Next Article in Journal
Immunological Mechanisms and Effects of Bacterial Infections in Acute-on-Chronic Liver Failure
Previous Article in Journal
Inverse Bicontinuous and Discontinuous Phases of Lipids, and Membrane Curvature
Previous Article in Special Issue
Combined Transplantation of Mesenchymal Progenitor and Neural Stem Cells to Repair Cervical Spinal Cord Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Metabolic Landscape of Cancer Stem Cells: Insights and Implications for Therapy

by
Martina Milella
1,†,
Monica Rutigliano
1,†,
Savio Domenico Pandolfo
2,3,
Achille Aveta
2,
Felice Crocetto
2,
Matteo Ferro
4,
Antonio d’Amati
1,
Pasquale Ditonno
1,
Giuseppe Lucarelli
1,5,‡ and
Francesco Lasorsa
1,*,‡
1
Urology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area-Urology, University of Bari “Aldo Moro”, 70124 Bari, Italy
2
Department of Urology, University of L’Aquila, 67100 L’Aquila, Italy
3
Department of Neurosciences, Science of Reproduction and Odontostomatology, Federico II University, 80138 Naples, Italy
4
Urology Unit, Department of Health Science, University of Milan, 20122 Milan, Italy
5
SSD Urologia Clinicizzata, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work.
Cells 2025, 14(10), 717; https://doi.org/10.3390/cells14100717
Submission received: 5 April 2025 / Revised: 6 May 2025 / Accepted: 14 May 2025 / Published: 15 May 2025
(This article belongs to the Special Issue Stem Cell, Differentiation, Regeneration and Diseases)

Abstract

:
Cancer stem cells (CSCs) are a subpopulation with self-renewal and differentiation capacities believed to be responsible for tumor initiation, progression, and recurrence. These cells exhibit unique metabolic features that contribute to their stemness and survival in hostile tumor microenvironments. Like non-stem cancer cells, CSCs primarily rely on glycolysis for ATP production, akin to the Warburg effect. However, CSCs also show increased dependence on alternative metabolic pathways, such as oxidative phosphorylation (OXPHOS) and fatty acid metabolism, which provide necessary energy and building blocks for self-renewal and therapy resistance. The metabolic plasticity of CSCs enables them to adapt to fluctuating nutrient availability and hypoxic conditions within the tumor. Recent studies highlight the importance of these metabolic shifts in maintaining the CSC phenotype and promoting cancer progression. The CSC model suggests that a small, metabolically adaptable subpopulation drives tumor growth and therapy resistance. CSCs can switch between glycolysis and mitochondrial metabolism, enhancing their survival under stress and dormant states. Targeting CSC metabolism offers a promising therapeutic strategy; however, their adaptability complicates eradication. A multi-targeted approach addressing various metabolic pathways is essential for effective CSC elimination, underscoring the need for further research into specific CSC markers and mechanisms that distinguish their metabolism from normal stem cells for successful therapeutic intervention.

1. Introduction

Cancer stem cells (CSCs) are a subpopulation of cells with the ability to self-renew and differentiate and are thought to be responsible for tumor initiation, progression, and recurrence. These cells exhibit unique metabolic characteristics that contribute to their stemness and survival in harsh tumor microenvironments [1]. The metabolism of CSCs is distinct from that of non-stem cancer cells, as well as normal tissue cells.
Several studies have shown that CSCs heavily rely on glycolysis for energy production, similar to the Warburg effect observed in non-stem cancer cells. This metabolic shift toward glycolysis allows them to efficiently generate adenosine triphosphate (ATP) and sustain their rapid proliferation. In addition to glycolysis, CSCs also exhibit an increased reliance on other metabolic pathways, such as oxidative phosphorylation (OXPHOS) and fatty acid metabolism. These alternative metabolic pathways provide them with the necessary building blocks and energy sources to support their self-renewal and resistance to therapies [2]. The metabolic plasticity of these cells allows them to adapt to changing nutrient availability and hypoxic conditions in the tumor microenvironment.
In this review, we describe recent evidence about CSC metabolism and its role in cancer progression.

2. CSC Characteristics

While normal stem cells assist in the differentiation of normal tissues, cancer tissues are made up of a hierarchical structure of differentiated cells derived from cancer stem cells [3,4,5,6]. Cancer is defined as the uncontrolled proliferation of abnormal cells with varying structures and functions [7,8,9,10,11]. Two primary hypotheses have been proposed to explain the cellular diversity found in tumors. The first is the classic stochastic model, which describes cancer genesis and development as the gradual accumulation of mutations. The second approach, known as the cancer stem cell hypothesis, suggests that tumor growth is driven by a limited population of stem cells [12,13]. The presence of cancer stem cells may play a significant role in treatment resistance and recurrence. Recent research has shown that CSCs undergo metabolic changes that set them apart from non-CSCs, raising the possibility of developing curative therapies for CSCs by targeting specific metabolic pathways. However, there is debate over whether CSCs rely more on glycolysis or mitochondrial OXPHOS to maintain their stem cell characteristics [14,15,16]. CSCs have a notable migratory potential, allowing them to spread from the primary tumor to distant organs [17,18]. Various methods have been developed to isolate and characterize CSCs. They are typically found in specialized niches where they depend on specific growth factors like epidermal growth factor (EGF) and fibroblast growth factor (bFGF) to maintain their undifferentiated state and support self-renewal [19,20]. In addition to their ability to self-renew in an undifferentiated state, CSCs share many common features with normal stem cells, including the expression of surface markers (CD44, CD133, aldehyde dehydrogenase), the activation of specific signaling pathways (Notch, Hedgehog, or Wnt, relative quiescence), and the capacity to actively repair DNA [8,18,19,20,21,22].
Depending on the type of cancer, CSCs can be identified by a variety of surface markers, such as CXCR4, ESA, and Nestin. For example, in pancreatic cancer, CSCs are identified by combinations like CD133+/CXCR4+, CD24+/CD44+, and other profiles, while in breast cancer, they express CD24−/low/CD44+ [23,24]. In glioblastoma, CSCs are characterized by CD133+/ABCG2+ [25,26]. CD44 appears to play a role in tumor metastasis and invasion by facilitating CSC attachment to the extracellular matrix, promoting tumor cell migration, and contributing to metastatic spread by binding to its ligand hyaluronic acid [27,28,29,30].
Previous investigations have emphasized the role of renal stem/progenitor cells in kidney regeneration following tubular damage. These cells, characterized by markers like CD133 and CD24, possess self-renewal capabilities and the potential to differentiate into various renal cell types, such as tubular cells and podocytes. While CD133+ progenitor cells isolated from renal tumors are not tumorigenic by themselves, they can enhance tumor development and angiogenesis when co-transplanted with malignant cells, indicating their contribution to cancer progression. Further research identified a subset of CD105+, CD133+, and CD24+ CSCs with mesenchymal properties, which exhibit resistance to chemotherapy and demonstrate self-renewal and multipotency. Markers such as CD44 and CXCR4 are also vital for RCC CSCs [31]. CD44 is involved in cell migration, adhesion, and signaling, activating pathways like TGFβ, MAPK, and PI3K/AKT that support tumorigenesis. Meanwhile, CXCR4 directs CSCs to metastatic sites by interacting with SDF1, promoting cancer spread. These CSCs are highly resistant to chemotherapy, mainly due to the activation of the Notch and Wnt/β-catenin pathways, contributing to survival and drug resistance. Furthermore, CTR2, a copper transporter, influences drug resistance by modulating the uptake of platinum-based chemotherapies [19]. This transporter regulates copper balance in the cell, which can impact the effectiveness of treatment. RCC CSCs are pivotal in tumor growth, metastasis, and chemoresistance, highlighting their potential as therapeutic targets.

3. Glucose Metabolism in CSCs

Glucose is a primary energy source for both CSCs and differentiated tumor cells, offering rapid energy but with lower efficiency (Figure 1). Under normal oxygen conditions, pyruvate from glycolysis is used in OXPHOS to produce ATP [32]. In an anaerobic environment, pyruvate is converted to lactate by lactate dehydrogenase-A (LDH-A) and exported via monocarboxylate transporters (MCT) [33]. While glycolysis provides rapid ATP, especially under hypoxic conditions, its energetic yield is much lower than OXPHOS. OXPHOS remains a key pathway for energy production necessary for tumor growth, even in tumor cells [34,35,36,37,38]. Apart from ATP production, pyruvate and other glycolytic intermediates are crucial for amino acid and lipid biosynthesis (via the tricarboxylic acid cycle, TCA, and acetyl-CoA production, respectively). This is essential for the rapid proliferation of metastatic cancer cells, which require both energy and biosynthetic precursors within a short time frame [39,40,41]. Elevated glucose levels and increased expression of glucose transporters promote glycolysis and are associated with enhanced tumor cell and CSC viability. Glucose transporters can be classified into two types according to their mechanism of action: GLUT transporters and sodium-dependent SGLT transporters, which operate against the gradient by coupling with sodium ions [42,43,44]. In CSCs, GLUTs and SGLTs play distinct but complementary roles in glucose uptake. GLUTs, particularly GLUT1, are primarily responsible for the passive transport of glucose into the cell, supporting the high glycolytic activity and metabolic demands of CSCs, especially in low-oxygen environments. They help maintain CSC properties like self-renewal and resistance to therapies. On the other hand, SGLTs actively transport glucose along with sodium ions, which is particularly useful when glucose levels are low outside the cell. This process ensures that CSCs can still take up glucose under nutrient-poor conditions, supporting their survival [36]. CSCs exhibit increased glucose uptake due to their reliance on high glycolytic activity and low OXPHOS [45]. Reducing glucose concentration or inhibiting GLUT1 via genetic knockdown or pharmacological agents limits stemness and spheroid formation in pancreatic, ovarian, and glioblastoma CSCs without compromising cell viability [46]. Even so, CSCs are more glycolytic than differentiated cancer cells in a variety of malignancies [47,48,49]. Upregulation of glycolytic genes occurs before the expression of pluripotency markers, indicating that switching from OXPHOS to glycolysis promotes stemness in CSCs rather than pluripotency [50]. CSCs have greater glucose absorption and, hence, lactate and ATP generation, and die when glycolysis is inhibited or glucose is depleted [51,52]. Glycolytic CSCs are observed in CD133+ osteosarcoma-initiating cells, glioblastoma cells, breast cells, and liver cancer cells [33,47,48,49,50,51,52,53,54,55].
Lactate plays a key role in maintaining stem cell characteristics by upregulating the transcription factor SP1 (Specificity Protein 1), which, in turn, enhances tumor aggressiveness, invasiveness, and immune evasion via sterol regulatory element-binding protein 1 (SREBP1) [56,57,58,59,60,61]. In CSCs, hypoxia-inducible factor-1 (HIF-1) shifts metabolism toward glycolysis while suppressing OXPHOS and the tricarboxylic acid cycle [62]. HIF-1 also reduces reactive oxygen species (ROS) production and induces the expression of GLUT, hexokinase 2, pyruvate kinase, and lactate dehydrogenase A while downregulating pyruvate dehydrogenase [52]. These metabolic adaptations support CSC self-renewal and pluripotency, contributing to resistance to therapies across various types of cancer [63,64]. Consequently, it can be proposed that the metabolic profiles of cancer stem cells may exhibit variability, influenced by both the tissue of origin and the specific metastatic niche.

Mitochondria in CSCs

Beyond serving as a primary ATP source for cancer cells, mitochondria are involved in regulating various signaling pathways. These include the release of cytochrome C to trigger apoptosis, ROS generation, and the synthesis of metabolites like acetyl-CoA, which plays a role in modulating protein acetylation [11,65,66,67]. Mitochondria are responsible for producing over 90% of cellular ATP via OXPHOS [68]. In contrast, mitochondrial dysfunction leads to the suppression of OXPHOS in most CSCs. When OXPHOS is upregulated, it is closely linked to processes like epithelial–mesenchymal transition (EMT) and tumor metastasis [69]. In environments with limited nutrients, oxidative stress stimulates tumor cells to rely on glycolysis for lactate and metabolite production, while OXPHOS continues to be the dominant pathway for cellular metabolism, fueling anabolic processes that drive tumor progression [70].
Recent investigations have demonstrated that CSCs from various tumor types, such as ovarian, cervical, lung, and glioma, exhibit an enhanced reliance on mitochondrial function and OXPHOS compared to their non-stem counterparts [20]. For instance, ovarian cancer-derived CSCs show increased expression of enzymes involved in OXPHOS and fatty acid metabolism [71]. Likewise, spheroids from ovarian and cervical carcinomas display a reprogrammed metabolic state, favoring the TCA cycle relative to non-CSCs [72]. In small-cell lung cancer, CSCs are more dependent on OXPHOS and mitochondrial activity than their non-stem cell counterparts [73]. Glioma-derived CSCs utilize less glucose and lactate while sustaining high ATP production via OXPHOS [74]. This pattern is also seen in CD133+ glioblastoma cells, where the overexpression of insulin-like growth factor 2 mRNA-binding protein 2 (IMP2) modulates mitochondrial dynamics and stemness markers such as CD133, SOX2, and NANOG [75]. Moreover, dormant cells surviving RAS ablation in pancreatic cancer show a similar reliance on mitochondrial function and OXPHOS instead of glycolysis to preserve stem cell properties [76]. A strong association between the metastatic potential of cancer cells and the transcriptional co-activator PGC-1α (PPARGC1A) has been reported [77]. PGC-1α has been found to play a pivotal role in regulating mitochondrial functions, including OXPHOS and mitochondrial biogenesis, which, in turn, contributes to enhanced cellular migration and invasion, as demonstrated in invasive breast cancer models. Additionally, PGC-1α overexpression has been observed in circulating tumor cells and breast CSCs, with suppression of its activity leading to reduced stem cell characteristics [78]. These findings highlight the essential role of mitochondrial integrity in sustaining CSC properties. Consequently, mitochondrial biogenesis has become recognized as a central feature of CSCs, which exhibit higher mitochondrial mass, increased membrane potential, augmented mitochondrial ROS production, and elevated oxygen consumption relative to differentiated tumor cells [79,80,81,82,83].
Consistent with these findings, mitochondrial activity and metabolic processes could play a key role in the spread of CSCs. Since preserving a functional mitochondrial network is critical for sustaining and propagating stem cell characteristics, targeting these organelles may offer a promising therapeutic approach to eliminate CSCs.

