; Wen, S.; Garcia-Manero, G.; Huang, P.; Hu, Y. Metabolic Alterations and Drug Sensitivity of Tyrosine Kinase Inhibitor Resistant Leukemia Cells with a FLT3/ITD Mutation. Cancer Lett. 2016, 377, 149–157. [Google Scholar] [CrossRef] [PubMed]
  74. Soltani, M.; Zhao, Y.; Xia, Z.; Ganjalikhani Hakemi, M.; Bazhin, A.V. The Importance of Cellular Metabolic Pathways in Pathogenesis and Selective Treatments of Hematological Malignancies. Front. Oncol. 2021, 11, 767026. [Google Scholar] [CrossRef]
  75. Vettore, L.; Westbrook, R.L.; Tennant, D.A. New Aspects of Amino Acid Metabolism in Cancer. Br. J. Cancer 2020, 122, 150–156. [Google Scholar] [CrossRef]
  76. Cormerais, Y.; Massard, P.A.; Vucetic, M.; Giuliano, S.; Tambutté, E.; Durivault, J.; Vial, V.; Endou, H.; Wempe, M.F.; Parks, S.K.; et al. The Glutamine Transporter ASCT2 (SLC1A5) Promotes Tumor Growth Independently of the Amino Acid Transporter LAT1 (SLC7A5). J. Biol. Chem. 2018, 293, 2877–2887. [Google Scholar] [CrossRef]
  77. Willems, L.; Jacque, N.; Jacquel, A.; Neveux, N.; Trovati Maciel, T.; Lambert, M.; Schmitt, A.; Poulain, L.; Green, A.S.; Uzunov, M.; et al. Inhibiting Glutamine Uptake Represents an Attractive New Strategy for Treating Acute Myeloid Leukemia. Blood 2013, 122, 3521–3532. [Google Scholar] [CrossRef]
  78. Nicklin, P.; Bergman, P.; Zhang, B.; Triantafellow, E.; Wang, H.; Nyfeler, B.; Yang, H.; Hild, M.; Kung, C.; Wilson, C.; et al. Bidirectional Transport of Amino Acids Regulates MTOR and Autophagy. Cell 2009, 136, 521–534. [Google Scholar] [CrossRef]
  79. Khamari, R.; Degand, C.; Fovez, Q.; Trinh, A.; Chomy, A.; Laine, W.; Dekiouk, S.; Ghesquiere, B.; Quesnel, B.; Marchetti, P.; et al. Key Role of Glutamine Metabolism in Persistence of Leukemic Cells upon Exposition to FLT3 Tyrosine Kinase Inhibitors. Exp. Hematol. 2024, 137, 104253. [Google Scholar] [CrossRef] [PubMed]
  80. Alshamleh, I.; Kurrle, N.; Makowka, P.; Bhayadia, R.; Kumar, R.; Süsser, S.; Seibert, M.; Ludig, D.; Wolf, S.; Koschade, S.E.; et al. PDP1 Is a Key Metabolic Gatekeeper and Modulator of Drug Resistance in FLT3-ITD-Positive Acute Myeloid Leukemia. Leukemia 2023, 37, 2367–2382. [Google Scholar] [CrossRef] [PubMed]
  81. Wang, E.S.; Frankfurt, O.; Orford, K.W.; Bennett, M.; Flinn, I.W.; Maris, M.; Konopleva, M. Phase 1 Study of CB-839, a First-in-Class, Orally Administered Small Molecule Inhibitor of Glutaminase in Patients with Relapsed/Refractory Leukemia. Blood 2015, 126, 2566. [Google Scholar] [CrossRef]
  82. Firmanty, P.; Chomczyk, M.; Dash, S.; Konopleva, M.; Baran, N. Feasibility and Safety of Targeting Mitochondria Function and Metabolism in Acute Myeloid Leukemia. Curr. Pharmacol. Rep. 2024, 10, 388–404. [Google Scholar] [CrossRef]
  83. Harding, J.J.; Telli, M.L.; Munster, P.N.; Le, M.H.; Molineaux, C.; Bennett, M.K.; Mittra, E.; Burris, H.A.; Clark, A.S.; Dunphy, M.; et al. Safety and Tolerability of Increasing Doses of CB-839, a First-in-Class, Orally Administered Small Molecule Inhibitor of Glutaminase, in Solid Tumors. J. Clin. Oncol. 2015, 33, 2512. [Google Scholar] [CrossRef]
  84. Maher, M.; Diesch, J.; Casquero, R.; Buschbeck, M. Epigenetic-Transcriptional Regulation of Fatty Acid Metabolism and Its Alterations in Leukaemia. Front. Genet. 2018, 9, 405. [Google Scholar] [CrossRef]
  85. Snaebjornsson, M.T.; Janaki-Raman, S.; Schulze, A. Greasing the Wheels of the Cancer Machine: The Role of Lipid Metabolism in Cancer. Cell Metab. 2020, 31, 62–76. [Google Scholar] [CrossRef]
  86. Balko, J.M.; Schwarz, L.J.; Luo, N.; Estrada, M.V.; Giltnane, J.M.; Dávila-González, D.; Wang, K.; Sánchez, V.; Dean, P.T.; Combs, S.E.; et al. Triple-Negative Breast Cancers with Amplification of JAK2 at the 9p24 Locus Demonstrate JAK2-Specific Dependence. Sci. Transl. Med. 2016, 8, 334ra53. [Google Scholar] [CrossRef]
  87. Samudio, I.; Konopleva, M. Targeting Leukemia’s “Fatty Tooth”. Blood 2015, 126, 1874–1875. [Google Scholar] [CrossRef]
  88. Pabst, T.; Kortz, L.; Fiedler, G.M.; Ceglarek, U.; Idle, J.R.; Beyoğlu, D. The Plasma Lipidome in Acute Myeloid Leukemia at Diagnosis in Relation to Clinical Disease Features. BBA Clin. 2017, 7, 105–114. [Google Scholar] [CrossRef]
  89. Mesbahi, Y.; Trahair, T.N.; Lock, R.B.; Connerty, P. Exploring the Metabolic Landscape of AML: From Haematopoietic Stem Cells to Myeloblasts and Leukaemic Stem Cells. Front. Oncol. 2022, 12, 807266. [Google Scholar] [CrossRef] [PubMed]
  90. Loeffler, D.; Schroeder, T. Symmetric and Asymmetric Activation of Hematopoietic Stem Cells. Curr. Opin. Hematol. 2021, 28, 262–268. [Google Scholar] [CrossRef] [PubMed]
  91. Ito, K.; Carracedo, A.; Weiss, D.; Arai, F.; Ala, U.; Avigan, D.E.; Schafer, Z.T.; Evans, R.M.; Suda, T.; Lee, C.-H.; et al. A PML–PPAR-δ Pathway for Fatty Acid Oxidation Regulates Hematopoietic Stem Cell Maintenance. Nat. Med. 2012, 18, 1350–1358. [Google Scholar] [CrossRef] [PubMed]
  92. Samudio, I.; Harmancey, R.; Fiegl, M.; Kantarjian, H.; Konopleva, M.; Korchin, B.; Kaluarachchi, K.; Bornmann, W.; Duvvuri, S.; Taegtmeyer, H.; et al. Pharmacologic Inhibition of Fatty Acid Oxidation Sensitizes Human Leukemia Cells to Apoptosis Induction. J. Clin. Investig. 2010, 120, 142–156. [Google Scholar] [CrossRef]
  93. Bonnefont, J.-P.; Bastin, J.; Behin, A.; Djouadi, F. Bezafibrate for an Inborn Mitochondrial Beta-Oxidation Defect. N. Engl. J. Med. 2009, 360, 838–840. [Google Scholar] [CrossRef]
  94. Yamaguchi, S.; Li, H.; Purevsuren, J.; Yamada, K.; Furui, M.; Takahashi, T.; Mushimoto, Y.; Kobayashi, H.; Hasegawa, Y.; Taketani, T.; et al. Bezafibrate Can Be a New Treatment Option for Mitochondrial Fatty Acid Oxidation Disorders: Evaluation by in Vitro Probe Acylcarnitine Assay. Mol. Genet. Metab. 2012, 107, 87–91. [Google Scholar] [CrossRef]
  95. Ogretmen, B. Sphingolipid Metabolism in Cancer Signalling and Therapy. Nat. Rev. Cancer 2018, 18, 33–50. [Google Scholar] [CrossRef]
  96. Schömel, N.; Geisslinger, G.; Wegner, M.-S. Influence of Glycosphingolipids on Cancer Cell Energy Metabolism. Prog. Lipid Res. 2020, 79, 101050. [Google Scholar] [CrossRef]
  97. Canals, D.; Clarke, C.J. Compartmentalization of Sphingolipid Metabolism: Implications for Signaling and Therapy. Pharmacol. Ther. 2022, 232, 108005. [Google Scholar] [CrossRef] [PubMed]
  98. Powell, J.A.; Lewis, A.C.; Zhu, W.; Toubia, J.; Pitman, M.R.; Wallington-Beddoe, C.T.; Moretti, P.A.B.; Iarossi, D.; Samaraweera, S.E.; Cummings, N.; et al. Targeting Sphingosine Kinase 1 Induces MCL1-Dependent Cell Death in Acute Myeloid Leukemia. Blood 2017, 129, 771–782. [Google Scholar] [CrossRef] [PubMed]
  99. Xie, S.Z.; Kaufmann, K.B.; Wang, W.; Chan-Seng-Yue, M.; Gan, O.I.; Laurenti, E.; Garcia-Prat, L.; Takayanagi, S.; Ng, S.W.K.; Xu, C.; et al. Sphingosine-1-Phosphate Receptor 3 Potentiates Inflammatory Programs in Normal and Leukemia Stem Cells to Promote Differentiation. Blood Cancer Discov. 2021, 2, 32–53. [Google Scholar] [CrossRef]
  100. Mendoza, A.E.-H.D.; Castello-Cros, R.; Imbuluzqueta, E.; Cirauqui, C.; Pippa, R.; Odero, M.D.; Blanco-Prieto, M.J. Lipid Nanosystems Enhance the Bioavailability and the Therapeutic Efficacy of FTY720 in Acute Myeloid Leukemia. J. Biomed. Nanotechnol. 2015, 11, 691–701. [Google Scholar] [CrossRef]
  101. Cristóbal, I.; Garcia-Orti, L.; Cirauqui, C.; Alonso, M.M.; Calasanz, M.J.; Odero, M.D. PP2A Impaired Activity Is a Common Event in Acute Myeloid Leukemia and Its Activation by Forskolin Has a Potent Anti-Leukemic Effect. Leukemia 2011, 25, 606–614. [Google Scholar] [CrossRef]
  102. Arriazu, E.; Vicente, C.; Pippa, R.; Peris, I.; Martínez-Balsalobre, E.; García-Ramírez, P.; Marcotegui, N.; Igea, A.; Alignani, D.; Rifón, J.; et al. A New Regulatory Mechanism of Protein Phosphatase 2A Activity via SET in Acute Myeloid Leukemia. Blood Cancer J. 2020, 10, 3. [Google Scholar] [CrossRef]
  103. Bai, A.-P.; Guo, Y. Ceramide Is a Potential Activator of Immune Responses Against Tumors. Gastroenterology 2018, 155, 579–580. [Google Scholar] [CrossRef]
  104. Morad, S.A.F.; Cabot, M.C. Ceramide-Orchestrated Signalling in Cancer Cells. Nat. Rev. Cancer 2013, 13, 51–65. [Google Scholar] [CrossRef] [PubMed]
  105. Maceyka, M.; Harikumar, K.B.; Milstien, S.; Spiegel, S. Sphingosine-1-Phosphate Signaling and Its Role in Disease. Trends Cell Biol. 2012, 22, 50–60. [Google Scholar] [CrossRef] [PubMed]
  106. Kumagai, K.; Hanada, K. Structure, Functions and Regulation of CERT, a Lipid-transfer Protein for the Delivery of Ceramide at the ER—Golgi Membrane Contact Sites. FEBS Lett. 2019, 593, 2366–2377. [Google Scholar] [CrossRef]
  107. Dany, M.; Gencer, S.; Nganga, R.; Thomas, R.J.; Oleinik, N.; Baron, K.D.; Szulc, Z.M.; Ruvolo, P.; Kornblau, S.; Andreeff, M.; et al. Targeting FLT3-ITD Signaling Mediates Ceramide-Dependent Mitophagy and Attenuates Drug Resistance in AML. Blood 2016, 128, 1944–1958. [Google Scholar] [CrossRef]
  108. Lewis, A.C.; Pope, V.S.; Tea, M.N.; Li, M.; Nwosu, G.O.; Nguyen, T.M.; Wallington-Beddoe, C.T.; Moretti, P.A.B.; Anderson, D.; Creek, D.J.; et al. Ceramide-Induced Integrated Stress Response Overcomes Bcl-2 Inhibitor Resistance in Acute Myeloid Leukemia. Blood 2022, 139, 3737–3751. [Google Scholar] [CrossRef] [PubMed]
  109. Nguyen, T.M.; Joyce, P.; Ross, D.M.; Bremmell, K.; Jambhrunkar, M.; Wong, S.S.; Prestidge, C.A. Combating Acute Myeloid Leukemia via Sphingosine Kinase 1 Inhibitor-Nanomedicine Combination Therapy with Cytarabine or Venetoclax. Pharmaceutics 2024, 16, 209. [Google Scholar] [CrossRef]
  110. Sun, X.; Li, Y.; Du, J.; Liu, F.; Wu, C.; Xiao, W.; Yu, G.; Chen, X.; Gale, R.P.; Zeng, H. Targeting Ceramide Transfer Protein Sensitizes AML to FLT3 Inhibitors via a GRP78-ATF6-CHOP Axis. Nat. Commun. 2025, 16, 1358. [Google Scholar] [CrossRef]