4. Lipid Metabolism in CSCs

Rapidly proliferating cancer cells have an increased requirement for lipids and cholesterol [84,85,86,87]. This need is fulfilled through enhanced uptake of external lipids and lipoproteins or, alternatively, by the activation of intracellular metabolic pathways that drive the synthesis of lipids and cholesterol, such as lipogenesis and the mevalonate pathway [88,89,90,91]. In this context, the lipid metabolism of CSCs is of particular interest. In cell membranes, lipids, particularly cholesterol and sphingolipids, form microdomains known as lipid rafts, which in cancer cells contain various signaling molecules and receptors involved in both oncogenic and apoptotic pathways [92]. Within the stem cell compartment, both hematopoietic stem cells and leukemia-initiating cells are highly reliant on fatty acid oxidation for their energy needs [93,94]. Interestingly, leukemia-initiating cells take advantage of the adipose tissue microenvironment to create conditions that support leukemic growth and resistance to chemotherapy. They promote lipolysis in gonadal fat, which leads to the release of free fatty acids that fuel fatty acid oxidation in leukemia-initiating cells, a process facilitated by upregulation of the fatty acid transporter CD36. A similar role for CD36 has also been observed in oral CSCs [95].
Lipid accumulation occurs in cancer cells in lipid droplets, likely originating from the endoplasmic reticulum or Golgi apparatus. Increased droplet levels are linked to tumor aggressiveness, and they have been detected in circulating tumor cells, with their quantification suggesting a prognostic tool for survival outcomes [96,97,98]. An elevated amount of droplets is a characteristic feature of CD133+ colorectal (CR)-CSCs, as demonstrated by Raman spectroscopy [99].
Unsaturated lipids are critical for the function and progression of various types of tumors, including breast, colon, renal, and prostate cancer [100,101]. Administering unsaturated fatty acids into mice with pre-established colorectal cancer increases specific subpopulations of CSCs, which enhances their stem-like traits and fosters both tumor growth and metastasis [102]. One key player in this process is stearoyl-CoA desaturase, an enzyme responsible for introducing double bonds into fatty acid chains [103]. This desaturation increases membrane fluidity, a crucial factor for maintaining the integrity and functions of cancer cells. In ovarian cancer, CSCs are particularly reliant on unsaturated lipids, which differentiate them from non-stem tumor cells. Inhibiting lipid desaturases in these cells reduces markers associated with stemness and prevents the initiation of tumors. This suggests that targeting lipid desaturation could serve as an effective strategy for limiting tumor growth and spread [104]. Lipid desaturation also plays a role in modifying membrane characteristics, which are key for cellular processes like division, migration, and signaling critical for the metastatic potential of cancer cells. Moreover, lipid desaturation interacts with several oncogenic pathways, including NF-kB, Wnt, and Hippo/YAP, all of which are involved in CSC regulation. These pathways are enhanced by unsaturated lipids, which promote the stabilization of important proteins like β-catenin and YAP, reinforcing CSC properties and survival [105]. Additionally, unsaturated fatty acids can trigger mesenchymal stem cells to release angiogenic factors such as IL-6, VEGF, and nitric oxide, further facilitating tumor blood vessel formation and metastasis. Lipid desaturation significantly influences the maintenance and function of CSCs, playing a crucial role in tumor progression and metastasis. Targeting lipid desaturases may provide a promising therapeutic approach for inhibiting tumor growth and reducing the risk of cancer recurrence and metastasis.

4.1. Key Lipid Regulators

Stearoyl-CoA desaturase 1 (SCD1) is an enzyme that converts saturated fatty acids into monounsaturated fatty acids (MUFAs) [106]. It is highly upregulated in various cancers and is associated with tumor progression and poor clinical outcomes. SCD1 plays a key role in the generation and maintenance of stem cell properties in CSCs and tumor-initiating cells (TICs), particularly in liver, breast, and ovarian cancers [107,108,109,110]. Moreover, SCD1 overexpression promotes CSC proliferation and inhibits apoptosis [111]. The increased activity of SCD1 and subsequent MUFA production are emerging as defining characteristics of CSCs.
Sterol regulatory element-binding protein 1 (SREBP1) is a critical transcription factor involved in the synthesis of fatty acids and cholesterol [112]. It is a central regulator of lipogenesis, influencing the expression of key enzymes like ATP citrate lyase (ACLY), acetyl-CoA carboxylase (ACC1), and fatty acid synthase (FASN) [113]. Overexpression of SREBP1 has been shown to support tumor proliferation and sustain the stem-like properties of CSCs [81]. Additionally, SREBP1 can upregulate the expression of SCD1 [55,93]. This dual role of SREBP1 in regulating lipid metabolism and supporting CSCs highlights its significance in both cancer progression and stem cell biology [114,115].
HMG-CoA reductase (HMG-CoAR) is the rate-limiting enzyme in the mevalonate pathway that is crucial for cholesterol, steroid hormones, and non-sterol isoprenoid synthesis. This enzyme is also the primary target of statins, commonly used to reduce serum cholesterol levels [116,117,118,119]. The pathway generates intermediates like farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (GGPP), which are critical for protein prenylation, a process that ensures the proper membrane attachment of small GTPases from the Ras and Rho families. Interestingly, inhibition of this pathway by simvastatin has been shown to decrease the number of CSCs, which could have potential implications for future cancer therapy [120,121,122,123].

4.2. Lipid Droplets

Lipid droplets (LDs) are cellular organelles that store lipids and are surrounded by a phospholipid monolayer [124,125,126]. In cancer cells, LDs are more abundant compared to normal cells [127]. When glycolysis is inhibited, LD-derived free fatty acids (FFAs) contribute to ATP production via fatty acid oxidation (FAO). Lipophagy, a selective autophagic process, breaks down LDs and transfers FFAs to mitochondria, aiding cell survival under metabolic stress induced by disrupted oncogenic signaling [128,129]. Moreover, LDs protect against lipid peroxidation, which can trigger ferroptosis [130]. Lipid accumulation in prostate cancer correlates with tumor stage, serving as a diagnostic marker [131]. Hypoxia induces LD formation through HIF-mediated repression of CPT1A, an enzyme crucial for FAO [132]. Besides de novo lipogenesis, increased lipid uptake from the extracellular space also promotes LD accumulation and tumorigenic potential in CSCs [133]. CSCs from various cancers, such as colorectal and ovarian, have higher LD levels than bulk tumor cells. For example, colorectal cancer stem cells show more lipids than normal colon cells and other tumor cell types, with lipid content correlating to CD133 expression and Wnt pathway activation. In this context, CSCs with more LDs demonstrate higher clonogenic and tumorigenic abilities [84]. Similarly, ovarian CSCs (ALDH+/CD133+) have increased LD content compared to ALDH−/CD133− cells. Elevated LDs provide an energy reserve when glycolysis is blocked and protect fatty acids from oxidative damage, supporting CSC survival and proliferation [134]. Conversely, inhibition of phospholipase A2 reduces LDs and induces apoptosis in cancer cells [100].

5. Regulation of Amino Acids Metabolism in CSCs

Glutamine, a non-essential amino acid, is crucial for regulating energy balance and metabolic equilibrium during the proliferation of CSCs [135,136,137]. Glutamine regulates redox homeostasis by supporting the synthesis of glutathione (GSH), a key antioxidant in cells, and facilitating NADPH production in many CSC populations. NADPH provides reductive power for the regeneration of ROS-detoxifying enzymes and GSH. Disruption of glutamine metabolism has been shown to inhibit self-renewal, reduce the expression of stemness-associated genes, and decrease pluripotency factors by increasing intracellular ROS levels [138,139,140,141]. The importance of GSH-mediated redox control has been highlighted in certain stem-like cell populations within triple-negative breast cancer. These cells are distinguished by elevated levels of GD2 ganglioside, a novel marker linked to breast cancer stem cells, and they depend on ASCT2 for the import of glutamine [88,101,142]. However, the mechanisms through which glutamine metabolism regulates stem cell properties are not the same across various cancer models. For example, in embryonal carcinoma stem-like cells from both human and mouse sources, a regulatory network involving TAp73, Myc, and SLC1A5 coordinates glutamine uptake and glutathione biosynthesis [86]. Moreover, the synthesis of glutathione driven by glutaminase (GLS) also plays a significant role in the survival and radioresistance of prostate cancer CSCs [143]. Glutamine-derived NADPH is essential for the survival of pancreatic cancer cell lines, such as PANC-1 and SW1990, grown under cancer stem cell conditions. In these cells, glutamine deprivation reduces the expression of stemness-associated genes, impairs self-renewal, and prevents sphere formation [144,145]. Studies have revealed that glutamine depletion can indirectly impact cell signaling pathways through redox imbalance. Liao et al. demonstrated that in A549 non-small cell lung cancer (NSCLC) cells, glutamine starvation reduces the side population (SP) by ROS-induced inhibition of the β-catenin signaling pathway [146]. This results in lower expression of the ABCG2 transporter and the stemness gene SOX2. Additionally, glutamine deprivation increases ROS levels in both SP cells and glioblastoma stem-like cell lines (GSC11, GSC23), impairing their ability to form neurospheres. The increased ROS promotes the degradation of β-catenin and the downregulation of its target genes, Survivin and Axin2. In turn, in some cancer cell lines (epithelial ovarian cancer PA1, OAW42, and colorectal cancer HCT116), glutamine deprivation induces stemness through mitochondrial ROS activation, which triggers the MAPK pathway. This leads to the phosphorylation of dynamin-related protein-1 (DRP1), promoting mitochondrial fragmentation. The fragmented mitochondria increase local ROS, enhancing CSC characteristics, such as CD44 and CD117/CD45 positivity. Interestingly, this process does not involve reduced glutathione synthesis, as no significant changes in cellular GSH are observed [147]. In this scenario, we have to highlight that glutamine serves two main purposes in cancer cells: it helps replenish important TCA cycle intermediates, which are crucial for energy production, and it provides building blocks for making nucleotides essential for cell division [148]. However, in some cancers, like NSCLCs, as we described before, tumor cells may use glucose to replace some of the roles usually played by glutamine [149]. This suggests that in certain environments, cancer cells might be able to adjust their metabolism and use glucose as an alternative to glutamine. In glioblastoma, CSCs rely on both glucose and glutamine. Glucose helps to produce fats, while glutamine maintains the TCA cycle, particularly for generating NADPH, which is crucial for maintaining the cell’s metabolism and survival [150]. Interestingly, CSCs seem to prefer glutamine for their energy needs, especially when dividing rapidly. Because CSCs are so dependent on glutamine for their metabolism, targeting this pathway could be a promising strategy in cancer treatment. By blocking their ability to use glutamine, we could disrupt their energy supply and hinder their growth, making it harder for these cells to survive and proliferate in the tumor [151].
The serine–glycine–one-carbon (SGOC) metabolic network is emerging as a pivotal axis in sustaining the identity and functionality of CSCs [152,153,154,155,156]. This metabolic adaptation supports core stemness features such as self-renewal, high proliferation potential, and epigenetic plasticity. In particular, the de novo serine synthesis pathway (SSP) is frequently upregulated in CSCs across various tumor types, including glioblastoma, breast, and pancreatic cancers [157]. The pathway is initiated by the glycolytic intermediate 3-phosphoglycerate (3-PG), which is diverted away from glycolysis toward serine biosynthesis via the action of phosphoglycerate dehydrogenase (PHGDH) [158]. Subsequent steps involve phosphoserine aminotransferase (PSAT1) and phosphoserine phosphatase (PSPH), culminating in the production of serine. This amino acid serves as a substrate for serine hydroxymethyltransferases (SHMT1/2), which catalyze the reversible conversion of serine to glycine, producing one-carbon units essential for nucleotide biosynthesis and methylation reactions [159,160,161].
In CSCs, these one-carbon units feed into the folate and methionine cycles, enabling the synthesis of purines, thymidylate, and S-adenosylmethionine (SAM). SAM is the universal methyl donor required for the maintenance of the epigenetic landscape characteristic of stem-like tumor cells [162]. Through this link, SGOC metabolism directly influences DNA and histone methylation, allowing CSCs to retain transcriptional programs associated with pluripotency and resistance to differentiation [163]. High levels of PHGDH and SHMT2 have been reported in CSC-enriched subpopulations and correlate with poor prognosis in multiple cancers [164]. Furthermore, oncogenic signaling pathways such as MYC, ATF4, and mTORC1 have been implicated in transcriptionally regulating key SGOC enzymes, reinforcing the pathway’s importance in CSC biology [165]. Therefore, SGOC metabolism provides critical methyl groups necessary for purine ring formation and for the conversion of deoxyuridine monophosphate (dUMP) to deoxythymidine monophosphate (dTMP) by thymidylate synthase [166]. Enzymes such as dihydrofolate reductase (DHFR), thymidylate synthase (TYMS), and amidophosphoribosyltransferase (PPAT) are often upregulated in CSCs, contributing to enhanced nucleotide availability [167,168]. This metabolic adaptation enables CSCs to rapidly replenish nucleotide pools following genotoxic stress and supports their resistance to DNA-damaging agents [169].

6. Metabolic Plasticity of CSC Within the TME

The tumor microenvironment (TME) significantly impacts the metabolism of CSCs and LSCs, influencing their metabolic plasticity and response to therapies. Stromal cells, immune cells, the extracellular matrix, and hypoxic conditions within the TME interact with CSCs to support their survival and therapeutic resistance [170]. Stromal cells, such as cancer-associated fibroblasts (CAFs), secrete growth factors and metabolites that provide essential nutrients to CSCs, contributing to their metabolism and resistance to treatments [171]. In the TME, immune cells can modulate the metabolism of CSCs by releasing cytokines and factors that stimulate either glycolysis or OXPHOS, depending on the conditions.
Hypoxia, a common feature of solid tumors, activates HIF-1α signaling, promoting a shift to anaerobic glycolysis and helping cells survive low oxygen levels, thereby enhancing CSC resistance [172,173]. HIF-1α, mTOR, AMPK, and c-Myc are the most studied metabolic key regulators [174]. CSCs exhibit marked metabolic heterogeneity and plasticity, which contributes to their resistance to conventional therapies. CSC subpopulations can dynamically shift between glycolysis, OXPHOS, and lipid metabolism in response to environmental stress, hypoxia, or treatment pressure. For instance, CSCs in hypoxic tumor regions preferentially rely on glycolysis, while those in well-oxygenated areas depend more on OXPHOS to fulfill their energy requirements. This metabolic flexibility is regulated by local signaling cues and pathways such as HIF-1α and mTOR [175].
Under hypoxia, HIF-1α stabilizes and promotes a shift to anaerobic glycolysis, enabling CSCs to produce ATP while minimizing ROS accumulation [63]. mTOR supports CSC growth by driving lipid synthesis, protein translation, and anabolic metabolism [176]. In contrast, AMPK is activated under energy stress and promotes catabolic pathways like autophagy and fatty acid oxidation, aiding CSC adaptation [177,178]. C-Myc enhances glycolysis and nucleotide biosynthesis by upregulating key metabolic enzymes, supporting CSC proliferation [179].
These transcriptional and signaling pathways converge on metabolic effectors that reinforce the stem-like phenotype. For instance, SREBP1 (sterol regulatory element-binding protein 1) regulates genes involved in fatty acid and cholesterol synthesis, supporting membrane biosynthesis and cellular signaling [180,181]. SCD1 catalyzes the formation of monounsaturated fatty acids, critical for membrane fluidity and energy storage, and has been associated with increased CSC survival and tumorigenicity [182,183,184]. CSCs tightly regulate redox balance, maintaining low intracellular ROS levels to preserve stemness, self-renewal, and therapy resistance. This is largely mediated by upregulated antioxidant systems, particularly the glutathione (GSH) and thioredoxin (Trx) pathways [185,186,187]. The GSH system detoxifies ROS via enzymes like glutamate–cysteine ligase and glutathione peroxidase, while the Trx system reduces oxidized proteins and modulates redox signaling [188]. Disrupting these pathways sensitizes CSCs to oxidative stress, reducing viability and tumorigenicity. Notably, dual inhibition of GSH and Trx leads to synergistic cancer cell death, underscoring their therapeutic potential [189]. Furthermore, CSCs exploit metabolic adaptations to support their antioxidant defenses. Enhanced flux through the pentose phosphate pathway (PPP) provides NADPH, a critical reducing equivalent for the regeneration of GSH and Trx [190]. This metabolic reprogramming underscores the interplay between redox regulation and cellular metabolism in CSC maintenance. Targeting the redox balance in CSCs represents a promising therapeutic strategy [191]. Agents that elevate ROS levels or inhibit antioxidant systems can selectively eradicate CSCs while sparing normal stem cells, which possess more robust antioxidant capacities [135]. This metabolic plasticity is a key factor in treatment resistance and disease relapse, as CSCs can transition to an alternative metabolism to escape therapeutic effects, becoming dormant or quiescent for extended periods [192].