  111. Swanton, C.; Marani, M.; Pardo, O.; Warne, P.H.; Kelly, G.; Sahai, E.; Elustondo, F.; Chang, J.; Temple, J.; Ahmed, A.A.; et al. Regulators of Mitotic Arrest and Ceramide Metabolism Are Determinants of Sensitivity to Paclitaxel and Other Chemotherapeutic Drugs. Cancer Cell 2007, 11, 498–512. [Google Scholar] [CrossRef]
  112. Chung, L.H.; Liu, D.; Liu, X.T.; Qi, Y. Ceramide Transfer Protein (CERT): An Overlooked Molecular Player in Cancer. Int. J. Mol. Sci. 2021, 22, 13184. [Google Scholar] [CrossRef]
  113. Sabatier, M.; Birsen, R.; Lauture, L.; Mouche, S.; Angelino, P.; Dehairs, J.; Goupille, L.; Boussaid, I.; Heiblig, M.; Boet, E.; et al. C/EBPα Confers Dependence to Fatty Acid Anabolic Pathways and Vulnerability to Lipid Oxidative Stress–Induced Ferroptosis in FLT3 -Mutant Leukemia. Cancer Discov. 2023, 13, 1720–1747. [Google Scholar] [CrossRef]
  114. Chang, H.-H.; Lee, L.-C.; Hsu, T.; Peng, Y.-H.; Huang, C.-H.; Yeh, T.-K.; Lu, C.-T.; Huang, Z.-T.; Hsueh, C.-C.; Kung, F.-C.; et al. Development of Potent and Selective Inhibitors of Methylenetetrahydrofolate Dehydrogenase 2 for Targeting Acute Myeloid Leukemia: SAR, Structural Insights, and Biological Characterization. J. Med. Chem. 2024, 67, 21106–21125. [Google Scholar] [CrossRef]
  115. Pikman, Y.; Puissant, A.; Alexe, G.; Furman, A.; Chen, L.M.; Frumm, S.M.; Ross, L.; Fenouille, N.; Bassil, C.F.; Lewis, C.A.; et al. Targeting MTHFD2 in Acute Myeloid Leukemia. J. Exp. Med. 2016, 213, 1285–1306. [Google Scholar] [CrossRef] [PubMed]
  116. Stavrou, V.; Fultang, L.; Booth, S.; Simone, D.D.; Bartnik, A.; Scarpa, U.; Gneo, L.; Panetti, S.; Potluri, S.; Almowaled, M.; et al. Invariant NKT Cells Metabolically Adapt to the Acute Myeloid Leukaemia Environment. Cancer Immunol. Immunother. 2023, 72, 543–560. [Google Scholar] [CrossRef] [PubMed]
  117. Shi, X.; Li, M.; Liu, Z.; Tiessen, J.; Li, Y.; Zhou, J.; Zhu, Y.; Mahesula, S.; Ding, Q.; Tan, L.; et al. Guanine Nucleotide Biosynthesis Blockade Impairs MLL Complex Formation and Sensitizes Leukemias to Menin Inhibition. Nat. Commun. 2025, 16, 2641. [Google Scholar] [CrossRef] [PubMed]
  118. Zhang, Y.W.; Velasco-Hernandez, T.; Mess, J.; Lalioti, M.-E.; Romero-Mulero, M.C.; Obier, N.; Karantzelis, N.; Rettkowski, J.; Schönberger, K.; Karabacz, N.; et al. GPRC5C Drives Branched-Chain Amino Acid Metabolism in Leukemogenesis. Blood Adv. 2023, 7, 7525–7538. [Google Scholar] [CrossRef]
  119. Baranello, M.P.; Bauer, L.; Jordan, C.T.; Benoit, D.S.W. Micelle Delivery of Parthenolide to Acute Myeloid Leukemia Cells. Cell Mol. Bioeng. 2015, 8, 455–470. [Google Scholar] [CrossRef] [PubMed]
  120. Jones, C.L.; Inguva, A.; Jordan, C.T. Targeting Energy Metabolism in Cancer Stem Cells: Progress and Challenges in Leukemia and Solid Tumors. Cell Stem Cell 2021, 28, 378–393. [Google Scholar] [CrossRef]
  121. Latini, S.; Venafra, V.; Massacci, G.; Bica, V.; Graziosi, S.; Pugliese, G.M.; Iannuccelli, M.; Frioni, F.; Minnella, G.; Marra, J.D.; et al. Unveiling the Signaling Network of FLT3-ITD AML Improves Drug Sensitivity Prediction. Elife 2024, 12, RP90532. [Google Scholar] [CrossRef]
  122. Chen, Y.; Zou, Z.; Găman, M.-A.; Xu, L.; Li, J. NADPH Oxidase Mediated Oxidative Stress Signaling in FLT3-ITD Acute Myeloid Leukemia. Cell Death Discov. 2023, 9, 208. [Google Scholar] [CrossRef]
  123. Sillar, J.R.; Germon, Z.P.; DeIuliis, G.N.; Dun, M.D. The Role of Reactive Oxygen Species in Acute Myeloid Leukaemia. Int. J. Mol. Sci. 2019, 20, 6003. [Google Scholar] [CrossRef]
  124. Starkov, A.A. The Role of Mitochondria in Reactive Oxygen Species Metabolism and Signaling. Ann. N. Y. Acad. Sci. 2008, 1147, 37–52. [Google Scholar] [CrossRef]
  125. Wise, D.R.; DeBerardinis, R.J.; Mancuso, A.; Sayed, N.; Zhang, X.-Y.; Pfeiffer, H.K.; Nissim, I.; Daikhin, E.; Yudkoff, M.; McMahon, S.B.; et al. Myc Regulates a Transcriptional Program That Stimulates Mitochondrial Glutaminolysis and Leads to Glutamine Addiction. Proc. Natl. Acad. Sci. USA 2008, 105, 18782–18787. [Google Scholar] [CrossRef]
  126. Pryde, K.R.; Hirst, J. Superoxide Is Produced by the Reduced Flavin in Mitochondrial Complex I. J. Biol. Chem. 2011, 286, 18056–18065. [Google Scholar] [CrossRef]
  127. Zhou, F.; Yin, Y.; Su, T.; Yu, L.; Yu, C.-A. Oxygen Dependent Electron Transfer in the Cytochrome Bc1 Complex. Biochim. Biophys. Acta (BBA) Bioenerg. 2012, 1817, 2103–2109. [Google Scholar] [CrossRef] [PubMed]
  128. Wang, Y.; Branicky, R.; Noë, A.; Hekimi, S. Superoxide Dismutases: Dual Roles in Controlling ROS Damage and Regulating ROS Signaling. J. Cell Biol. 2018, 217, 1915–1928. [Google Scholar] [CrossRef] [PubMed]
  129. Woolley, J.F.; Naughton, R.; Stanicka, J.; Gough, D.R.; Bhatt, L.; Dickinson, B.C.; Chang, C.J.; Cotter, T.G. H2O2 Production Downstream of FLT3 Is Mediated by P22phox in the Endoplasmic Reticulum and Is Required for STAT5 Signalling. PLoS ONE 2012, 7, e34050. [Google Scholar] [CrossRef] [PubMed]
  130. Lagunas-Rangel, F.A.; Linnea-Niemi, J.V.; Kudłak, B.; Williams, M.J.; Jönsson, J.; Schiöth, H.B. Role of the Synergistic Interactions of Environmental Pollutants in the Development of Cancer. Geohealth 2022, 6, e2021GH000552. [Google Scholar] [CrossRef] [PubMed]
  131. Stanicka, J.; Russell, E.G.; Woolley, J.F.; Cotter, T.G. NADPH Oxidase-Generated Hydrogen Peroxide Induces DNA Damage in Mutant FLT3-Expressing Leukemia Cells. J. Biol. Chem. 2015, 290, 9348–9361. [Google Scholar] [CrossRef]
  132. Wang, X.-X.; Wei, J.-Z.; Jiao, J.; Jiang, S.-Y.; Yu, D.-H.; Li, D. Genome-Wide DNA Methylation and Gene Expression Patterns Provide Insight into Polycystic Ovary Syndrome Development. Oncotarget 2014, 5, 6603–6610. [Google Scholar] [CrossRef]
  133. Böhmer, A.; Barz, S.; Schwab, K.; Kolbe, U.; Gabel, A.; Kirkpatrick, J.; Ohlenschläger, O.; Görlach, M.; Böhmer, F.-D. Modulation of FLT3 Signal Transduction through Cytoplasmic Cysteine Residues Indicates the Potential for Redox Regulation. Redox Biol. 2020, 28, 101325. [Google Scholar] [CrossRef]
  134. Wu, M.; Li, L.; Hamaker, M.; Small, D.; Duffield, A.S. FLT3-ITD Cooperates with Rac1 to Modulate the Sensitivity of Leukemic Cells to Chemotherapeutic Agents via Regulation of DNA Repair Pathways. Haematologica 2019, 104, 2418–2428. [Google Scholar] [CrossRef]
  135. Cauchy, P.; James, S.R.; Zacarias-Cabeza, J.; Ptasinska, A.; Imperato, M.R.; Assi, S.A.; Piper, J.; Canestraro, M.; Hoogenkamp, M.; Raghavan, M.; et al. Chronic FLT3-ITD Signaling in Acute Myeloid Leukemia Is Connected to a Specific Chromatin Signature. Cell Rep. 2015, 12, 821–836. [Google Scholar] [CrossRef] [PubMed]
  136. Sallmyr, A.; Fan, J.; Datta, K.; Kim, K.-T.; Grosu, D.; Shapiro, P.; Small, D.; Rassool, F. Internal Tandem Duplication of FLT3 (FLT3/ITD) Induces Increased ROS Production, DNA Damage, and Misrepair: Implications for Poor Prognosis in AML. Blood 2008, 111, 3173–3182. [Google Scholar] [CrossRef] [PubMed]
  137. Gallipoli, P.; Giotopoulos, G.; Tzelepis, K.; Costa, A.S.H.; Vohra, S.; Medina-Perez, P.; Basheer, F.; Marando, L.; Di Lisio, L.; Dias, J.M.L.; et al. Glutaminolysis Is a Metabolic Dependency in FLT3ITD Acute Myeloid Leukemia Unmasked by FLT3 Tyrosine Kinase Inhibition. Blood 2018, 131, 1639–1653. [Google Scholar] [CrossRef] [PubMed]
  138. Kannan, S.; Irwin, M.E.; Herbrich, S.M.; Cheng, T.; Patterson, L.L.; Aitken, M.J.L.; Bhalla, K.; You, M.J.; Konopleva, M.; Zweidler-McKay, P.A.; et al. Targeting the NRF2/HO-1 Antioxidant Pathway in FLT3-ITD-Positive AML Enhances Therapy Efficacy. Antioxidants 2022, 11, 717. [Google Scholar] [CrossRef]
  139. Rushworth, S.A.; Zaitseva, L.; Langa, S.; Bowles, K.M.; MacEwan, D.J. FLIP Regulation of HO-1 and TNF Signalling in Human Acute Myeloid Leukemia Provides a Unique Secondary Anti-Apoptotic Mechanism. Oncotarget 2010, 1, 359–366. [Google Scholar] [CrossRef]
  140. Hasan, S.K.; Jayakumar, S.; Espina Barroso, E.; Jha, A.; Catalano, G.; Sandur, S.K.; Noguera, N.I. Molecular Targets of Oxidative Stress: Focus on Nuclear Factor Erythroid 2–Related Factor 2 Function in Leukemia and Other Cancers. Cells 2025, 14, 713. [Google Scholar] [CrossRef]
  141. Yagishita, Y.; Gatbonton-Schwager, T.N.; McCallum, M.L.; Kensler, T.W. Current Landscape of NRF2 Biomarkers in Clinical Trials. Antioxidants 2020, 9, 716. [Google Scholar] [CrossRef]
  142. Zhang, D.; Hou, Z.; Aldrich, K.E.; Lockwood, L.; Odom, A.L.; Liby, K.T. A Novel Nrf2 Pathway Inhibitor Sensitizes Keap1-Mutant Lung Cancer Cells to Chemotherapy. Mol. Cancer Ther. 2021, 20, 1692–1701. [Google Scholar] [CrossRef]
  143. Pearson, K.J.; Lewis, K.N.; Price, N.L.; Chang, J.W.; Perez, E.; Cascajo, M.V.; Tamashiro, K.L.; Poosala, S.; Csiszar, A.; Ungvari, Z.; et al. Nrf2 Mediates Cancer Protection but Not Prolongevity Induced by Caloric Restriction. Proc. Natl. Acad. Sci. USA 2008, 105, 2325–2330. [Google Scholar] [CrossRef]
  144. Jones, C.L.; Stevens, B.M.; D’Alessandro, A.; Culp-Hill, R.; Reisz, J.A.; Pei, S.; Gustafson, A.; Khan, N.; DeGregori, J.; Pollyea, D.A.; et al. Cysteine Depletion Targets Leukemia Stem Cells through Inhibition of Electron Transport Complex II. Blood 2019, 134, 389–394. [Google Scholar] [CrossRef]
  145. Ryl, T.; Kuchen, E.E.; Bell, E.; Shao, C.; Flórez, A.F.; Mönke, G.; Gogolin, S.; Friedrich, M.; Lamprecht, F.; Westermann, F.; et al. Cell-Cycle Position of Single MYC-Driven Cancer Cells Dictates Their Susceptibility to a Chemotherapeutic Drug. Cell Syst. 2017, 5, 237–250.e8. [Google Scholar] [CrossRef]
  146. Blagosklonny, M. V Target for Cancer Therapy: Proliferating Cells or Stem Cells. Leukemia 2006, 20, 385–391. [Google Scholar] [CrossRef]