7. Epigenetic Regulation of Metabolic Pathways in CSCs

It is now well established that oncogenic mutations driving tumor initiation and progression are closely associated with widespread epigenetic disruptions. These include global DNA hypomethylation, localized hypermethylation at CpG (cytosine–phosphate–guanine) island promoters, altered histone modification profiles, and changes in nucleosome positioning [193]. Epigenetic regulation encompasses reversible chemical modifications to DNA and histone proteins that modulate gene expression without altering the nucleotide sequence. Principal mechanisms include DNA methylation, post-translational histone modifications—such as acetylation, methylation, phosphorylation, and ubiquitination—and the action of non-coding RNAs. These processes influence chromatin architecture and accessibility, thereby regulating transcriptional activity [194,195]. Epigenetic states are shaped by both genetic predisposition and environmental exposures, linking them to various pathological conditions, notably cancer [193]. Epimutations—aberrant epigenetic changes—can impair gene regulation, promoting genomic instability and cancer progression [196,197,198,199]. They may drive oncogenesis by evading growth control, resisting apoptosis, sustaining proliferation, and enhancing metastasis [137,141].
DNA methylation, catalyzed by DNA methyltransferase (DNMT) enzymes, mainly targets cytosine residues within CpG islands, typically located in gene promoter regions. In cancer, global DNA hypomethylation promotes chromosomal instability and oncogene activation, while focal hypermethylation silences tumor suppressor genes [200,201]. 5-hydroxymethylcytosine is generated through the oxidation of 5-methylcytosine and plays a role in maintaining stem cell plasticity and contributing to tumor progression [202].
Acetylation of histone H3 at lysine 9 (H3K9ac) promotes gene activation, whereas trimethylation at H3K27 (H3K27me3) is linked to gene silencing [203]. Dysregulation of these marks contributes to tumor development by altering chromatin dynamics [204,205]. Enzymes that modify histones are, therefore, promising targets for therapy, and histone deacetylase inhibitors (HDACis) are already used clinically in cancer treatment.
Non-coding RNAs (ncRNAs) are important epigenetic modulators that influence gene expression without altering DNA sequences [206]. Long ncRNAs (typically over 200 nucleotides in length) regulate transcription by interacting with chromatin-modifying complexes at specific genomic loci [147]. In contrast, microRNAs (miRNAs -17–25 nucleotides) act mainly at the post-transcriptional level by binding to untranslated regions of target mRNAs, leading to their degradation or translational repression through RNA interference pathways [207]. Both lncRNAs and miRNAs contribute to the precise regulation of gene networks involved in tumor initiation and progression [208,209,210,211].
Epigenetic alterations are increasingly recognized as key regulators of CSC identity. A well-characterized example is the epithelial-to-mesenchymal transition (EMT), which has been associated with the acquisition of CSC-like properties. In breast cancer, EMT correlates with elevated expression of CSC markers (e.g., CD44high/CD24low), enhanced self-renewal capacity, and increased tumor-initiating potential [212,213,214]. Emerging evidence indicates that EMT is governed, in part, by epigenetic mechanisms—particularly through modifications that regulate the transcription of mesenchymal factors such as ZEB1—highlighting the dynamic interplay between epigenetic control and CSC plasticity [215].

8. Optimizing Strategies for CSC Targeting

Targeting CSCs holds promise for long-term cancer remission. Traditional identification methods rely on flow cytometry and functional assays, but surface and cytoplasmic markers often lack specificity due to technical limitations and tumor heterogeneity. Incorporating metabolic markers may enhance the accuracy of CSC detection by capturing their distinct functional properties [216,217]. To evaluate metabolic therapies, experimental models are essential. While in vitro systems—such as CSC-enriched cultures and cell lines—offer initial insights, they often fail to replicate the tumor microenvironment. In contrast, patient-derived xenografts (PDX) and organoids provide more physiologically relevant models for assessing therapeutic responses [218].
Enhanced mitochondrial biogenesis can be a marker for identifying cancer cells with enhanced self-renewal capacity across various cancers [219,220]. Specifically, using fluorescent probes like MitoTracker allows for effective identification of CSCs regardless of their metabolic phenotype, whether glycolytic or reliant on OXPHOS. For instance, metabolic profiling of MCF7 breast cancer cells using MitoTracker has shown that a high mitochondrial mass is associated with CSC populations that have anabolic properties [72]. Mitochondrial enrichment in breast cancer stem cells enhances DNA repair and may help them resist anticancer treatments [221]. AML cells increase their mitochondrial mass by transferring mitochondria from bone marrow stromal cells, which supports survival and long-term culture potential [222]. Mitochondrial movement and mitophagy contribute to cancer stemness, and measuring mitochondrial membrane potential helps identify CSCs [223]. In breast cancer, a molecular toolkit has been developed for CSC identification based on markers like increased mitochondrial biogenesis, ROS production, and NADH levels [224].
Conventional therapies often fail to eradicate CSCs as they primarily target differentiated cancer cells. Therefore, developing strategies to specifically eliminate CSCs is a key challenge in modern cancer research (Table 1). A crucial step in this process is distinguishing CSCs from normal stem cells, as this differentiation could lead to the development of targeted treatments that spare healthy cells. Understanding the fundamental metabolic traits could lead to the identification of potential therapeutic targets [155,156]. Inhibitors of mTOR, for instance, impair lipid and protein biosynthesis, while agents like metformin and 2-deoxy-D-glucose (2-DG) disrupt glycolysis and mitochondrial metabolism [225]. The combination of 2-DG with fenformin, an AMPK activator, has shown enhanced antitumor efficacy in colon cancer models, including patient-derived xenografts. Metformin, an anti-diabetic agent, has been shown to suppress glycolysis, the TCA cycle, and nucleotide synthesis, leading to reduced CSC formation and tumor growth [226]. In a Phase II clinical trial (NCT01579812), metformin led to a 2.4-fold reduction in ovarian ALDH+CD133+ CSCs, increased cisplatin sensitivity, and improved both progression-free and overall survival [227]. Despite these promising advances, CSCs can develop resistance to metabolic therapies. Consequently, current research focuses on combinatorial approaches that simultaneously target multiple metabolic pathways or integrate metabolic inhibitors with immunotherapy, aiming to overcome compensatory survival mechanisms [228]. In pancreatic cancer, metformin induces cell death by inhibiting mitochondrial complex I, preventing the metabolic shift to glycolysis [229,230]. However, their survival is compromised when this metabolic adaptability is disrupted, as observed in K-ras-resistant pancreatic CSCs. Accumulating evidence supports the inclusion of metabolic alterations as a hallmark of cancer, positioning the metabolic profile of CSCs as a crucial target for therapeutic intervention [231]. High-throughput analysis and systems biology have accelerated the discovery of key metabolic regulators essential for CSC maintenance. Altogether, the distinctive metabolic characteristics of CSCs hold potential for therapeutic intervention without compromising normal tissue homeostasis.

9. Conclusions

The CSC model suggests that a small, metabolically adaptable subpopulation drives tumor growth and therapy resistance. CSCs can shift between glycolysis and mitochondrial metabolism, enabling survival under stress and in a quiescent state.
Targeting CSC metabolism is a promising strategy, but their adaptability and dormancy complicate eradication. A comprehensive strategy that targets multiple metabolic pathways is essential to effectively eliminate CSCs. Further research is essential to identify specific CSC markers, distinguish their metabolism from normal stem cells, and develop effective therapies.

Author Contributions

Conceptualization, M.M. and M.R.; methodology, M.M., G.L. and F.L.; software, M.M.; validation, G.L.; formal analysis, M.M.; investigation, M.M. and M.R.; data curation, M.M.; writing—original draft preparation, M.M.; writing—review and editing, M.R., S.D.P., A.A., F.C., M.F., A.d., P.D., G.L. and F.L.; visualization, G.L.; supervision, P.D. and G.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

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. Lasorsa, F.; Rutigliano, M.; Milella, M.; Ferro, M.; Pandolfo, S.D.; Crocetto, F.; Autorino, R.; Battaglia, M.; Ditonno, P.; Lucarelli, G. Cancer Stem Cells in Renal Cell Carcinoma: Origins and Biomarkers. Int. J. Mol. Sci. 2023, 24, 13179. [Google Scholar] [CrossRef] [PubMed]
  2. Tanabe, S. Cancer Stem Cell Markers and Related Network Pathways; Springer International Publishing: Cham, Switzerland, 2022; Volume 1393, ISBN 978-3-031-12973-5. [Google Scholar]
  3. Kolios, G.; Moodley, Y. Introduction to Stem Cells and Regenerative Medicine. Respiration 2013, 85, 3–10. [Google Scholar] [CrossRef] [PubMed]
  4. Chu, X.; Tian, W.; Ning, J.; Xiao, G.; Zhou, Y.; Wang, Z.; Zhai, Z.; Tanzhu, G.; Yang, J.; Zhou, R. Cancer Stem Cells: Advances in Knowledge and Implications for Cancer Therapy. Signal Transduct. Target. Ther. 2024, 9, 170. [Google Scholar] [CrossRef] [PubMed]
  5. Batlle, E.; Clevers, H. Cancer Stem Cells Revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef]
  6. Bisht, S.; Nigam, M.; Kunjwal, S.S.; Sergey, P.; Mishra, A.P.; Sharifi-Rad, J. Cancer Stem Cells: From an Insight into the Basics to Recent Advances and Therapeutic Targeting. Stem Cells Int. 2022, 2022, 9653244. [Google Scholar] [CrossRef]
  7. Denham, M.; Conley, B.; Olsson, F.; Cole, T.J.; Mollard, R. Stem Cells: An Overview. Curr. Protoc. Cell Biol. 2005, 28. [Google Scholar] [CrossRef]
  8. Vats, A.; Bielby, R.; Tolley, N.; Nerem, R.; Polak, J. Stem Cells. Lancet 2005, 366, 592–602. [Google Scholar] [CrossRef]
  9. Shenghui, H.; Nakada, D.; Morrison, S.J. Mechanisms of Stem Cell Self-Renewal. Annu. Rev. Cell Dev. Biol. 2009, 25, 377–406. [Google Scholar] [CrossRef]
  10. Falanga, V. Stem Cells in Tissue Repair and Regeneration. J. Investig. Dermatol. 2012, 132, 1538–1541. [Google Scholar] [CrossRef]
  11. Peiris-Pagès, M.; Martinez-Outschoorn, U.E.; Pestell, R.G.; Sotgia, F.; Lisanti, M.P. Cancer Stem Cell Metabolism. Breast Cancer Res. 2016, 18, 55. [Google Scholar] [CrossRef]
  12. Pattabiraman, D.R.; Weinberg, R.A. Tackling the Cancer Stem Cells—What Challenges Do They Pose? Nat. Rev. Drug Discov. 2014, 13, 497–512. [Google Scholar] [CrossRef] [PubMed]
  13. Aguilar-Gallardo, C.; Simón, C. Cells, Stem Cells, and Cancer Stem Cells. Semin. Reprod. Med. 2013, 31, 005–013. [Google Scholar] [CrossRef] [PubMed]
  14. Sancho, P.; Barneda, D.; Heeschen, C. Hallmarks of Cancer Stem Cell Metabolism. Br. J. Cancer 2016, 114, 1305–1312. [Google Scholar] [CrossRef] [PubMed]
  15. Deshmukh, A.; Deshpande, K.; Arfuso, F.; Newsholme, P.; Dharmarajan, A. Cancer Stem Cell Metabolism: A Potential Target for Cancer Therapy. Mol. Cancer 2016, 15, 69. [Google Scholar] [CrossRef]
  16. Yasuda, T.; Ishimoto, T.; Baba, H. Conflicting Metabolic Alterations in Cancer Stem Cells and Regulation by the Stromal Niche. Regen. Ther. 2021, 17, 8–12. [Google Scholar] [CrossRef]
  17. Brabletz, T.; Jung, A.; Spaderna, S.; Hlubek, F.; Kirchner, T. Migrating Cancer Stem Cells—An Integrated Concept of Malignant Tumour Progression. Nat. Rev. Cancer 2005, 5, 744–749. [Google Scholar] [CrossRef]
  18. Moriyama, T.; Ohuchida, K.; Mizumoto, K.; Cui, L.; Ikenaga, N.; Sato, N.; Tanaka, M. Enhanced Cell Migration and Invasion of CD133+ Pancreatic Cancer Cells Cocultured with Pancreatic Stromal Cells. Cancer 2010, 116, 3357–3368. [Google Scholar] [CrossRef]
  19. Fillmore, C.M.; Gupta, P.B.; Rudnick, J.A.; Caballero, S.; Keller, P.J.; Lander, E.S.; Kuperwasser, C. Estrogen Expands Breast Cancer Stem-like Cells through Paracrine FGF/Tbx3 Signaling. Proc. Natl. Acad. Sci. USA 2010, 107, 21737–21742. [Google Scholar] [CrossRef]
  20. Reynolds, B.A.; Weiss, S. Clonal and Population Analyses Demonstrate That an EGF-Responsive Mammalian Embryonic CNS Precursor Is a Stem Cell. Dev. Biol. 1996, 175, 1–13. [Google Scholar] [CrossRef]
  21. Lucarelli, G.; Galleggiante, V.; Rutigliano, M.; Vavallo, A.; Ditonno, P.; Battaglia, M. Isolation and Characterization of Cancer Stem Cells in Renal Cell Carcinoma. Urol. J. 2015, 82, 46–53. [Google Scholar] [CrossRef]
  22. Galleggiante, V.; Rutigliano, M.; Sallustio, F.; Ribatti, D.; Ditonno, P.; Bettocchi, C.; Selvaggi, F.P.; Lucarelli, G.; Battaglia, M. CTR2 Identifies a Population of Cancer Cells with Stem Cell-like Features in Patients with Clear Cell Renal Cell Carcinoma. J. Urol. 2014, 192, 1831–1841. [Google Scholar] [CrossRef] [PubMed]
  23. Matsuda, Y.; Ishiwata, T.; Yoshimura, H.; Yamashita, S.; Ushijima, T.; Arai, T. Systemic Administration of Small Interfering RNA Targeting Human Nestin Inhibits Pancreatic Cancer Cell Proliferation and Metastasis. Pancreas 2016, 45, 93–100. [Google Scholar] [CrossRef] [PubMed]
  24. Warrier, S.; Bhuvanalakshmi, G.; Arfuso, F.; Rajan, G.; Millward, M.; Dharmarajan, A. Cancer Stem-like Cells from Head and Neck Cancers Are Chemosensitized by the Wnt Antagonist, sFRP4, by Inducing Apoptosis, Decreasing Stemness, Drug Resistance and Epithelial to Mesenchymal Transition. Cancer Gene Ther. 2014, 21, 381–388. [Google Scholar] [CrossRef] [PubMed]
  25. Karsten, U.; Goletz, S. What Makes Cancer Stem Cell Markers Different? SpringerPlus 2013, 2, 301. [Google Scholar] [CrossRef]
  26. Tomao, F.; Papa, A.; Rossi, L.; Strudel, M.; Vici, P.; Lo Russo, G.; Tomao, S. Emerging Role of Cancer Stem Cells in the Biology and Treatment of Ovarian Cancer: Basic Knowledge and Therapeutic Possibilities for an Innovative Approach. J. Exp. Clin. Cancer Res. 2013, 32, 48. [Google Scholar] [CrossRef]
  27. Moitra, K.; Lou, H.; Dean, M. Multidrug Efflux Pumps and Cancer Stem Cells: Insights Into Multidrug Resistance and Therapeutic Development. Clin. Pharmacol. Ther. 2011, 89, 491–502. [Google Scholar] [CrossRef]
  28. Plaks, V.; Kong, N.; Werb, Z. The Cancer Stem Cell Niche: How Essential Is the Niche in Regulating Stemness of Tumor Cells? Cell Stem Cell 2015, 16, 225–238. [Google Scholar] [CrossRef]
  29. Ju, F.; Atyah, M.M.; Horstmann, N.; Gul, S.; Vago, R.; Bruns, C.J.; Zhao, Y.; Dong, Q.-Z.; Ren, N. Characteristics of the Cancer Stem Cell Niche and Therapeutic Strategies. Stem Cell Res. Ther. 2022, 13, 233. [Google Scholar] [CrossRef]
  30. Pan, Y.; Yuan, C.; Zeng, C.; Sun, C.; Xia, L.; Wang, G.; Chen, X.; Zhang, B.; Liu, J.; Ding, Z. Cancer Stem Cells and Niches: Challenges in Immunotherapy Resistance. Mol. Cancer 2025, 24, 52. [Google Scholar] [CrossRef]
  31. Lucarelli, G.; Loizzo, D.; Franzin, R.; Battaglia, S.; Ferro, M.; Cantiello, F.; Castellano, G.; Bettocchi, C.; Ditonno, P.; Battaglia, M. Metabolomic Insights into Pathophysiological Mechanisms and Biomarker Discovery in Clear Cell Renal Cell Carcinoma. Expert Rev. Mol. Diagn. 2019, 19, 397–407. [Google Scholar] [CrossRef]
  32. Lucarelli, G.; Galleggiante, V.; Rutigliano, M.; Sanguedolce, F.; Cagiano, S.; Bufo, P.; Lastilla, G.; Maiorano, E.; Ribatti, D.; Giglio, A.; et al. Metabolomic Profile of Glycolysis and the Pentose Phosphate Pathway Identifies the Central Role of Glucose-6-Phosphate Dehydrogenase in Clear Cell-Renal Cell Carcinoma. Oncotarget 2015, 6, 13371–13386. [Google Scholar] [CrossRef] [PubMed]
  33. De Francesco, E.M.; Sotgia, F.; Lisanti, M.P. Cancer Stem Cells (CSCs): Metabolic Strategies for Their Identification and Eradication. Biochem. J. 2018, 475, 1611–1634. [Google Scholar] [CrossRef] [PubMed]
  34. Zheng, J. Energy Metabolism of Cancer: Glycolysis versus Oxidative Phosphorylation (Review). Oncol. Lett. 2012, 4, 1151–1157. [Google Scholar] [CrossRef] [PubMed]
  35. 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]
  36. Ashton, T.M.; McKenna, W.G.; Kunz-Schughart, L.A.; Higgins, G.S. Oxidative Phosphorylation as an Emerging Target in Cancer Therapy. Clin. Cancer Res. 2018, 24, 2482–2490. [Google Scholar] [CrossRef]
  37. Pascual, G.; Domínguez, D.; Benitah, S.A. The Contributions of Cancer Cell Metabolism to Metastasis. Dis. Models Mech. 2018, 11, dmm032920. [Google Scholar] [CrossRef]
  38. Lucarelli, G.; Rutigliano, M.; Sallustio, F.; Ribatti, D.; Giglio, A.; Lepore Signorile, M.; Grossi, V.; Sanese, P.; Napoli, A.; Maiorano, E.; et al. Integrated Multi-Omics Characterization Reveals a Distinctive Metabolic Signature and the Role of NDUFA4L2 in Promoting Angiogenesis, Chemoresistance, and Mitochondrial Dysfunction in Clear Cell Renal Cell Carcinoma. Aging 2018, 10, 3957–3985. [Google Scholar] [CrossRef]
  39. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef]
  40. Garber, A.J.; Karl, I.E.; Kipnis, D.M. Alanine and Glutamine Synthesis and Release from Skeletal Muscle. II. The Precursor Role of Amino Acids in Alanine and Glutamine Synthesis. J. Biol. Chem. 1976, 251, 836–843. [Google Scholar] [CrossRef]
  41. Zhang, Y.; Zhang, S.; Sun, H.; Xu, L. The Pathogenesis and Therapeutic Implications of Metabolic Reprogramming in Renal Cell Carcinoma. Cell Death Discov. 2025, 11, 186. [Google Scholar] [CrossRef]
  42. Macheda, M.L.; Rogers, S.; Best, J.D. Molecular and Cellular Regulation of Glucose Transporter (GLUT) Proteins in Cancer. J. Cell. Physiol. 2005, 202, 654–662. [Google Scholar] [CrossRef] [PubMed]
  43. Ganapathy, V.; Thangaraju, M.; Prasad, P.D. Nutrient Transporters in Cancer: Relevance to Warburg Hypothesis and Beyond. Pharmacol. Ther. 2009, 121, 29–40. [Google Scholar] [CrossRef] [PubMed]
  44. Furuta, E.; Okuda, H.; Kobayashi, A.; Watabe, K. Metabolic Genes in Cancer: Their Roles in Tumor Progression and Clinical Implications. Biochim. Biophys. Acta BBA-Rev. Cancer 2010, 1805, 141–152. [Google Scholar] [CrossRef] [PubMed]
  45. Kaur, J.; Bhattacharyya, S. Cancer Stem Cells: Metabolic Characterization for Targeted Cancer Therapy. Front. Oncol. 2021, 11, 756888. [Google Scholar] [CrossRef]
  46. Shibuya, K.; Okada, M.; Suzuki, S.; Seino, M.; Seino, S.; Takeda, H.; Kitanaka, C. Targeting the Facilitative Glucose Transporter GLUT1 Inhibits the Self-Renewal and Tumor-Initiating Capacity of Cancer Stem Cells. Oncotarget 2015, 6, 651–661. [Google Scholar] [CrossRef]
  47. Ciavardelli, D.; Rossi, C.; Barcaroli, D.; Volpe, S.; Consalvo, A.; Zucchelli, M.; De Cola, A.; Scavo, E.; Carollo, R.; D’Agostino, D.; et al. Breast Cancer Stem Cells Rely on Fermentative Glycolysis and Are Sensitive to 2-Deoxyglucose Treatment. Cell Death Dis. 2014, 5, e1336. [Google Scholar] [CrossRef]
  48. Liao, J.; Qian, F.; Tchabo, N.; Mhawech-Fauceglia, P.; Beck, A.; Qian, Z.; Wang, X.; Huss, W.J.; Lele, S.B.; Morrison, C.D.; et al. Ovarian Cancer Spheroid Cells with Stem Cell-Like Properties Contribute to Tumor Generation, Metastasis and Chemotherapy Resistance through Hypoxia-Resistant Metabolism. PLoS ONE 2014, 9, e84941. [Google Scholar] [CrossRef]
  49. 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. J. Biol. Chem. 2011, 286, 32843–32853. [Google Scholar] [CrossRef]
  50. Folmes, C.D.L.; Nelson, T.J.; Martinez-Fernandez, A.; Arrell, D.K.; Lindor, J.Z.; Dzeja, P.P.; Ikeda, Y.; Perez-Terzic, C.; Terzic, A. Somatic Oxidative Bioenergetics Transitions into Pluripotency-Dependent Glycolysis to Facilitate Nuclear Reprogramming. Cell Metab. 2011, 14, 264–271. [Google Scholar] [CrossRef]
  51. Liu, P.-P.; Liao, J.; Tang, Z.-J.; Wu, W.-J.; Yang, J.; Zeng, Z.-L.; Hu, Y.; Wang, P.; Ju, H.-Q.; Xu, R.-H.; et al. Metabolic Regulation of Cancer Cell Side Population by Glucose through Activation of the Akt Pathway. Cell Death Differ. 2014, 21, 124–135. [Google Scholar] [CrossRef]
  52. Palorini, R.; Votta, G.; Balestrieri, C.; Monestiroli, A.; Olivieri, S.; Vento, R.; Chiaradonna, F. Energy Metabolism Characterization of a Novel Cancer Stem Cell- L Ike Line 3 AB-OS. J. Cell. Biochem. 2014, 115, 368–379. [Google Scholar] [CrossRef] [PubMed]
  53. Mizushima, E.; Tsukahara, T.; Emori, M.; Murata, K.; Akamatsu, A.; Shibayama, Y.; Hamada, S.; Watanabe, Y.; Kaya, M.; Hirohashi, Y.; et al. Osteosarcoma-initiating Cells Show High Aerobic Glycolysis and Attenuation of Oxidative Phosphorylation Mediated by LIN28B. Cancer Sci. 2020, 111, 36–46. [Google Scholar] [CrossRef] [PubMed]
  54. Feng, W.; Gentles, A.; Nair, R.V.; Huang, M.; Lin, Y.; Lee, C.Y.; Cai, S.; Scheeren, F.A.; Kuo, A.H.; Diehn, M. Targeting Unique Metabolic Properties of Breast Tumor Initiating Cells. Stem Cells 2014, 32, 1734–1745. [Google Scholar] [CrossRef]
  55. Song, K.; Kwon, H.; Han, C.; Zhang, J.; Dash, S.; Lim, K.; Wu, T. Active Glycolytic Metabolism in CD133(+) Hepatocellular Cancer Stem Cells: Regulation by MIR-122. Oncotarget 2015, 6, 40822–40835. [Google Scholar] [CrossRef]
  56. Estrella, V.; Chen, T.; Lloyd, M.; Wojtkowiak, J.; Cornnell, H.H.; Ibrahim-Hashim, A.; Bailey, K.; Balagurunathan, Y.; Rothberg, J.M.; Sloane, B.F.; et al. Acidity Generated by the Tumor Microenvironment Drives Local Invasion. Cancer Res. 2013, 73, 1524–1535. [Google Scholar] [CrossRef]
  57. Martinez-Outschoorn, U.E.; Prisco, M.; Ertel, A.; Tsirigos, A.; Lin, Z.; Pavlides, S.; Wang, C.; Flomenberg, N.; Knudsen, E.S.; Howell, A.; et al. Ketones and Lactate Increase Cancer Cell “Stemness,” Driving Recurrence, Metastasis and Poor Clinical Outcome in Breast Cancer: Achieving Personalized Medicine via Metabolo-Genomics. Cell Cycle 2011, 10, 1271–1286. [Google Scholar] [CrossRef]
  58. Lin, S.; Sun, L.; Lyu, X.; Ai, X.; Du, D.; Su, N.; Li, H.; Zhang, L.; Yu, J.; Yuan, S. Lactate-Activated Macrophages Induced Aerobic Glycolysis and Epithelial-Mesenchymal Transition in Breast Cancer by Regulation of CCL5-CCR5 Axis: A Positive Metabolic Feedback Loop. Oncotarget 2017, 8, 110426–110443. [Google Scholar] [CrossRef]
  59. Tasdogan, A.; Faubert, B.; Ramesh, V.; Ubellacker, J.M.; Shen, B.; Solmonson, A.; Murphy, M.M.; Gu, Z.; Gu, W.; Martin, M.; et al. Metabolic Heterogeneity Confers Differences in Melanoma Metastatic Potential. Nature 2020, 577, 115–120. [Google Scholar] [CrossRef]
  60. Brand, A.; Singer, K.; Koehl, G.E.; Kolitzus, M.; Schoenhammer, G.; Thiel, A.; Matos, C.; Bruss, C.; Klobuch, S.; Peter, K.; et al. LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. Cell Metab. 2016, 24, 657–671. [Google Scholar] [CrossRef]
  61. Lasorsa, F.; Di Meo, N.A.; Rutigliano, M.; Ferro, M.; Terracciano, D.; Tataru, O.S.; Battaglia, M.; Ditonno, P.; Lucarelli, G. Emerging Hallmarks of Metabolic Reprogramming in Prostate Cancer. Int. J. Mol. Sci. 2023, 24, 910. [Google Scholar] [CrossRef]
  62. Yuen, C.A.; Asuthkar, S.; Guda, M.R.; Tsung, A.J.; Velpula, K.K. Cancer Stem Cell Molecular Reprogramming of the Warburg Effect in Glioblastomas: A New Target Gleaned from an Old Concept. CNS Oncol. 2016, 5, 101–108. [Google Scholar] [CrossRef] [PubMed]