  147. Wang, B.; Pei, J.; Xu, S.; Liu, J.; Yu, J. A Glutamine Tug-of-War between Cancer and Immune Cells: Recent Advances in Unraveling the Ongoing Battle. J. Exp. Clin. Cancer Res. 2024, 43, 74. [Google Scholar] [CrossRef]
  148. Matés, J.M.; Di Paola, F.J.; Campos-Sandoval, J.A.; Mazurek, S.; Márquez, J. Therapeutic Targeting of Glutaminolysis as an Essential Strategy to Combat Cancer. Semin. Cell Dev. Biol. 2020, 98, 34–43. [Google Scholar] [CrossRef] [PubMed]
  149. Shen, Y.-A.; Chen, C.-L.; Huang, Y.-H.; Evans, E.E.; Cheng, C.-C.; Chuang, Y.-J.; Zhang, C.; Le, A. Inhibition of Glutaminolysis in Combination with Other Therapies to Improve Cancer Treatment. Curr. Opin. Chem. Biol. 2021, 62, 64–81. [Google Scholar] [CrossRef] [PubMed]
  150. Akins, N.S.; Nielson, T.C.; Le, H.V. Inhibition of Glycolysis and Glutaminolysis: An Emerging Drug Discovery Approach to Combat Cancer. Curr. Top. Med. Chem. 2018, 18, 494–504. [Google Scholar] [CrossRef] [PubMed]
  151. Fan, Y.; Xue, H.; Li, Z.; Huo, M.; Gao, H.; Guan, X. Exploiting the Achilles’ Heel of Cancer: Disrupting Glutamine Metabolism for Effective Cancer Treatment. Front. Pharmacol. 2024, 15, 1345522. [Google Scholar] [CrossRef]
  152. Stine, Z.E.; Schug, Z.T.; Salvino, J.M.; Dang, C.V. Targeting Cancer Metabolism in the Era of Precision Oncology. Nat. Rev. Drug Discov. 2022, 21, 141–162. [Google Scholar] [CrossRef] [PubMed]
  153. Koch, K.; Hartmann, R.; Tsiampali, J.; Uhlmann, C.; Nickel, A.-C.; He, X.; Kamp, M.A.; Sabel, M.; Barker, R.A.; Steiger, H.-J.; et al. A Comparative Pharmaco-Metabolomic Study of Glutaminase Inhibitors in Glioma Stem-like Cells Confirms Biological Effectiveness but Reveals Differences in Target-Specificity. Cell Death Discov. 2020, 6, 20. [Google Scholar] [CrossRef] [PubMed]
  154. Rattigan, K.M.; Zarou, M.M.; Helgason, V. Metabolism in Stem Cell Driven Leukaemia: Parallels between Haematopoiesis and Immunity. Blood 2023, 141, 2553–2565. [Google Scholar] [CrossRef]
  155. Appelbaum, F.R.; Gundacker, H.; Head, D.R.; Slovak, M.L.; Willman, C.L.; Godwin, J.E.; Anderson, J.E.; Petersdorf, S.H. Age and Acute Myeloid Leukemia. Blood 2006, 107, 3481–3485. [Google Scholar] [CrossRef]
  156. Juliusson, G.; Antunovic, P.; Derolf, A.; Lehmann, S.; Möllgård, L.; Stockelberg, D.; Tidefelt, U.; Wahlin, A.; Höglund, M. Age and Acute Myeloid Leukemia: Real World Data on Decision to Treat and Outcomes from the Swedish Acute Leukemia Registry. Blood 2009, 113, 4179–4187. [Google Scholar] [CrossRef]
  157. Norsworthy, K.J.; Luo, L.; Hsu, V.; Gudi, R.; Dorff, S.E.; Przepiorka, D.; Deisseroth, A.; Shen, Y.-L.; Sheth, C.M.; Charlab, R.; et al. FDA Approval Summary: Ivosidenib for Relapsed or Refractory Acute Myeloid Leukemia with an Isocitrate Dehydrogenase-1 Mutation. Clin. Cancer Res. 2019, 25, 3205–3209. [Google Scholar] [CrossRef]
  158. Kim, E.S. Enasidenib: First Global Approval. Drugs 2017, 77, 1705–1711. [Google Scholar] [CrossRef]
  159. Bross, P.F.; Beitz, J.; Chen, G.; Chen, X.H.; Duffy, E.; Kieffer, L.; Roy, S.; Sridhara, R.; Rahman, A.; Williams, G.; et al. Approval Summary: Gemtuzumab Ozogamicin in Relapsed Acute Myeloid Leukemia. Clin. Cancer Res. 2001, 7, 1490–1496. [Google Scholar]
  160. Krauss, A.C.; Gao, X.; Li, L.; Manning, M.L.; Patel, P.; Fu, W.; Janoria, K.G.; Gieser, G.; Bateman, D.A.; Przepiorka, D.; et al. FDA Approval Summary: (Daunorubicin and Cytarabine) Liposome for Injection for the Treatment of Adults with High-Risk Acute Myeloid Leukemia. Clin. Cancer Res. 2019, 25, 2685–2690. [Google Scholar] [CrossRef] [PubMed]
  161. Addanki, S.; Kim, L.; Stevens, A. Understanding and Targeting Metabolic Vulnerabilities in Acute Myeloid Leukemia: An Updated Comprehensive Review. Cancers 2025, 17, 1355. [Google Scholar] [CrossRef] [PubMed]
  162. Yeşlyurt, S.G.; Koyun, D.; Toprak, S.K.; Özcan, M.; Özen, C. A Predictive Metabolomic Model for FLT3 and NPM1 Mutations in Acute Myeloid Leukemia Patients. J. Pharm. Biomed. Anal. 2025, 260, 116789. [Google Scholar] [CrossRef]
  163. Hvinden, I.C.; Cadoux-Hudson, T.; Schofield, C.J.; McCullagh, J.S.O. Metabolic Adaptations in Cancers Expressing Isocitrate Dehydrogenase Mutations. Cell Rep. Med. 2021, 2, 100469. [Google Scholar] [CrossRef]
  164. Liu, J.; Zhang, C.; Hu, W.; Feng, Z. Tumor Suppressor P53 and Metabolism. J. Mol. Cell Biol. 2019, 11, 284–292. [Google Scholar] [CrossRef]
  165. Dai, Y.-J.; Hu, F.; He, S.-Y.; Tian, X.-P.; Li, H.-H.; Qin, Z.-Y.; Chen, S.; Liang, Y. A Distinct Metabolic Signature in DNMT3A-Mutated Leukemia. Blood 2019, 134, 1426. [Google Scholar] [CrossRef]
  166. Fujino, T.; Goyama, S.; Sugiura, Y.; Inoue, D.; Yamasaki, S.; Matsumoto, A.; Sato, N.; Morinaga, H.; Shikata, S.; Fukuyama, T.; et al. Mutant ASXL1 Promotes Expansion of the Phenotypic Hematopoietic Stem Cell Compartment. Blood 2019, 134, 821. [Google Scholar] [CrossRef]