  63. Schito, L.; Semenza, G.L. Hypoxia-Inducible Factors: Master Regulators of Cancer Progression. Trends Cancer 2016, 2, 758–770. [Google Scholar] [CrossRef] [PubMed]
  64. Tong, W.-W.; Tong, G.-H.; Liu, Y. Cancer Stem Cells and Hypoxia-Inducible Factors (Review). Int. J. Oncol. 2018, 53, 469–476. [Google Scholar] [CrossRef]
  65. Wang, S.-F.; Tseng, L.-M.; Lee, H.-C. Role of Mitochondrial Alterations in Human Cancer Progression and Cancer Immunity. J. Biomed. Sci. 2023, 30, 61. [Google Scholar] [CrossRef]
  66. Wallace, D.C. Mitochondria and Cancer. Nat. Rev. Cancer 2012, 12, 685–698. [Google Scholar] [CrossRef]
  67. Vyas, S.; Zaganjor, E.; Haigis, M.C. Mitochondria and Cancer. Cell 2016, 166, 555–566. [Google Scholar] [CrossRef]
  68. Fernández-Vizarra, E.; López-Calcerrada, S.; Sierra-Magro, A.; Pérez-Pérez, R.; Formosa, L.E.; Hock, D.H.; Illescas, M.; Peñas, A.; Brischigliaro, M.; Ding, S.; et al. Two Independent Respiratory Chains Adapt OXPHOS Performance to Glycolytic Switch. Cell Metab. 2022, 34, 1792–1808.e6. [Google Scholar] [CrossRef]
  69. De Luca, A.; Fiorillo, M.; Peiris-Pagès, M.; Ozsvari, B.; Smith, D.L.; Sanchez-Alvarez, R.; Martinez-Outschoorn, U.E.; Cappello, A.R.; Pezzi, V.; Lisanti, M.P.; et al. Mitochondrial Biogenesis Is Required for the Anchorage-Independent Survival and Propagation of Stem-like Cancer Cells. Oncotarget 2015, 6, 14777–14795. [Google Scholar] [CrossRef]
  70. Wen, G.-M.; Xu, X.-Y.; Xia, P. Metabolism in Cancer Stem Cells: Targets for Clinical Treatment. Cells 2022, 11, 3790. [Google Scholar] [CrossRef]
  71. Pastò, A.; Bellio, C.; Pilotto, G.; Ciminale, V.; Silic-Benussi, M.; Guzzo, G.; Rasola, A.; Frasson, C.; Nardo, G.; Zulato, E.; et al. Cancer Stem Cells from Epithelial Ovarian Cancer Patients Privilege Oxidative Phosphorylation, and Resist Glucose Deprivation. Oncotarget 2014, 5, 4305–4319. [Google Scholar] [CrossRef]
  72. Sato, M.; Kawana, K.; Adachi, K.; Fujimoto, A.; Yoshida, M.; Nakamura, H.; Nishida, H.; Inoue, T.; Taguchi, A.; Takahashi, J.; et al. Spheroid Cancer Stem Cells Display Reprogrammed Metabolism and Obtain Energy by Actively Running the Tricarboxylic Acid (TCA) Cycle. Oncotarget 2016, 7, 33297–33305. [Google Scholar] [CrossRef] [PubMed]
  73. Gao, C.; Shen, Y.; Jin, F.; Miao, Y.; Qiu, X. Cancer Stem Cells in Small Cell Lung Cancer Cell Line H446: Higher Dependency on Oxidative Phosphorylation and Mitochondrial Substrate-Level Phosphorylation than Non-Stem Cancer Cells. PLoS ONE 2016, 11, e0154576. [Google Scholar] [CrossRef] [PubMed]
  74. Vlashi, E.; Lagadec, C.; Vergnes, L.; Matsutani, T.; Masui, K.; Poulou, M.; Popescu, R.; Della Donna, L.; Evers, P.; Dekmezian, C.; et al. Metabolic State of Glioma Stem Cells and Nontumorigenic Cells. Proc. Natl. Acad. Sci. USA 2011, 108, 16062–16067. [Google Scholar] [CrossRef] [PubMed]
  75. Janiszewska, M.; Suvà, M.L.; Riggi, N.; Houtkooper, R.H.; Auwerx, J.; Clément-Schatlo, V.; Radovanovic, I.; Rheinbay, E.; Provero, P.; Stamenkovic, I. Imp2 Controls Oxidative Phosphorylation and Is Crucial for Preserving Glioblastoma Cancer Stem Cells. Genes Dev. 2012, 26, 1926–1944. [Google Scholar] [CrossRef]
  76. Viale, A.; Pettazzoni, P.; Lyssiotis, C.A.; Ying, H.; Sánchez, N.; Marchesini, M.; Carugo, A.; Green, T.; Seth, S.; Giuliani, V.; et al. Oncogene Ablation-Resistant Pancreatic Cancer Cells Depend on Mitochondrial Function. Nature 2014, 514, 628–632. [Google Scholar] [CrossRef]
  77. LeBleu, V.S.; O’Connell, J.T.; Gonzalez Herrera, K.N.; Wikman, H.; Pantel, K.; Haigis, M.C.; De Carvalho, F.M.; Damascena, A.; Domingos Chinen, L.T.; Rocha, R.M.; et al. PGC-1α Mediates Mitochondrial Biogenesis and Oxidative Phosphorylation in Cancer Cells to Promote Metastasis. Nat. Cell Biol. 2014, 16, 992–1003. [Google Scholar] [CrossRef]
  78. Jiang, W.G.; Douglas-Jones, A.; Mansel, R.E. Expression of Peroxisome-proliferator Activated Receptor-gamma (PPARγ) and the PPARγ Co-activator, PGC-1, in Human Breast Cancer Correlates with Clinical Outcomes. Int. J. Cancer 2003, 106, 752–757. [Google Scholar] [CrossRef]
  79. Lamb, R.; Harrison, H.; Hulit, J.; Smith, D.L.; Lisanti, M.P.; Sotgia, F. Mitochondria as New Therapeutic Targets for Eradicating Cancer Stem Cells: Quantitative Proteomics and Functional Validation via MCT1/2 Inhibition. Oncotarget 2014, 5, 11029–11037. [Google Scholar] [CrossRef]
  80. Sancho, P.; Burgos-Ramos, E.; Tavera, A.; Bou Kheir, T.; Jagust, P.; Schoenhals, M.; Barneda, D.; Sellers, K.; Campos-Olivas, R.; Graña, O.; et al. MYC/PGC-1α Balance Determines the Metabolic Phenotype and Plasticity of Pancreatic Cancer Stem Cells. Cell Metab. 2015, 22, 590–605. [Google Scholar] [CrossRef]
  81. Vlashi, E.; Lagadec, C.; Vergnes, L.; Reue, K.; Frohnen, P.; Chan, M.; Alhiyari, Y.; Dratver, M.B.; Pajonk, F. Metabolic Differences in Breast Cancer Stem Cells and Differentiated Progeny. Breast Cancer Res. Treat. 2014, 146, 525–534. [Google Scholar] [CrossRef]
  82. Farnie, G.; Sotgia, F.; Lisanti, M.P. High Mitochondrial Mass Identifies a Sub-Population of Stem-like Cancer Cells That Are Chemo-Resistant. Oncotarget 2015, 6, 30472–30486. [Google Scholar] [CrossRef] [PubMed]
  83. Vazquez, F.; Lim, J.-H.; Chim, H.; Bhalla, K.; Girnun, G.; Pierce, K.; Clish, C.B.; Granter, S.R.; Widlund, H.R.; Spiegelman, B.M.; et al. PGC1α Expression Defines a Subset of Human Melanoma Tumors with Increased Mitochondrial Capacity and Resistance to Oxidative Stress. Cancer Cell 2013, 23, 287–301. [Google Scholar] [CrossRef] [PubMed]
  84. Di Meo, N.A.; Lasorsa, F.; Rutigliano, M.; Milella, M.; Ferro, M.; Battaglia, M.; Ditonno, P.; Lucarelli, G. The Dark Side of Lipid Metabolism in Prostate and Renal Carcinoma: Novel Insights into Molecular Diagnostic and Biomarker Discovery. Expert Rev. Mol. Diagn. 2023, 23, 297–313. [Google Scholar] [CrossRef] [PubMed]
  85. Lucarelli, G.; Ferro, M.; Loizzo, D.; Bianchi, C.; Terracciano, D.; Cantiello, F.; Bell, L.N.; Battaglia, S.; Porta, C.; Gernone, A.; et al. Integration of Lipidomics and Transcriptomics Reveals Reprogramming of the Lipid Metabolism and Composition in Clear Cell Renal Cell Carcinoma. Metabolites 2020, 10, 509. [Google Scholar] [CrossRef]
  86. Ferro, M.; Terracciano, D.; Buonerba, C.; Lucarelli, G.; Bottero, D.; Perdonà, S.; Autorino, R.; Serino, A.; Cantiello, F.; Damiano, R.; et al. The Emerging Role of Obesity, Diet and Lipid Metabolism in Prostate Cancer. Future Oncol. 2017, 13, 285–293. [Google Scholar] [CrossRef]
  87. Bianchi, C.; Meregalli, C.; Bombelli, S.; Di Stefano, V.; Salerno, F.; Torsello, B.; De Marco, S.; Bovo, G.; Cifola, I.; Mangano, E.; et al. The Glucose and Lipid Metabolism Reprogramming Is Grade-Dependent in Clear Cell Renal Cell Carcinoma Primary Cultures and Is Targetable to Modulate Cell Viability and Proliferation. Oncotarget 2017, 8, 113502–113515. [Google Scholar] [CrossRef]
  88. Beloribi-Djefaflia, S.; Vasseur, S.; Guillaumond, F. Lipid Metabolic Reprogramming in Cancer Cells. Oncogenesis 2016, 5, e189. [Google Scholar] [CrossRef]
  89. Delmas, D.; Mialhe, A.; Cotte, A.K.; Connat, J.-L.; Bouyer, F.; Hermetet, F.; Aires, V. Lipid Metabolism in Cancer: Exploring Phospholipids as Potential Biomarkers. Biomed. Pharmacother. 2025, 187, 118095. [Google Scholar] [CrossRef]
  90. Mordzińska-Rak, A.; Verdeil, G.; Hamon, Y.; Błaszczak, E.; Trombik, T. Dysregulation of Cholesterol Homeostasis in Cancer Pathogenesis. Cell. Mol. Life Sci. 2025, 82, 168. [Google Scholar] [CrossRef]
  91. Fu, W.; Sun, A.; Dai, H. Lipid Metabolism Involved in Progression and Drug Resistance of Breast Cancer. Genes Dis. 2025, 12, 101376. [Google Scholar] [CrossRef]
  92. Mollinedo, F.; Gajate, C. Lipid Rafts as Major Platforms for Signaling Regulation in Cancer. Adv. Biol. Regul. 2015, 57, 130–146. [Google Scholar] [CrossRef] [PubMed]
  93. Ito, K.; Carracedo, A.; Weiss, D.; Arai, F.; Ala, U.; Schafer, Z.T.; Evans, R.M.; Suda, T.; Lee, C.-H.; Pandolfi, P. A PML-PPARδ Pathway for Fatty Acid Oxidation Regulates Haematopoietic Stem Cell Maintenance. Nat. Med. 2012, 18, 1350–1358. [Google Scholar] [CrossRef] [PubMed]
  94. Ye, H.; Adane, B.; Khan, N.; Sullivan, T.; Minhajuddin, M.; Gasparetto, M.; Stevens, B.; Pei, S.; Balys, M.; Ashton, J.M.; et al. Leukemic Stem Cells Evade Chemotherapy by Metabolic Adaptation to an Adipose Tissue Niche. Cell Stem Cell 2016, 19, 23–37. [Google Scholar] [CrossRef]
  95. Pascual, G.; Avgustinova, A.; Mejetta, S.; Martín, M.; Castellanos, A.; Attolini, C.S.-O.; Berenguer, A.; Prats, N.; Toll, A.; Hueto, J.A.; et al. Targeting Metastasis-Initiating Cells through the Fatty Acid Receptor CD36. Nature 2017, 541, 41–45. [Google Scholar] [CrossRef]
  96. Yue, S.; Li, J.; Lee, S.-Y.; Lee, H.J.; Shao, T.; Song, B.; Cheng, L.; Masterson, T.A.; Liu, X.; Ratliff, T.L.; et al. Cholesteryl Ester Accumulation Induced by PTEN Loss and PI3K/AKT Activation Underlies Human Prostate Cancer Aggressiveness. Cell Metab. 2014, 19, 393–406. [Google Scholar] [CrossRef]
  97. de Gonzalo-Calvo, D.; López-Vilaró, L.; Nasarre, L.; Perez-Olabarria, M.; Vázquez, T.; Escuin, D.; Badimon, L.; Barnadas, A.; Lerma, E. Intratumor Cholesteryl Ester Accumulation Is Associated with Human Breast Cancer Proliferation and Aggressive Potential: A Molecular and Clinicopathological Study. BMC Cancer 2015, 15, 460. [Google Scholar] [CrossRef]
  98. Mitra, R.; Chao, O.; Urasaki, Y.; Goodman, O.B.; Le, T.T. Detection of Lipid-Rich Prostate Circulating Tumour Cells with Coherent Anti-Stokes Raman Scattering Microscopy. BMC Cancer 2012, 12, 540. [Google Scholar] [CrossRef]
  99. Tirinato, L.; Liberale, C.; Di Franco, S.; Candeloro, P.; Benfante, A.; La Rocca, R.; Potze, L.; Marotta, R.; Ruffilli, R.; Rajamanickam, V.P.; et al. Lipid Droplets: A New Player in Colorectal Cancer Stem Cells Unveiled by Spectroscopic Imaging. Stem Cells 2015, 33, 35–44. [Google Scholar] [CrossRef]
  100. Peck, B.; Schug, Z.T.; Zhang, Q.; Dankworth, B.; Jones, D.T.; Smethurst, E.; Patel, R.; Mason, S.; Jiang, M.; Saunders, R.; et al. Inhibition of Fatty Acid Desaturation Is Detrimental to Cancer Cell Survival in Metabolically Compromised Environments. Cancer Metab. 2016, 4, 6. [Google Scholar] [CrossRef]
  101. La Civita, E.; Sirica, R.; Crocetto, F.; Ferro, M.; Lasorsa, F.; Lucarelli, G.; Imbimbo, C.; Formisano, P.; Beguinot, F.; Terracciano, D. FABP4-Mediated ERK Phosphorylation Promotes Renal Cancer Cell Migration. BMC Cancer 2025, 25, 575. [Google Scholar] [CrossRef]
  102. Kawahara, I.; Mori, T.; Goto, K.; Fujii, K.; Ohmori, H.; Kishi, S.; Fujiwara-Tani, R.; Kuniyasu, H. Fatty Acids Induce Stemness in the Stromal Cells of a CT26 Mouse Tumor Model. Pathobiology 2017, 84, 237–242. [Google Scholar] [CrossRef] [PubMed]
  103. Kim, Y.-C.; Ntambi, J.M. Regulation of Stearoyl-CoA Desaturase Genes: Role in Cellular Metabolism and Preadipocyte Differentiation. Biochem. Biophys. Res. Commun. 1999, 266, 1–4. [Google Scholar] [CrossRef] [PubMed]
  104. Taraboletti, G.; Perin, L.; Bottazzi, B.; Mantovani, A.; Giavazzi, R.; Salmona, M. Membrane Fluidity Affects Tumor-cell Motility, Invasion and Lung-colonizing Potential. Int. J. Cancer 1989, 44, 707–713. [Google Scholar] [CrossRef] [PubMed]
  105. Noto, A.; De Vitis, C.; Pisanu, M.E.; Roscilli, G.; Ricci, G.; Catizone, A.; Sorrentino, G.; Chianese, G.; Taglialatela-Scafati, O.; Trisciuoglio, D.; et al. Stearoyl-CoA-Desaturase 1 Regulates Lung Cancer Stemness via Stabilization and Nuclear Localization of YAP/TAZ. Oncogene 2017, 36, 4573–4584. [Google Scholar] [CrossRef]
  106. Castro, L.F.C.; Wilson, J.M.; Gonçalves, O.; Galante-Oliveira, S.; Rocha, E.; Cunha, I. The Evolutionary History of the Stearoyl-CoA Desaturase Gene Family in Vertebrates. BMC Evol. Biol. 2011, 11, 132. [Google Scholar] [CrossRef]
  107. Bansal, S.; Berk, M.; Alkhouri, N.; Partrick, D.A.; Fung, J.J.; Feldstein, A. Stearoyl-CoA Desaturase Plays an Important Role in Proliferation and Chemoresistance in Human Hepatocellular Carcinoma. J. Surg. Res. 2014, 186, 29–38. [Google Scholar] [CrossRef]
  108. Lai, K.K.Y.; Kweon, S.-M.; Chi, F.; Hwang, E.; Kabe, Y.; Higashiyama, R.; Qin, L.; Yan, R.; Wu, R.P.; Lai, K.; et al. Stearoyl-CoA Desaturase Promotes Liver Fibrosis and Tumor Development in Mice via a Wnt Positive-Signaling Loop by Stabilization of Low-Density Lipoprotein-Receptor-Related Proteins 5 and 6. Gastroenterology 2017, 152, 1477–1491. [Google Scholar] [CrossRef]
  109. Colacino, J.A.; McDermott, S.P.; Sartor, M.A.; Wicha, M.S.; Rozek, L.S. Transcriptomic Profiling of Curcumin-Treated Human Breast Stem Cells Identifies a Role for Stearoyl-Coa Desaturase in Breast Cancer Prevention. Breast Cancer Res. Treat. 2016, 158, 29–41. [Google Scholar] [CrossRef]
  110. Li, J.; Condello, S.; Thomes-Pepin, J.; Ma, X.; Xia, Y.; Hurley, T.D.; Matei, D.; Cheng, J.-X. Lipid Desaturation Is a Metabolic Marker and Therapeutic Target of Ovarian Cancer Stem Cells. Cell Stem Cell 2017, 20, 303–314.e5. [Google Scholar] [CrossRef]
  111. Mason, P.; Liang, B.; Li, L.; Fremgen, T.; Murphy, E.; Quinn, A.; Madden, S.L.; Biemann, H.-P.; Wang, B.; Cohen, A.; et al. SCD1 Inhibition Causes Cancer Cell Death by Depleting Mono-Unsaturated Fatty Acids. PLoS ONE 2012, 7, e33823. [Google Scholar] [CrossRef]
  112. Pandey, P.R.; Xing, F.; Sharma, S.; Watabe, M.; Pai, S.K.; Iiizumi-Gairani, M.; Fukuda, K.; Hirota, S.; Mo, Y.-Y.; Watabe, K. Elevated Lipogenesis in Epithelial Stem-like Cell Confers Survival Advantage in Ductal Carcinoma in Situ of Breast Cancer. Oncogene 2013, 32, 5111–5122. [Google Scholar] [CrossRef] [PubMed]
  113. Shimano, H.; Sato, R. SREBP-Regulated Lipid Metabolism: Convergent Physiology—Divergent Pathophysiology. Nat. Rev. Endocrinol. 2017, 13, 710–730. [Google Scholar] [CrossRef] [PubMed]
  114. Sun, Y.; He, W.; Luo, M.; Zhou, Y.; Chang, G.; Ren, W.; Wu, K.; Li, X.; Shen, J.; Zhao, X.; et al. SREBP1 Regulates Tumorigenesis and Prognosis of Pancreatic Cancer through Targeting Lipid Metabolism. Tumor Biol. 2015, 36, 4133–4141. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, H.; Zhang, Z.; Song, L.; Gao, J.; Liu, Y. Lipid Metabolism of Cancer Stem Cells (Review). Oncol. Lett. 2022, 23, 119. [Google Scholar] [CrossRef]
  116. Ginestier, C.; Monville, F.; Wicinski, J.; Cabaud, O.; Cervera, N.; Josselin, E.; Finetti, P.; Guille, A.; Larderet, G.; Viens, P.; et al. Mevalonate Metabolism Regulates Basal Breast Cancer Stem Cells and Is a Potential Therapeutic Target. Stem Cells 2012, 30, 1327–1337. [Google Scholar] [CrossRef]
  117. Ganguly, K.; Rauth, S.; Marimuthu, S.; Kumar, S.; Batra, S.K. Unraveling Mucin Domains in Cancer and Metastasis: When Protectors Become Predators. Cancer Metastasis Rev. 2020, 39, 647–659. [Google Scholar] [CrossRef]
  118. Liu, C.; Chen, H.; Hu, B.; Shi, J.; Chen, Y.; Huang, K. New Insights into the Therapeutic Potentials of Statins in Cancer. Front. Pharmacol. 2023, 14, 1188926. [Google Scholar] [CrossRef]
  119. Ediriweera, M.K. Use of Cholesterol Metabolism for Anti-Cancer Strategies. Drug Discov. Today 2022, 27, 103347. [Google Scholar] [CrossRef]
  120. Mullen, P.J.; Yu, R.; Longo, J.; Archer, M.C.; Penn, L.Z. The Interplay between Cell Signalling and the Mevalonate Pathway in Cancer. Nat. Rev. Cancer 2016, 16, 718–731. [Google Scholar] [CrossRef]
  121. Lucarelli, G.; Loizzo, D.; Ferro, M.; Rutigliano, M.; Vartolomei, M.D.; Cantiello, F.; Buonerba, C.; Di Lorenzo, G.; Terracciano, D.; De Cobelli, O.; et al. Metabolomic Profiling for the Identification of Novel Diagnostic Markers and Therapeutic Targets in Prostate Cancer: An Update. Expert Rev. Mol. Diagn. 2019, 19, 377–387. [Google Scholar] [CrossRef]
  122. Di Lorenzo, G.; Sonpavde, G.; Pond, G.; Lucarelli, G.; Rossetti, S.; Facchini, G.; Scagliarini, S.; Cartenì, G.; Federico, P.; Daniele, B.; et al. Statin Use and Survival in Patients with Metastatic Castration-Resistant Prostate Cancer Treated with Abiraterone Acetate. Eur. Urol. Focus 2018, 4, 874–879. [Google Scholar] [CrossRef] [PubMed]
  123. Gordon, J.A.; Buonerba, C.; Pond, G.; Crona, D.; Gillessen, S.; Lucarelli, G.; Rossetti, S.; Dorff, T.; Artale, S.; Locke, J.A.; et al. Statin Use and Survival in Patients with Metastatic Castration-Resistant Prostate Cancer Treated with Abiraterone or Enzalutamide after Docetaxel Failure: The International Retrospective Observational STABEN Study. Oncotarget 2018, 9, 19861–19873. [Google Scholar] [CrossRef] [PubMed]
  124. Tirinato, L.; Pagliari, F.; Limongi, T.; Marini, M.; Falqui, A.; Seco, J.; Candeloro, P.; Liberale, C.; Di Fabrizio, E. An Overview of Lipid Droplets in Cancer and Cancer Stem Cells. Stem Cells Int. 2017, 2017, 1656053. [Google Scholar] [CrossRef] [PubMed]
  125. Yi, M.; Li, J.; Chen, S.; Cai, J.; Ban, Y.; Peng, Q.; Zhou, Y.; Zeng, Z.; Peng, S.; Li, X.; et al. Emerging Role of Lipid Metabolism Alterations in Cancer Stem Cells. J. Exp. Clin. Cancer Res. 2018, 37, 118. [Google Scholar] [CrossRef]
  126. Ma, Z.; Pan, S.; Yang, Y.; Ren, H.; Yin, S.; Chen, Q.; An, Z.; Zhao, X.; Xu, Z. Lipid Droplets: Emerging Therapeutic Targets for Age-Related Metabolic Diseases. Ageing Res. Rev. 2025, 108, 102758. [Google Scholar] [CrossRef]
  127. Bombelli, S.; Torsello, B.; De Marco, S.; Lucarelli, G.; Cifola, I.; Grasselli, C.; Strada, G.; Bovo, G.; Perego, R.A.; Bianchi, C. 36-kDa Annexin A3 Isoform Negatively Modulates Lipid Storage in Clear Cell Renal Cell Carcinoma Cells. Am. J. Pathol. 2020, 190, 2317–2326. [Google Scholar] [CrossRef]
  128. Maan, M.; Peters, J.M.; Dutta, M.; Patterson, A.D. Lipid Metabolism and Lipophagy in Cancer. Biochem. Biophys. Res. Commun. 2018, 504, 582–589. [Google Scholar] [CrossRef]
  129. Lue, H.; Podolak, J.; Kolahi, K.; Cheng, L.; Rao, S.; Garg, D.; Xue, C.-H.; Rantala, J.K.; Tyner, J.W.; Thornburg, K.L.; et al. Metabolic Reprogramming Ensures Cancer Cell Survival despite Oncogenic Signaling Blockade. Genes Dev. 2017, 31, 2067–2084. [Google Scholar] [CrossRef]
  130. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An Iron-Dependent Form of Nonapoptotic Cell Death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef]
  131. O’Malley, J.; Kumar, R.; Kuzmin, A.N.; Pliss, A.; Yadav, N.; Balachandar, S.; Wang, J.; Attwood, K.; Prasad, P.N.; Chandra, D. Lipid Quantification by Raman Microspectroscopy as a Potential Biomarker in Prostate Cancer. Cancer Lett. 2017, 397, 52–60. [Google Scholar] [CrossRef]
  132. Du, W.; Zhang, L.; Brett-Morris, A.; Aguila, B.; Kerner, J.; Hoppel, C.L.; Puchowicz, M.; Serra, D.; Herrero, L.; Rini, B.I.; et al. HIF Drives Lipid Deposition and Cancer in ccRCC via Repression of Fatty Acid Metabolism. Nat. Commun. 2017, 8, 1769. [Google Scholar] [CrossRef] [PubMed]
  133. Menard, J.A.; Christianson, H.C.; Kucharzewska, P.; Bourseau-Guilmain, E.; Svensson, K.J.; Lindqvist, E.; Chandran, V.I.; Kjellén, L.; Welinder, C.; Bengzon, J.; et al. Metastasis Stimulation by Hypoxia and Acidosis-Induced Extracellular Lipid Uptake Is Mediated by Proteoglycan-Dependent Endocytosis. Cancer Res. 2016, 76, 4828–4840. [Google Scholar] [CrossRef] [PubMed]
  134. Bailey, A.P.; Koster, G.; Guillermier, C.; Hirst, E.M.A.; MacRae, J.I.; Lechene, C.P.; Postle, A.D.; Gould, A.P. Antioxidant Role for Lipid Droplets in a Stem Cell Niche of Drosophila. Cell 2015, 163, 340–353. [Google Scholar] [CrossRef]
  135. Chen, J.; Cui, L.; Lu, S.; Xu, S. Amino Acid Metabolism in Tumor Biology and Therapy. Cell Death Dis. 2024, 15, 42. [Google Scholar] [CrossRef]
  136. 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]
  137. Wei, Z.; Liu, X.; Cheng, C.; Yu, W.; Yi, P. Metabolism of Amino Acids in Cancer. Front. Cell Dev. Biol. 2021, 8, 603837. [Google Scholar] [CrossRef]
  138. Sharif, T.; Dai, C.; Martell, E.; Ghassemi-Rad, M.S.; Hanes, M.R.; Murphy, P.J.; Kennedy, B.E.; Venugopal, C.; Subapanditha, M.; Giacomantonio, C.A.; et al. TAp73 Modifies Metabolism and Positively Regulates Growth of Cancer Stem–Like Cells in a Redox-Sensitive Manner. Clin. Cancer Res. 2019, 25, 2001–2017. [Google Scholar] [CrossRef]
  139. Jaggupilli, A.; Ly, S.; Nguyen, K.; Anand, V.; Yuan, B.; El-Dana, F.; Yan, Y.; Arvanitis, Z.; Piyarathna, D.W.B.; Putluri, N.; et al. Metabolic Stress Induces GD2+ Cancer Stem Cell-like Phenotype in Triple-Negative Breast Cancer. Br. J. Cancer 2022, 126, 615–627. [Google Scholar] [CrossRef]
  140. Mukha, A.; Kahya, U.; Linge, A.; Chen, O.; Löck, S.; Lukiyanchuk, V.; Richter, S.; Alves, T.C.; Peitzsch, M.; Telychko, V.; et al. GLS-Driven Glutamine Catabolism Contributes to Prostate Cancer Radiosensitivity by Regulating the Redox State, Stemness and ATG5-Mediated Autophagy. Theranostics 2021, 11, 7844–7868. [Google Scholar] [CrossRef]
  141. Yoshikawa, M.; Tsuchihashi, K.; Ishimoto, T.; Yae, T.; Motohara, T.; Sugihara, E.; Onishi, N.; Masuko, T.; Yoshizawa, K.; Kawashiri, S.; et al. xCT Inhibition Depletes CD44v-Expressing Tumor Cells That Are Resistant to EGFR-Targeted Therapy in Head and Neck Squamous Cell Carcinoma. Cancer Res. 2013, 73, 1855–1866. [Google Scholar] [CrossRef]
  142. Battula, V.L.; Shi, Y.; Evans, K.W.; Wang, R.-Y.; Spaeth, E.L.; Jacamo, R.O.; Guerra, R.; Sahin, A.A.; Marini, F.C.; Hortobagyi, G.; et al. Ganglioside GD2 Identifies Breast Cancer Stem Cells and Promotes Tumorigenesis. J. Clin. Investig. 2012, 122, 2066–2078. [Google Scholar] [CrossRef] [PubMed]
  143. Pacifico, F.; Leonardi, A.; Crescenzi, E. Glutamine Metabolism in Cancer Stem Cells: A Complex Liaison in the Tumor Microenvironment. Int. J. Mol. Sci. 2023, 24, 2337. [Google Scholar] [CrossRef] [PubMed]
  144. Li, D.; Fu, Z.; Chen, R.; Zhao, X.; Zhou, Y.; Zeng, B.; Yu, M.; Zhou, Q.; Lin, Q.; Gao, W.; et al. Inhibition of Glutamine Metabolism Counteracts Pancreatic Cancer Stem Cell Features and Sensitizes Cells to Radiotherapy. Oncotarget 2015, 6, 31151–31163. [Google Scholar] [CrossRef]
  145. Fernandez-Marcos, P.J.; Nóbrega-Pereira, S. NADPH: New Oxygen for the ROS Theory of Aging. Oncotarget 2016, 7, 50814–50815. [Google Scholar] [CrossRef]
  146. Liao, J.; Liu, P.-P.; Hou, G.; Shao, J.; Yang, J.; Liu, K.; Lu, W.; Wen, S.; Hu, Y.; Huang, P. Regulation of Stem-like Cancer Cells by Glutamine through β-Catenin Pathway Mediated by Redox Signaling. Mol. Cancer 2017, 16, 51. [Google Scholar] [CrossRef]
  147. Prasad, P.; Ghosh, S.; Roy, S.S. Glutamine Deficiency Promotes Stemness and Chemoresistance in Tumor Cells through DRP1-Induced Mitochondrial Fragmentation. Cell. Mol. Life Sci. 2021, 78, 4821–4845. [Google Scholar] [CrossRef]
  148. Cluntun, A.A.; Lukey, M.J.; Cerione, R.A.; Locasale, J.W. Glutamine Metabolism in Cancer: Understanding the Heterogeneity. Trends Cancer 2017, 3, 169–180. [Google Scholar] [CrossRef]
  149. Davidson, S.M.; Papagiannakopoulos, T.; Olenchock, B.A.; Heyman, J.E.; Keibler, M.A.; Luengo, A.; Bauer, M.R.; Jha, A.K.; O’Brien, J.P.; Pierce, K.A.; et al. Environment Impacts the Metabolic Dependencies of Ras-Driven Non-Small Cell Lung Cancer. Cell Metab. 2016, 23, 517–528. [Google Scholar] [CrossRef]
  150. Raimundo, N.; Baysal, B.E.; Shadel, G.S. Revisiting the TCA Cycle: Signaling to Tumor Formation. Trends Mol. Med. 2011, 17, 641–649. [Google Scholar] [CrossRef]
  151. Jin, J.; Byun, J.-K.; Choi, Y.-K.; Park, K.-G. Targeting Glutamine Metabolism as a Therapeutic Strategy for Cancer. Exp. Mol. Med. 2023, 55, 706–715. [Google Scholar] [CrossRef]
  152. Yang, M.; Vousden, K.H. Serine and One-Carbon Metabolism in Cancer. Nat. Rev. Cancer 2016, 16, 650–662. [Google Scholar] [CrossRef] [PubMed]
  153. Huang, D.; Cai, H.; Huang, H. Serine Metabolism in Tumor Progression and Immunotherapy. Discov. Oncol. 2025, 16, 628. [Google Scholar] [CrossRef] [PubMed]
  154. Lyu, H.; Bao, S.; Cai, L.; Wang, M.; Liu, Y.; Sun, Y.; Hu, X. The Role and Research Progress of Serine Metabolism in Tumor Cells. Front. Oncol. 2025, 15, 1509662. [Google Scholar] [CrossRef] [PubMed]
  155. Tassinari, V.; Jia, W.; Chen, W.-L.; Candi, E.; Melino, G. The Methionine Cycle and Its Cancer Implications. Oncogene 2024, 43, 3483–3488. [Google Scholar] [CrossRef]
  156. Lim, E.W.; Metallo, C.M. Tracing the Diverse Paths of One-Carbon Metabolism in Cancer and Beyond. Cold Spring Harb. Perspect. Med. 2024, 14, a041533. [Google Scholar] [CrossRef]
  157. Mattaini, K.R.; Sullivan, M.R.; Vander Heiden, M.G. The Importance of Serine Metabolism in Cancer. J. Cell Biol. 2016, 214, 249–257. [Google Scholar] [CrossRef]
  158. Cai, Z.; Li, W.; Hager, S.; Wilson, J.L.; Afjehi-Sadat, L.; Heiss, E.H.; Weichhart, T.; Heffeter, P.; Weckwerth, W. Targeting PHGDH Reverses the Immunosuppressive Phenotype of Tumor-Associated Macrophages through α-Ketoglutarate and mTORC1 Signaling. Cell. Mol. Immunol. 2024, 21, 448–465. [Google Scholar] [CrossRef]
  159. Labuschagne, C.F.; van den Broek, N.J.F.; Mackay, G.M.; Vousden, K.H.; Maddocks, O.D.K. Serine, but Not Glycine, Supports One-Carbon Metabolism and Proliferation of Cancer Cells. Cell Rep. 2014, 7, 1248–1258. [Google Scholar] [CrossRef]
  160. Subramani, S. A Mammalian Pexophagy Target. Nat. Cell Biol. 2015, 17, 1371–1373. [Google Scholar] [CrossRef]
  161. Zhao, M.; Hou, Y.; Du, Y.; Yang, L.; Qin, Y.; Peng, M.; Liu, S.; Wan, X.; Qiao, Y.; Zeng, H.; et al. Drosha-Independent miR-6778–5p Strengthens Gastric Cancer Stem Cell Stemness via Regulation of Cytosolic One-Carbon Folate Metabolism. Cancer Lett. 2020, 478, 8–21. [Google Scholar] [CrossRef]
  162. Qi, Y.-N.; Liu, Z.; Hong, L.-L.; Li, P.; Ling, Z.-Q. Methyltransferase-like Proteins in Cancer Biology and Potential Therapeutic Targeting. J. Hematol. Oncol. 2023, 16, 89. [Google Scholar] [CrossRef] [PubMed]
  163. Zhang, W.C.; Shyh-Chang, N.; Yang, H.; Rai, A.; Umashankar, S.; Ma, S.; Soh, B.S.; Sun, L.L.; Tai, B.C.; Nga, M.E.; et al. Glycine Decarboxylase Activity Drives Non-Small Cell Lung Cancer Tumor-Initiating Cells and Tumorigenesis. Cell 2012, 148, 259–272. [Google Scholar] [CrossRef] [PubMed]
  164. He, L.; Endress, J.; Cho, S.; Li, Z.; Zheng, Y.; Asara, J.M.; Blenis, J. Suppression of Nuclear GSK3 Signaling Promotes Serine/One-Carbon Metabolism and Confers Metabolic Vulnerability in Lung Cancer Cells. Sci. Adv. 2022, 8, eabm8786. [Google Scholar] [CrossRef] [PubMed]
  165. Nigdelioglu, R.; Hamanaka, R.B.; Meliton, A.Y.; O’Leary, E.; Witt, L.J.; Cho, T.; Sun, K.; Bonham, C.; Wu, D.; Woods, P.S.; et al. Transforming Growth Factor (TGF)-β Promotes de Novo Serine Synthesis for Collagen Production. J. Biol. Chem. 2016, 291, 27239–27251. [Google Scholar] [CrossRef]
  166. Lane, A.N.; Fan, T.W.-M. Regulation of Mammalian Nucleotide Metabolism and Biosynthesis. Nucleic Acids Res. 2015, 43, 2466–2485. [Google Scholar] [CrossRef]
  167. Wang, L.; Yang, R.; Kong, Y.; Zhou, J.; Chen, Y.; Li, R.; Chen, C.; Tang, X.; Chen, X.; Xia, J.; et al. Integrative Single-Cell and Bulk Transcriptomes Analyses Reveals Heterogeneity of Serine-Glycine-One-Carbon Metabolism with Distinct Prognoses and Therapeutic Vulnerabilities in HNSCC. Int. J. Oral Sci. 2024, 16, 44. [Google Scholar] [CrossRef]
  168. Zhang, F.; Ye, J.; Guo, W.; Zhang, F.; Wang, L.; Han, A. TYMS-TM4SF4 Axis Promotes the Progression of Colorectal Cancer by EMT and Upregulating Stem Cell Marker. Am. J. Cancer Res. 2022, 13, 1009. [Google Scholar]
  169. Lv, Y.; Wang, X.; Li, X.; Xu, G.; Bai, Y.; Wu, J.; Piao, Y.; Shi, Y.; Xiang, R.; Wang, L. Nucleotide de Novo Synthesis Increases Breast Cancer Stemness and Metastasis via cGMP-PKG-MAPK Signaling Pathway. PLoS Biol. 2020, 18, e3000872. [Google Scholar] [CrossRef]
  170. Liang, T.; Tao, T.; Wu, K.; Liu, L.; Xu, W.; Zhou, D.; Fang, H.; Ding, Q.; Huang, G.; Wu, S. Cancer-Associated Fibroblast-Induced Remodeling of Tumor Microenvironment in Recurrent Bladder Cancer. Adv. Sci. 2023, 10, 2303230. [Google Scholar] [CrossRef]
  171. Missiaen, R.; Lesner, N.P.; Simon, M.C. HIF: A Master Regulator of Nutrient Availability and Metabolic Cross-talk in the Tumor Microenvironment. EMBO J. 2023, 42, e112067. [Google Scholar] [CrossRef]
  172. Gilkes, D.M.; Semenza, G.L.; Wirtz, D. Hypoxia and the Extracellular Matrix: Drivers of Tumour Metastasis. Nat. Rev. Cancer 2014, 14, 430–439. [Google Scholar] [CrossRef] [PubMed]
  173. Zhu, Y.; Li, X.; Wang, L.; Hong, X.; Yang, J. Metabolic Reprogramming and Crosstalk of Cancer-Related Fibroblasts and Immune Cells in the Tumor Microenvironment. Front. Endocrinol. 2022, 13, 988295. [Google Scholar] [CrossRef] [PubMed]
  174. Moldogazieva, N.T.; Mokhosoev, I.M.; Terentiev, A.A. Metabolic Heterogeneity of Cancer Cells: An Interplay between HIF-1, GLUTs, and AMPK. Cancers 2020, 12, 862. [Google Scholar] [CrossRef] [PubMed]
  175. Cannino, G.; Ciscato, F.; Masgras, I.; Sánchez-Martín, C.; Rasola, A. Metabolic Plasticity of Tumor Cell Mitochondria. Front. Oncol. 2018, 8, 333. [Google Scholar] [CrossRef]
  176. Lamming, D.W.; Ye, L.; Katajisto, P.; Goncalves, M.D.; Saitoh, M.; Stevens, D.M.; Davis, J.G.; Salmon, A.B.; Richardson, A.; Ahima, R.S.; et al. Rapamycin-Induced Insulin Resistance Is Mediated by mTORC2 Loss and Uncoupled from Longevity. Science 2012, 335, 1638–1643. [Google Scholar] [CrossRef]
  177. Hardie, D.G.; Alessi, D.R. LKB1 and AMPK and the Cancer-Metabolism Link—Ten Years After. BMC Biol. 2013, 11, 36. [Google Scholar] [CrossRef]
  178. Grossi, V.; Lucarelli, G.; Forte, G.; Peserico, A.; Matrone, A.; Germani, A.; Rutigliano, M.; Stella, A.; Bagnulo, R.; Loconte, D.; et al. Loss of STK11 Expression Is an Early Event in Prostate Carcinogenesis and Predicts Therapeutic Response to Targeted Therapy against MAPK/P38. Autophagy 2015, 11, 2102–2113. [Google Scholar] [CrossRef]
  179. Dang, C.V. MYC on the Path to Cancer. Cell 2012, 149, 22–35. [Google Scholar] [CrossRef]
  180. Morton, J.D.; Shimomura, L. Sterol Regulatory Element-Binding Proteins: Activators of Cholesterol and Fatty Acid Biosynthesis. Curr. Opin. Lipidol. 1999, 10, 143–150. [Google Scholar] [CrossRef]
  181. He, Y.; Qi, S.; Chen, L.; Zhu, J.; Liang, L.; Chen, X.; Zhang, H.; Zhuo, L.; Zhao, S.; Liu, S.; et al. The Roles and Mechanisms of SREBP1 in Cancer Development and Drug Response. Genes Dis. 2024, 11, 100987. [Google Scholar] [CrossRef]
  182. Min, J.-Y.; Kim, D.-H. Stearoyl-CoA Desaturase 1 as a Therapeutic Biomarker: Focusing on Cancer Stem Cells. Int. J. Mol. Sci. 2023, 24, 8951. [Google Scholar] [CrossRef] [PubMed]
  183. Sen, U.; Coleman, C.; Sen, T. Stearoyl Coenzyme A Desaturase-1: Multitasker in Cancer, Metabolism, and Ferroptosis. Trends Cancer 2023, 9, 480–489. [Google Scholar] [CrossRef] [PubMed]
  184. Ascenzi, F.; De Vitis, C.; Maugeri-Saccà, M.; Napoli, C.; Ciliberto, G.; Mancini, R. SCD1, Autophagy and Cancer: Implications for Therapy. J. Exp. Clin. Cancer Res. CR 2021, 40, 265. [Google Scholar] [CrossRef] [PubMed]
  185. Ding, S.; Li, C.; Cheng, N.; Cui, X.; Xu, X.; Zhou, G. Redox Regulation in Cancer Stem Cells. Oxid. Med. Cell. Longev. 2015, 2015, 750798. [Google Scholar] [CrossRef]
  186. Seitz, R.; Tümen, D.; Kunst, C.; Heumann, P.; Schmid, S.; Kandulski, A.; Müller, M.; Gülow, K. Exploring the Thioredoxin System as a Therapeutic Target in Cancer: Mechanisms and Implications. Antioxidants 2024, 13, 1078. [Google Scholar] [CrossRef]
  187. Abdullah, N.A.; Md Hashim, N.F.; Muhamad Zakuan, N.; Chua, J.X. Thioredoxin System in Colorectal Cancer: Its Role in Carcinogenesis, Disease Progression, and Response to Treatment. Life Sci. 2024, 348, 122711. [Google Scholar] [CrossRef]
  188. Harris, I.S.; Treloar, A.E.; Inoue, S.; Sasaki, M.; Gorrini, C.; Lee, K.C.; Yung, K.Y.; Brenner, D.; Knobbe-Thomsen, C.B.; Cox, M.A.; et al. Glutathione and Thioredoxin Antioxidant Pathways Synergize to Drive Cancer Initiation and Progression. Cancer Cell 2015, 27, 211–222. [Google Scholar] [CrossRef]
  189. Panieri, E.; Santoro, M.M. ROS Homeostasis and Metabolism: A Dangerous Liason in Cancer Cells. Cell Death Dis. 2016, 7, e2253. [Google Scholar] [CrossRef]
  190. Dando, I.; Cordani, M.; Dalla Pozza, E.; Biondani, G.; Donadelli, M.; Palmieri, M. Antioxidant Mechanisms and ROS-Related MicroRNAs in Cancer Stem Cells. Oxid. Med. Cell. Longev. 2015, 2015, 425708. [Google Scholar] [CrossRef]
  191. Fiorillo, M.; Tóth, F.; Brindisi, M.; Sotgia, F.; Lisanti, M.P. Deferiprone (DFP) Targets Cancer Stem Cell (CSC) Propagation by Inhibiting Mitochondrial Metabolism and Inducing ROS Production. Cells 2020, 9, 1529. [Google Scholar] [CrossRef]
  192. Chen, K.; Zhang, C.; Ling, S.; Wei, R.; Wang, J.; Xu, X. The Metabolic Flexibility of Quiescent CSC: Implications for Chemotherapy Resistance. Cell Death Dis. 2021, 12, 835. [Google Scholar] [CrossRef] [PubMed]
  193. Dawson, M.A.; Kouzarides, T. Cancer Epigenetics: From Mechanism to Therapy. Cell 2012, 150, 12–27. [Google Scholar] [CrossRef] [PubMed]
  194. Talbert, P.B.; Henikoff, S. Histone Variants—Ancient Wrap Artists of the Epigenome. Nat. Rev. Mol. Cell Biol. 2010, 11, 264–275. [Google Scholar] [CrossRef] [PubMed]
  195. Greer, E.L.; Shi, Y. Histone Methylation: A Dynamic Mark in Health, Disease and Inheritance. Nat. Rev. Genet. 2012, 13, 343–357. [Google Scholar] [CrossRef]
  196. Radpour, R.; Barekati, Z.; Kohler, C.; Holzgreve, W.; Zhong, X.Y. New Trends in Molecular Biomarker Discovery for Breast Cancer. Genet. Test. Mol. Biomark. 2009, 13, 565–571. [Google Scholar] [CrossRef]
  197. Lønning, P.E.; Nikolaienko, O.; Knappskog, S. Constitutional Epimutations: From Rare Events Toward Major Cancer Risk Factors? JCO Precis. Oncol. 2025, 9, e2400746. [Google Scholar] [CrossRef]
  198. Zhou, W.; Reizel, Y. On Correlative and Causal Links of Replicative Epimutations. Trends Genet. TIG 2025, 41, 60–75. [Google Scholar] [CrossRef]
  199. Mohammad, A.; Jha, S. Epimutations and Their Effect on Chromatin Organization: Exciting Avenues for Cancer Treatment. Cancers 2022, 15, 215. [Google Scholar] [CrossRef]
  200. Mandhair, H.K.; Novak, U.; Radpour, R. Epigenetic Regulation of Autophagy: A Key Modification in Cancer Cells and Cancer Stem Cells. World J. Stem Cells 2021, 13, 542–567. [Google Scholar] [CrossRef]
  201. Radpour, R.; Haghighi, M.M.; Fan, A.X.-C.; Torbati, P.M.; Hahn, S.; Holzgreve, W.; Zhong, X.Y. High-Throughput Hacking of the Methylation Patterns in Breast Cancer by In Vitro Transcription and Thymidine-Specific Cleavage Mass Array on MALDI-TOF Silico-Chip. Mol. Cancer Res. 2008, 6, 1702–1709. [Google Scholar] [CrossRef]
  202. Radpour, R.; Sikora, M.; Grussenmeyer, T.; Kohler, C.; Barekati, Z.; Holzgreve, W.; Lefkovits, I.; Zhong, X.Y. Simultaneous Isolation of DNA, RNA, and Proteins for Genetic, Epigenetic, Transcriptomic, and Proteomic Analysis. J. Proteome Res. 2009, 8, 5264–5274. [Google Scholar] [CrossRef] [PubMed]
  203. Tan, M.; Luo, H.; Lee, S.; Jin, F.; Yang, J.S.; Montellier, E.; Buchou, T.; Cheng, Z.; Rousseaux, S.; Rajagopal, N.; et al. Identification of 67 Histone Marks and Histone Lysine Crotonylation as a New Type of Histone Modification. Cell 2011, 146, 1016–1028. [Google Scholar] [CrossRef] [PubMed]
  204. Mancarella, D.; Plass, C. Epigenetic Signatures in Cancer: Proper Controls, Current Challenges and the Potential for Clinical Translation. Genome Med. 2021, 13, 23. [Google Scholar] [CrossRef]
  205. Radpour, R.; Stucki, M.; Riether, C.; Ochsenbein, A.F. Epigenetic Silencing of Immune-Checkpoint Receptors in Bone Marrow- Infiltrating T Cells in Acute Myeloid Leukemia. Front. Oncol. 2021, 11, 663406. [Google Scholar] [CrossRef]
  206. Esteller, M. Non-Coding RNAs in Human Disease. Nat. Rev. Genet. 2011, 12, 861–874. [Google Scholar] [CrossRef]
  207. Jaggi, B.; Poon, S.S.S.; Macaulay, C.; Palcic, B. Imaging System for Morphometric Assessment of Absorption or Fluorescence in Stained Cells. Cytometry 1988, 9, 566–572. [Google Scholar] [CrossRef]
  208. Zhang, P.; Brinton, L.T.; Williams, K.; Sher, S.; Orwick, S.; Tzung-Huei, L.; Mims, A.S.; Coss, C.C.; Kulp, S.K.; Youssef, Y.; et al. Targeting DNA Damage Repair Functions of Two Histone Deacetylases, HDAC8 and SIRT6, Sensitizes Acute Myeloid Leukemia to NAMPT Inhibition. Clin. Cancer Res. 2021, 27, 2352–2366. [Google Scholar] [CrossRef]
  209. Zhang, H.; Wang, J.; Liu, C.; Yan, K.; Wang, X.; Sheng, X. Interactions between Long Non-Coding RNAs and M6 A Modification in Cancer. Discov. Oncol. 2025, 16, 579. [Google Scholar] [CrossRef]
  210. Peng, J.; Liu, W.; Tian, J.; Shu, Y.; Zhao, R.; Wang, Y. Non-Coding RNAs as Key Regulators of Epithelial-Mesenchymal Transition in Breast Cancer. Front. Cell Dev. Biol. 2025, 13, 1544310. [Google Scholar] [CrossRef]
  211. Solaimani, M.; Hosseinzadeh, S.; Abasi, M. Non-Coding RNAs, a Double-Edged Sword in Breast Cancer Prognosis. Cancer Cell Int. 2025, 25, 123. [Google Scholar] [CrossRef]
  212. Jolly, M.K.; Jia, D.; Boareto, M.; Mani, S.A.; Pienta, K.J.; Ben-Jacob, E.; Levine, H. Coupling the Modules of EMT and Stemness: A Tunable ‘Stemness Window’ Model. Oncotarget 2015, 6, 25161–25174. [Google Scholar] [CrossRef] [PubMed]
  213. Mani, S.A.; Guo, W.; Liao, M.-J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The Epithelial-Mesenchymal Transition Generates Cells with Properties of Stem Cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef] [PubMed]
  214. Ye, X.; Tam, W.L.; Shibue, T.; Kaygusuz, Y.; Reinhardt, F.; Ng Eaton, E.; Weinberg, R.A. Distinct EMT Programs Control Normal Mammary Stem Cells and Tumour-Initiating Cells. Nature 2015, 525, 256–260. [Google Scholar] [CrossRef]
  215. Sarkar, M.; Nguyen, T.; Gundre, E.; Ogunlusi, O.; El-Sobky, M.; Giri, B.; Sarkar, T.R. Cancer-Associated Fibroblasts: The Chief Architect in the Tumor Microenvironment. Front. Cell Dev. Biol. 2023, 11, 1089068. [Google Scholar] [CrossRef]
  216. Di Meo, N.A.; Lasorsa, F.; Rutigliano, M.; Loizzo, D.; Ferro, M.; Stella, A.; Bizzoca, C.; Vincenti, L.; Pandolfo, S.D.; Autorino, R.; et al. Renal Cell Carcinoma as a Metabolic Disease: An Update on Main Pathways, Potential Biomarkers, and Therapeutic Targets. Int. J. Mol. Sci. 2022, 23, 14360. [Google Scholar] [CrossRef]
  217. Lucarelli, G.; Rutigliano, M.; Ferro, M.; Giglio, A.; Intini, A.; Triggiano, F.; Palazzo, S.; Gigante, M.; Castellano, G.; Ranieri, E.; et al. Activation of the Kynurenine Pathway Predicts Poor Outcome in Patients with Clear Cell Renal Cell Carcinoma. Urol. Oncol. Semin. Orig. Investig. 2017, 35, 461.e15–461.e27. [Google Scholar] [CrossRef]
  218. Qian, J.; Rankin, E.B. Hypoxia-Induced Phenotypes That Mediate Tumor Heterogeneity. In Hypoxia and Cancer Metastasis; Gilkes, D.M., Ed.; Advances in Experimental Medicine and Biology; Springer International Publishing: Cham, Switzerland, 2019; Volume 1136, pp. 43–55. ISBN 978-3-030-12733-6. [Google Scholar]
  219. De Francesco, E.M.; Maggiolini, M.; Tanowitz, H.B.; Sotgia, F.; Lisanti, M.P. Targeting Hypoxic Cancer Stem Cells (CSCs) with Doxycycline: Implications for Optimizing Anti-Angiogenic Therapy. Oncotarget 2017, 8, 56126–56142. [Google Scholar] [CrossRef]
  220. Bonuccelli, G.; Peiris-Pages, M.; Ozsvari, B.; Martinez-Outschoorn, U.E.; Sotgia, F.; Lisanti, M.P. Targeting Cancer Stem Cell Propagation with Palbociclib, a CDK4/6 Inhibitor: Telomerase Drives Tumor Cell Heterogeneity. Oncotarget 2017, 8, 9868–9884. [Google Scholar] [CrossRef]
  221. García-Heredia, J.M.; Carnero, A. Role of Mitochondria in Cancer Stem Cell Resistance. Cells 2020, 9, 1693. [Google Scholar] [CrossRef]
  222. 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]
  223. Ye, X.-Q.; Wang, G.-H.; Huang, G.-J.; Bian, X.-W.; Qian, G.-S.; Yu, S.-C. Heterogeneity of Mitochondrial Membrane Potential: A Novel Tool to Isolate and Identify Cancer Stem Cells from a Tumor Mass? Stem Cell Rev. Rep. 2011, 7, 153–160. [Google Scholar] [CrossRef] [PubMed]
  224. Bonuccelli, G.; De Francesco, E.M.; De Boer, R.; Tanowitz, H.B.; Lisanti, M.P. NADH Autofluorescence, a New Metabolic Biomarker for Cancer Stem Cells: Identification of Vitamin C and CAPE as Natural Products Targeting “Stemness”. Oncotarget 2017, 8, 20667–20678. [Google Scholar] [CrossRef] [PubMed]
  225. Bao, B.; Wang, Z.; Ali, S.; Ahmad, A.; Azmi, A.S.; Sarkar, S.H.; Banerjee, S.; Kong, D.; Li, Y.; Thakur, S.; et al. Metformin Inhibits Cell Proliferation, Migration and Invasion by Attenuating CSC Function Mediated by Deregulating miRNAs in Pancreatic Cancer Cells. Cancer Prev. Res. 2012, 5, 355–364. [Google Scholar] [CrossRef] [PubMed]
  226. Wheaton, W.W.; Weinberg, S.E.; Hamanaka, R.B.; Soberanes, S.; Sullivan, L.B.; Anso, E.; Glasauer, A.; Dufour, E.; Mutlu, G.M.; Budigner, G.S.; et al. Metformin Inhibits Mitochondrial Complex I of Cancer Cells to Reduce Tumorigenesis. eLife 2014, 3, e02242. [Google Scholar] [CrossRef]
  227. Brown, J.R.; Chan, D.K.; Shank, J.J.; Griffith, K.A.; Fan, H.; Szulawski, R.; Yang, K.; Reynolds, R.K.; Johnston, C.; McLean, K.; et al. Phase II Clinical Trial of Metformin as a Cancer Stem Cell-Targeting Agent in Ovarian Cancer. JCI Insight 2020, 5, e133247. [Google Scholar] [CrossRef]
  228. Kaur, G.; Sharma, P.; Dogra, N.; Singh, S. Eradicating Cancer Stem Cells: Concepts, Issues, and Challenges. Curr. Treat. Options Oncol. 2018, 19, 20. [Google Scholar] [CrossRef]
  229. Dong, C.; Yuan, T.; Wu, Y.; Wang, Y.; Fan, T.W.M.; Miriyala, S.; Lin, Y.; Yao, J.; Shi, J.; Kang, T.; et al. Loss of FBP1 by Snail-Mediated Repression Provides Metabolic Advantages in Basal-like Breast Cancer. Cancer Cell 2013, 23, 316–331. [Google Scholar] [CrossRef]
  230. Flavahan, W.A.; Wu, Q.; Hitomi, M.; Rahim, N.; Kim, Y.; Sloan, A.E.; Weil, R.J.; Nakano, I.; Sarkaria, J.N.; Stringer, B.W.; et al. Brain Tumor Initiating Cells Adapt to Restricted Nutrition through Preferential Glucose Uptake. Nat. Neurosci. 2013, 16, 1373–1382. [Google Scholar] [CrossRef]
  231. Lucarelli, G.; Lasorsa, F.; Milella, M.; d’Amati, A.; Ingravallo, G.; Silecchia, M.; Errede, M.; Bianchi, C.; Spilotros, M.; Battaglia, M.; et al. Transcriptomic and Proteo-Metabolic Determinants of the Grading System in Clear Cell Renal Cell Carcinoma. Urol. Oncol. Semin. Orig. Investig. 2025; in press. [Google Scholar] [CrossRef]
Figure 1. Metabolic features of CSCs and related therapeutic targets. CSCs exhibit high metabolic flexibility, alternating between glycolysis and OXPHOS to support stemness and adapt to stress. Key pathways, including serine/glycine one-carbon metabolism, glutaminolysis, lipid metabolism, and the TCA cycle, are regulated by oncogenes like MYC, mTOR, and HIF-1α. Targeting these metabolic nodes offers promising strategies to eliminate CSCs and prevent tumor recurrence.
Figure 1. Metabolic features of CSCs and related therapeutic targets. CSCs exhibit high metabolic flexibility, alternating between glycolysis and OXPHOS to support stemness and adapt to stress. Key pathways, including serine/glycine one-carbon metabolism, glutaminolysis, lipid metabolism, and the TCA cycle, are regulated by oncogenes like MYC, mTOR, and HIF-1α. Targeting these metabolic nodes offers promising strategies to eliminate CSCs and prevent tumor recurrence.
Cells 14 00717 g001
Table 1. Putative metabolic strategies to target CSCs. OXPHOS: oxidative phosphorylation; FASN: Fatty Acid Synthase; CPT1: Carnitine Palmitoyltransferase 1; ROS: reactive species of oxygen; mTOR: mammalian target of rapamycin.
Table 1. Putative metabolic strategies to target CSCs. OXPHOS: oxidative phosphorylation; FASN: Fatty Acid Synthase; CPT1: Carnitine Palmitoyltransferase 1; ROS: reactive species of oxygen; mTOR: mammalian target of rapamycin.
Therapeutic StrategyDrugClinical Trials
Glycolysis2-DG
Lonidamine
NCT00633087
NCT02758860
OXPHOSMetformin
Oligomycin
Complex I inhibitors
NCT01579812
none
NCT02882321, NCT03291938
GlutaminolysisCB-839 (glutaminase inhibitor)NCT02071862, NCT03163667, NCT03872427
LipogenesisOrlistat (FASN inhibitor)none
Fatty acid β-oxidationEtomoxir (CPT1 inhibitor)none
ROS productionSalinomycin
Fenretinide
none
NCT00646230
mTORSirolimusNCT03433183
EverolimusNCT00876395, NCT01007942,
NCT04485559
TemsirolimusNCT01614197, NCT03433183
RidaforolimusNCT00538239
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

Milella, M.; Rutigliano, M.; Pandolfo, S.D.; Aveta, A.; Crocetto, F.; Ferro, M.; d’Amati, A.; Ditonno, P.; Lucarelli, G.; Lasorsa, F. The Metabolic Landscape of Cancer Stem Cells: Insights and Implications for Therapy. Cells 2025, 14, 717. https://doi.org/10.3390/cells14100717

AMA Style

Milella M, Rutigliano M, Pandolfo SD, Aveta A, Crocetto F, Ferro M, d’Amati A, Ditonno P, Lucarelli G, Lasorsa F. The Metabolic Landscape of Cancer Stem Cells: Insights and Implications for Therapy. Cells. 2025; 14(10):717. https://doi.org/10.3390/cells14100717

Chicago/Turabian Style

Milella, Martina, Monica Rutigliano, Savio Domenico Pandolfo, Achille Aveta, Felice Crocetto, Matteo Ferro, Antonio d’Amati, Pasquale Ditonno, Giuseppe Lucarelli, and Francesco Lasorsa. 2025. "The Metabolic Landscape of Cancer Stem Cells: Insights and Implications for Therapy" Cells 14, no. 10: 717. https://doi.org/10.3390/cells14100717

APA Style

Milella, M., Rutigliano, M., Pandolfo, S. D., Aveta, A., Crocetto, F., Ferro, M., d’Amati, A., Ditonno, P., Lucarelli, G., & Lasorsa, F. (2025). The Metabolic Landscape of Cancer Stem Cells: Insights and Implications for Therapy. Cells, 14(10), 717. https://doi.org/10.3390/cells14100717

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