  167. Hope, K.J.; Jin, L.; Dick, J.E. Acute Myeloid Leukemia Originates from a Hierarchy of Leukemic Stem Cell Classes That Differ in Self-Renewal Capacity. Nat. Immunol. 2004, 5, 738–743. [Google Scholar] [CrossRef] [PubMed]
  168. Wang, A.; Zhong, H. Roles of the Bone Marrow Niche in Hematopoiesis, Leukemogenesis, and Chemotherapy Resistance in Acute Myeloid Leukemia. Hematology 2018, 23, 729–739. [Google Scholar] [CrossRef] [PubMed]
  169. Salvia, A.M.; Cuviello, F.; Coluzzi, S.; Nuccorini, R.; Attolico, I.; Pascale, S.P.; Bisaccia, F.; Pizzuti, M.; Ostuni, A. Expression of Some ATP-Binding Cassette Transporters in Acute Myeloid Leukemia. Hematol. Rep. 2017, 9, 7406. [Google Scholar] [CrossRef]
  170. Stevens, A.M.; Schafer, E.S.; Li, M.; Terrell, M.; Rashid, R.; Paek, H.; Bernhardt, M.B.; Weisnicht, A.; Smith, W.T.; Keogh, N.J.; et al. Repurposing Atovaquone as a Therapeutic against Acute Myeloid Leukemia (AML): Combination with Conventional Chemotherapy Is Feasible and Well Tolerated. Cancers 2023, 15, 1344. [Google Scholar] [CrossRef] [PubMed]
  171. Lee, E.A.; Angka, L.; Rota, S.-G.; Hanlon, T.; Mitchell, A.; Hurren, R.; Wang, X.M.; Gronda, M.; Boyaci, E.; Bojko, B.; et al. Targeting Mitochondria with Avocatin B Induces Selective Leukemia Cell Death. Cancer Res. 2015, 75, 2478–2488. [Google Scholar] [CrossRef]
  172. Tan, S.-F.; Liu, X.; Fox, T.E.; Barth, B.M.; Sharma, A.; Turner, S.D.; Awwad, A.; Dewey, A.; Doi, K.; Spitzer, B.; et al. Acid Ceramidase Is Upregulated in AML and Represents a Novel Therapeutic Target. Oncotarget 2016, 7, 83208–83222. [Google Scholar] [CrossRef]
  173. Yap, T.A.; Daver, N.; Mahendra, M.; Zhang, J.; Kamiya-Matsuoka, C.; Meric-Bernstam, F.; Kantarjian, H.M.; Ravandi, F.; Collins, M.E.; Francesco, M.E.D.; et al. Complex I Inhibitor of Oxidative Phosphorylation in Advanced Solid Tumors and Acute Myeloid Leukemia: Phase I Trials. Nat. Med. 2023, 29, 115–126. [Google Scholar] [CrossRef]
  174. Reed, G.A.; Schiller, G.J.; Kambhampati, S.; Tallman, M.S.; Douer, D.; Minden, M.D.; Yee, K.W.; Gupta, V.; Brandwein, J.; Jitkova, Y.; et al. A Phase 1 Study of Intravenous Infusions of Tigecycline in Patients with Acute Myeloid Leukemia. Cancer Med. 2016, 5, 3031–3040. [Google Scholar] [CrossRef]
  175. Mussai, F.; Egan, S.; Higginbotham-Jones, J.; Perry, T.; Beggs, A.; Odintsova, E.; Loke, J.; Pratt, G.; U, K.P.; Lo, A.; et al. Arginine Dependence of Acute Myeloid Leukemia Blast Proliferation: A Novel Therapeutic Target. Blood 2015, 125, 2386–2396. [Google Scholar] [CrossRef]
  176. Tsai, H.-J.; Jiang, S.S.; Hung, W.-C.; Borthakur, G.; Lin, S.-F.; Pemmaraju, N.; Jabbour, E.; Bomalaski, J.S.; Chen, Y.-P.; Hsiao, H.-H.; et al. A Phase II Study of Arginine Deiminase (ADI-PEG20) in Relapsed/Refractory or Poor-Risk Acute Myeloid Leukemia Patients. Sci. Rep. 2017, 7, 11253. [Google Scholar] [CrossRef] [PubMed]
  177. Senapati, J.; Kantarjian, H.M.; Bazinet, A.; Reville, P.; Short, N.J.; Daver, N.; Borthakur, G.; Bataller, A.; Jabbour, E.; DiNardo, C.; et al. Lower Intensity Therapy with Cladribine/Low Dose Cytarabine/Venetoclax in Older Patients with Acute Myeloid Leukemia Compares Favorably with Intensive Chemotherapy among Patients Undergoing Allogeneic Stem Cell Transplantation. Cancer 2024, 130, 3333–3343. [Google Scholar] [CrossRef] [PubMed]
  178. DiNardo, C.D.; Jonas, B.A.; Pullarkat, V.; Thirman, M.J.; Garcia, J.S.; Wei, A.H.; Konopleva, M.; Döhner, H.; Letai, A.; Fenaux, P.; et al. Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N. Engl. J. Med. 2020, 383, 617–629. [Google Scholar] [CrossRef] [PubMed]
  179. Fathi, A.T.; Braun, T.P.; Ambinder, A.J.; Borthakur, G.; Redner, R.L.; Arevalo, M.; Gutierrez, S.; Limon, A.; Faller, D.V. Iadademstat and Gilteritinib for the Treatment of FLT3-Mutated Relapsed/Refractory Acute Myeloid Leukemia: The Frida Study. Blood 2023, 142, 5974. [Google Scholar] [CrossRef]
  180. Vidal, R.S.; Quarti, J.; Rumjanek, F.D.; Rumjanek, V.M. Metabolic Reprogramming During Multidrug Resistance in Leukemias. Front. Oncol. 2018, 8, 90. [Google Scholar] [CrossRef]
  181. Farge, T.; Saland, E.; Toni, F.D.; Aroua, N.; Hosseini, M.; Perry, R.; Bosc, C.; Sugita, M.; Stuani, L.; Fraisse, M.; et al. Chemotherapy-Resistant Human Acute Myeloid Leukemia Cells Are Not Enriched for Leukemic Stem Cells but Require Oxidative Metabolism. Cancer Discov. 2017, 7, 716–735. [Google Scholar] [CrossRef]
  182. Moschoi, R.; Imbert, V.; Nebout, M.; Chiche, J.; Mary, D.; Prebet, T.; Saland, E.; Castellano, R.; Pouyet, L.; Collette, Y.; et al. Protective Mitochondrial Transfer from Bone Marrow Stromal Cells to Acute Myeloid Leukemic Cells during Chemotherapy. Blood 2016, 128, 253–264. [Google Scholar] [CrossRef] [PubMed]
  183. Gregory, M.A.; D’Alessandro, A.; Alvarez-Calderon, F.; Kim, J.; Nemkov, T.; Adane, B.; Rozhok, A.I.; Kumar, A.; Kumar, V.; Pollyea, D.A.; et al. ATM/G6PD-Driven Redox Metabolism Promotes FLT3 Inhibitor Resistance in Acute Myeloid Leukemia. Proc. Natl. Acad. Sci. USA 2016, 113, E6669–E6678. [Google Scholar] [CrossRef]
  184. Lu, X.; Han, L.; Busquets, J.; Collins, M.; Lodi, A.; Marszalek, J.R.; Konopleva, M.; Tiziani, S. The Combined Treatment with the FLT3-Inhibitor AC220 and the Complex I Inhibitor IACS-010759 Synergistically Depletes Wt- and FLT3-Mutated Acute Myeloid Leukemia Cells. Front. Oncol. 2021, 11, 686765. [Google Scholar] [CrossRef]
  185. Taylor, S.J.; Steidl, U. Metabolic StrugGLS after FLT3 Inhibition in AML. Blood 2018, 131, 1631–1632. [Google Scholar] [CrossRef]
  186. Sharma, P.; Borthakur, G. Targeting Metabolic Vulnerabilities to Overcome Resistance to Therapy in Acute Myeloid Leukemia. Cancer Drug Resist. 2023, 6, 567–589. [Google Scholar] [CrossRef] [PubMed]
  187. Pollyea, D.A.; Stevens, B.M.; Jones, C.L.; Winters, A.; Pei, S.; Minhajuddin, M.; D’Alessandro, A.; Culp-Hill, R.; Riemondy, K.A.; Gillen, A.E.; et al. Venetoclax with Azacitidine Disrupts Energy Metabolism and Targets Leukemia Stem Cells in Patients with Acute Myeloid Leukemia. Nat. Med. 2018, 24, 1859–1866. [Google Scholar] [CrossRef]
  188. Zhang, Y.; Luo, T.; Ding, X.; Chang, Y.; Liu, C.; Zhang, Y.; Hao, S.; Yin, Q.; Jiang, B. Inhibition of Mitochondrial Complex III Induces Differentiation in Acute Myeloid Leukemia. Biochem. Biophys. Res. Commun. 2021, 547, 162–168. [Google Scholar] [CrossRef]
  189. Alvarez-Calderon, F.; Gregory, M.A.; Pham-Danis, C.; DeRyckere, D.; Stevens, B.M.; Zaberezhnyy, V.; Hill, A.A.; Gemta, L.; Kumar, A.; Kumar, V.; et al. Tyrosine Kinase Inhibition in Leukemia Induces an Altered Metabolic State Sensitive to Mitochondrial Perturbations. Clin. Cancer Res. 2015, 21, 1360–1372. [Google Scholar] [CrossRef]
  190. Zhang, Y.; Zhou, F.; Guan, J.; Zhou, L.; Chen, B. Action Mechanism of Metformin and Its Application in Hematological Malignancy Treatments: A Review. Biomolecules 2023, 13, 250. [Google Scholar] [CrossRef] [PubMed]
  191. Wang, F.; Liu, Z.; Zeng, J.; Zhu, H.; Li, J.; Cheng, X.; Jiang, T.; Zhang, L.; Zhang, C.; Chen, T.; et al. Metformin Synergistically Sensitizes FLT3-ITD-Positive Acute Myeloid Leukemia to Sorafenib by Promoting MTOR-Mediated Apoptosis and Autophagy. Leuk. Res. 2015, 39, 1421–1427. [Google Scholar] [CrossRef]
  192. Li, Y.; Zeng, P.; Xiao, J.; Huang, P.; Liu, P. Modulation of Energy Metabolism to Overcome Drug Resistance in Chronic Myeloid Leukemia Cells through Induction of Autophagy. Cell Death Discov. 2022, 8, 212. [Google Scholar] [CrossRef]
  193. Sobhakumari, A.; Orcutt, K.P.; Love-Homan, L.; Kowalski, C.E.; Parsons, A.D.; Knudson, C.M.; Simons, A.L. 2-Deoxy-d-Glucose Suppresses the In Vivo Antitumor Efficacy of Erlotinib in Head and Neck Squamous Cell Carcinoma Cells. Oncol. Res. 2016, 24, 55–64. [Google Scholar] [CrossRef]
  194. Bjelosevic, S.; Gruber, E.; Newbold, A.; Shembrey, C.; Devlin, J.R.; Hogg, S.J.; Kats, L.; Todorovski, I.; Fan, Z.; Abrehart, T.C.; et al. Serine Biosynthesis Is a Metabolic Vulnerability in FLT3-ITD-Driven Acute Myeloid Leukemia. Cancer Discov. 2021, 11, 1582–1599. [Google Scholar] [CrossRef] [PubMed]
  195. Janssen, M.; Schmidt, C.; Bruch, P.-M.; Blank, M.F.; Rohde, C.; Waclawiczek, A.; Heid, D.; Renders, S.; Göllner, S.; Vierbaum, L.; et al. Venetoclax Synergizes with Gilteritinib in FLT3 Wild-Type High-Risk Acute Myeloid Leukemia by Suppressing MCL-1. Blood 2022, 140, 2594–2610. [Google Scholar] [CrossRef]
  196. Milnerowicz, S.; Maszewska, J.; Skowera, P.; Stelmach, M.; Lejman, M. AML under the Scope: Current Strategies and Treatment Involving FLT3 Inhibitors and Venetoclax-Based Regimens. Int. J. Mol. Sci. 2023, 24, 15849. [Google Scholar] [CrossRef] [PubMed]
  197. Zhou, F.-J.; Zeng, C.-X.; Kuang, W.; Cheng, C.; Liu, H.-C.; Yan, X.-Y.; Chen, X.-P.; Zhou, G.; Cao, S. Metformin Exerts a Synergistic Effect with Venetoclax by Downregulating Mcl-1 Protein in Acute Myeloid Leukemia. J. Cancer 2021, 12, 6727–6739. [Google Scholar] [CrossRef] [PubMed]
  198. Zhao, Y.; Zhang, X.; Ding, X.; Wang, Y.; Li, Z.; Zhao, R.; Cheng, H.-E.; Sun, Y. Efficacy and Safety of FLT3 Inhibitors in Monotherapy of Hematological and Solid Malignancies: A Systemic Analysis of Clinical Trials. Front. Pharmacol. 2024, 15, 1294668. [Google Scholar] [CrossRef]
  199. Perrone, S.; Ottone, T.; Zhdanovskaya, N.; Molica, M. How Acute Myeloid Leukemia (AML) Escapes from FMS-Related Tyrosine Kinase 3 (FLT3) Inhibitors? Still an Overrated Complication? Cancer Drug Resist. 2023, 6, 223–238. [Google Scholar] [CrossRef]
Figure 1. Metabolic dependencies in FLT3-ITD-mutated AML. Overview of key altered pathways supporting leukemic cell survival and proliferation.
Figure 1. Metabolic dependencies in FLT3-ITD-mutated AML. Overview of key altered pathways supporting leukemic cell survival and proliferation.
Jpm 15 00431 g001
Figure 2. Mitochondrial bioenergetics in FLT3-ITD AML. Schematic representation of mitochondrial metabolism highlighting OXPHOS as a central hub integrating glycolysis, TCA cycle, fatty acid oxidation, and purine synthesis in FLT3-ITD-mutated AML cells. Low spare respiratory capacity indicates mitochondrial vulnerability.
Figure 2. Mitochondrial bioenergetics in FLT3-ITD AML. Schematic representation of mitochondrial metabolism highlighting OXPHOS as a central hub integrating glycolysis, TCA cycle, fatty acid oxidation, and purine synthesis in FLT3-ITD-mutated AML cells. Low spare respiratory capacity indicates mitochondrial vulnerability.
Jpm 15 00431 g002
Table 1. Emerging therapeutic approaches targeting metabolism in AML.
Table 1. Emerging therapeutic approaches targeting metabolism in AML.
DrugMetabolic TargetTrialReferences
2-Deoxy-D-glucose (2-DG)GlycolysisPre-clinical (in vitro and in vivo)[68]
AtovaquoneOXPHOSPre-clinical (in vitro and in vivo) Feasibility Trial (NCT03568994)[170]
Avocatin B, Etomoxir and ST-1326Fatty acid oxidationPre-clinical (in vitro and in vivo)[57,92,171]
3-bromopyruvate (3BrPA)GlycolysisPre-clinical (in vitro and in vivo)[66]
LCL204SphingolipidsPre-clinical (in vivo)[172]
IACS-010759OXPHOSPhase I (NCT02882321)[173]
Telaglenastat (CB-839)GlutaminolysisPhase I (NCT02071927)[174]
TigecyclineOXPHOSPhase I (NCT01332786)[175]
BCT-100 Arginine metabolismPhase I/II (NCT03455140)[175]
ADI-PEG 20Arginine metabolismPhase II trial (NCT01910012)[176]
VenetoclaxOXPHOSPhase I/II/III (NCT01994837,
NCT02203773,
NCT02993523, NCT04801779, NCT05177731, NCT05048615, NCT03586609, NCT03625505,
NCT04140487
NCT03455504)
[177,178]
IadademstatOXPHOSNCT05546580[179]
Table 2. Metabolic-oriented therapeutic strategies in FLT3-ITD-mutated AML.
Table 2. Metabolic-oriented therapeutic strategies in FLT3-ITD-mutated AML.
Drug/CombinationTargeted Metabolic PathwayMechanism of ActionEffectReferences
Quizartinib (1–5 nM) +
CB-839
GlutaminolysisInhibits glutamine metabolism; increased
ROS and mitochondrial dysfunction
Synergistic lethality;
overcomes resistance mechanisms
(in vitro and in vivo)
[79]
Quizartinib (100 nM) +
IACS-010759 (10 nM)
OXPHOS
(Complex I inhibition)
Inhibits mitochondrial electron transport chain Complex IEnhances sensitivity to FLT3i;
impairs energy production
(in vitro)
[184]
Venetoclax (100 nM in vitro/100 mg/kg in vivo/400 mg/die in clinical trial) +
Azacitidine (1 µM in vitro/3 mg/kg in vivo/75 mg/7 days in clinical trial)
Mitochondrial metabolism,
Glutathione synthesis
Impairs OXPHOS,
reduces GSH levels,
inhibits Bcl-2
Targets LSCs; induces apoptosis; synergizes with FLT3i via MCL-1 suppression
(in vitro, in vivo, and clinical trial)
[187]
Antimycin A
(1 µM)
OXPHOS (Complex III inhibition)Blocks electron flow in ETC Complex IIIReduces cell viability; promotes differentiation
(in vitro)
[188]
Oligomycin A
(0.5–10 nM in vitro/100 µg/kg in vivo)
OXPHOS (Complex V/ATP synthase inhibition)Inhibits ATP productionEnhances sensitivity to FLT3 inhibitors
(in vitro and in vivo)
[189]
Metformin
(0.5–10 mM in vitro/200 mg/kg in vivo)
Mitochondrial metabolism/mTOR pathwayInhibits OXPHOS and glycolysis; activates AMPKPromotes apoptosis; synergizes with FLT3i and venetoclax
(in vitro and in vivo)
[190,191]
Quizartinib + L-Asparaginase
(2 UI/mL)
Amino acid metabolism (glutamine/asparagine depletion)Depletes glutamine/asparagine; metabolic stressEffective in quizartinib-resistant FLT3+ cells
(in vitro)
[79]
Crenolanib (6 µM in vitro/15 mg/Kg/die in vivo) + MP-A08 (SPHK1 inhibitor, 6–20 µM/100 mg/kg)Sphingolipid metabolism (ceramide pathway)Induces ceramide accumulation; activates stress/apoptotic signalingPromotes mitophagy and apoptosis; sensitizes to venetoclax[110]
2-Deoxy-D-glucose (2-DG, 20 mM)GlycolysisInhibits hexokinase; blocks glucose utilizationEnhances cytotoxic effects of TKIs
(in vitro and in vivo)
[192,193]
WQ-2101
(PHGDH inhibitor)
Serine synthesisInhibits de novo serine synthesis pathwayEnhances chemotherapy response in FLT3-mut AML
(in vitro and in vivo)
[194]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Banella, C.; Catalano, G.; Calvani, M.; Candi, E.; Noguera, N.I.; Travaglini, S. Metabolic Signature of FLT3-Mutated AML: Clinical and Therapeutic Implications. J. Pers. Med. 2025, 15, 431. https://doi.org/10.3390/jpm15090431

AMA Style

Banella C, Catalano G, Calvani M, Candi E, Noguera NI, Travaglini S. Metabolic Signature of FLT3-Mutated AML: Clinical and Therapeutic Implications. Journal of Personalized Medicine. 2025; 15(9):431. https://doi.org/10.3390/jpm15090431

Chicago/Turabian Style

Banella, Cristina, Gianfranco Catalano, Maura Calvani, Eleonora Candi, Nelida Ines Noguera, and Serena Travaglini. 2025. "Metabolic Signature of FLT3-Mutated AML: Clinical and Therapeutic Implications" Journal of Personalized Medicine 15, no. 9: 431. https://doi.org/10.3390/jpm15090431

APA Style

Banella, C., Catalano, G., Calvani, M., Candi, E., Noguera, N. I., & Travaglini, S. (2025). Metabolic Signature of FLT3-Mutated AML: Clinical and Therapeutic Implications. Journal of Personalized Medicine, 15(9), 431. https://doi.org/10.3390/jpm15090431

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop