Appearance
This report is written by MaltSci based on the latest literature and research findings
How does metabolic reprogramming drive cancer progression?
Abstract
Cancer remains a leading cause of morbidity and mortality globally, underscoring the urgent need to understand its underlying mechanisms. Among these, metabolic reprogramming has emerged as a hallmark of cancer, marked by profound alterations in cellular metabolism that enable cancer cells to sustain rapid proliferation and survive environmental challenges. This review synthesizes current knowledge on metabolic adaptations, including the shift from oxidative phosphorylation to aerobic glycolysis (the Warburg effect), altered lipid metabolism, and amino acid metabolism, which collectively enhance tumor growth and resistance to therapies. The role of oncogenes and tumor suppressor genes in modulating these pathways is crucial, as they regulate metabolic flexibility and influence tumor behavior. Additionally, the tumor microenvironment, with its complex interactions among non-cancerous cells, plays a significant role in shaping the metabolic landscape of tumors, often complicating treatment strategies. Targeting these unique metabolic needs offers promising therapeutic avenues, potentially leading to more effective cancer treatments. This review aims to provide a comprehensive overview of the mechanisms driving metabolic reprogramming in cancer, highlighting the importance of these adaptations in tumor progression and the development of innovative therapeutic strategies.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Mechanisms of Metabolic Reprogramming in Cancer
- 2.1 Aerobic Glycolysis and the Warburg Effect
- 2.2 Altered Lipid Metabolism
- 2.3 Amino Acid Metabolism and Nitrogen Source Utilization
- 3 Role of Oncogenes and Tumor Suppressors
- 3.1 Oncogenic Signaling Pathways
- 3.2 Tumor Suppressor Gene Functions in Metabolism
- 4 Impact of the Tumor Microenvironment
- 4.1 Hypoxia and Metabolic Adaptations
- 4.2 Interactions with Immune Cells
- 5 Therapeutic Implications
- 5.1 Targeting Metabolic Pathways
- 5.2 Combination Therapies with Metabolic Inhibitors
- 6 Future Directions and Research Gaps
- 6.1 Emerging Metabolic Targets
- 6.2 Personalized Medicine Approaches
- 7 Conclusion
1 Introduction
Cancer remains one of the leading causes of morbidity and mortality worldwide, necessitating an urgent need for a deeper understanding of its underlying mechanisms. Among these, metabolic reprogramming has emerged as a hallmark of cancer, characterized by the profound alterations in cellular metabolism that enable cancer cells to sustain rapid proliferation and survival in the face of various environmental challenges. This reprogramming is not merely a byproduct of cancerous transformation but is intricately linked to the ability of tumors to adapt to their microenvironment, evade apoptosis, and resist therapeutic interventions [1][2].
The significance of metabolic reprogramming in cancer progression cannot be overstated. As tumors develop, they undergo substantial shifts in metabolic pathways, transitioning from oxidative phosphorylation to aerobic glycolysis, enhancing lipogenesis, and altering amino acid metabolism. These changes not only provide the necessary energy and building blocks for macromolecule synthesis but also contribute to the tumor's ability to thrive in nutrient-poor and hypoxic conditions [3][4]. The understanding of these metabolic adaptations has opened new avenues for therapeutic strategies aimed at targeting the unique metabolic needs of cancer cells, potentially leading to more effective treatments [5][6].
Current research indicates that metabolic reprogramming is driven by a complex interplay of genetic mutations, oncogenic signaling pathways, and the tumor microenvironment. Oncogenes and tumor suppressor genes play pivotal roles in regulating these metabolic pathways, thus influencing tumor behavior and response to therapy [1][2]. Moreover, the tumor microenvironment, which consists of various non-cancerous cells and extracellular components, further complicates the metabolic landscape by exerting influences that can enhance or inhibit tumor growth [4][7].
This review is organized as follows: the first section will delve into the mechanisms of metabolic reprogramming in cancer, focusing on key processes such as aerobic glycolysis (the Warburg effect), altered lipid metabolism, and amino acid metabolism [2][2]. The subsequent section will explore the roles of oncogenes and tumor suppressors in modulating these metabolic pathways [1][4]. Following this, we will examine the impact of the tumor microenvironment on metabolic adaptations, including the effects of hypoxia and interactions with immune cells [4][7]. The therapeutic implications of targeting these metabolic pathways will then be discussed, highlighting potential strategies for combination therapies that include metabolic inhibitors [5][6]. Finally, we will identify future directions and research gaps in the field, emphasizing emerging metabolic targets and personalized medicine approaches [5][6].
In summary, understanding the intricacies of metabolic reprogramming in cancer is not only vital for elucidating the mechanisms underlying tumor progression but also holds promise for the development of innovative therapeutic strategies aimed at overcoming the challenges posed by this complex disease. Through this review, we aim to synthesize current knowledge and provide a comprehensive overview of the metabolic adaptations that drive cancer progression, thereby laying the groundwork for future research and therapeutic interventions.
2 Mechanisms of Metabolic Reprogramming in Cancer
2.1 Aerobic Glycolysis and the Warburg Effect
Metabolic reprogramming is a hallmark of cancer that significantly influences tumor progression and survival. One of the most well-studied aspects of this phenomenon is the preference of cancer cells for aerobic glycolysis, commonly referred to as the Warburg effect. This metabolic alteration enables tumor cells to convert glucose into lactate even in the presence of sufficient oxygen, a process that not only supports rapid proliferation but also contributes to the evasion of apoptosis and immune surveillance.
The Warburg effect is characterized by several key features that facilitate cancer progression. Firstly, it allows for a rapid generation of ATP and metabolic intermediates essential for biosynthetic processes, which are crucial for the growth and invasiveness of cancer cells. Key enzymes involved in this pathway include glucose transporters (GLUTs), hexokinases (HKs), phosphofructokinases (PFKs), lactate dehydrogenases (LDHs), and pyruvate kinase M2 (PKM2) [8]. These enzymes not only regulate glycolysis but also influence other metabolic pathways, contributing to the overall metabolic flexibility of cancer cells.
Moreover, the expression of various transcriptional regulatory factors, such as FOXM1, p53, NF-κB, HIF1α, and c-Myc, plays a critical role in modulating the Warburg effect. For instance, the tumor suppressor protein p53 has been shown to negatively influence the Warburg effect, suggesting that its loss can lead to enhanced aerobic glycolysis and promote oncogenic metabolic adaptation [9].
Additionally, metabolic reprogramming is closely linked to the tumor microenvironment. Cancer cells often exist in a hypoxic environment, which further drives the reliance on glycolysis. The acidic microenvironment generated by lactate production not only supports tumor growth but also facilitates immune evasion by inhibiting T cell function and promoting an immunosuppressive milieu [10].
Research indicates that the metabolic adaptations associated with the Warburg effect can lead to chemoresistance, complicating treatment strategies. By relying heavily on glycolysis, cancer cells can develop resistance to therapies that target oxidative phosphorylation or other metabolic pathways [11]. Furthermore, the interplay between cancer cells and immune cells in the tumor microenvironment often results in competition for nutrients, which can further exacerbate tumor growth and survival [8].
In summary, metabolic reprogramming, particularly through the Warburg effect, drives cancer progression by enhancing the proliferative capacity of tumor cells, enabling them to survive in hostile environments, and promoting resistance to therapies. Understanding these mechanisms provides insights into potential therapeutic targets aimed at disrupting the metabolic advantages conferred by aerobic glycolysis, offering new avenues for cancer treatment [12].
2.2 Altered Lipid Metabolism
Metabolic reprogramming is increasingly recognized as a hallmark of cancer, particularly in the context of altered lipid metabolism, which plays a critical role in driving cancer progression. The reprogramming of lipid metabolism in cancer cells involves a complex interplay of biochemical pathways that facilitate tumor growth, survival, and metastasis.
Lipid metabolism encompasses various processes including lipid uptake, synthesis, transport, and degradation. In cancer, these processes are often dysregulated, leading to enhanced lipid synthesis and accumulation. For instance, cancer cells exhibit increased de novo fatty acid synthesis, elevated lipid uptake from the surrounding environment, and altered fatty acid oxidation, all of which contribute to their metabolic demands for rapid proliferation and survival in adverse conditions such as nutrient deprivation and hypoxia [13][14].
The altered lipid metabolic pathways not only provide energy and structural components for cell membranes but also serve as signaling molecules that influence oncogenic signaling pathways. Dysregulated lipid metabolism has been linked to the activation of oncogenic pathways that promote cell proliferation and survival, thereby exacerbating cancer progression [15][16]. For example, lipids act as second messengers in various signaling cascades, and their aberrant accumulation can lead to altered cellular signaling that favors tumorigenesis [17].
Furthermore, lipid metabolic reprogramming impacts the tumor microenvironment (TME), which includes stromal and immune cells. The reprogramming of lipid metabolism in these non-cancerous cells can further modulate tumor behavior, enhancing tumor growth and immune evasion [18][19]. For instance, tumor-associated adipocytes can influence lipid metabolism in cancer cells, contributing to therapeutic resistance and promoting a supportive microenvironment for tumor progression [20].
The role of non-coding RNAs, such as long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), in regulating lipid metabolic reprogramming has also gained attention. These molecules can modulate key metabolic pathways and influence the overall lipid metabolism landscape within tumors, thereby affecting cancer cell behavior and therapeutic responses [16].
Recent studies have highlighted potential therapeutic strategies targeting lipid metabolism as a means to inhibit cancer progression. These strategies include the use of small-molecule inhibitors that specifically target enzymes involved in lipid synthesis and oxidation, aiming to disrupt the metabolic adaptations that cancer cells rely on for growth and survival [14][21].
In summary, metabolic reprogramming, particularly through altered lipid metabolism, drives cancer progression by providing the necessary energy and building blocks for rapid tumor growth, modulating oncogenic signaling pathways, and influencing the tumor microenvironment. Understanding these mechanisms offers valuable insights into potential therapeutic interventions that target metabolic pathways to improve cancer treatment outcomes.
2.3 Amino Acid Metabolism and Nitrogen Source Utilization
Metabolic reprogramming is a fundamental characteristic of cancer that enables tumor cells to adapt to their microenvironment, sustain rapid growth, and evade therapeutic interventions. A critical aspect of this reprogramming is the alteration of amino acid metabolism, which plays a significant role in supporting the anabolic demands of cancer cells.
Amino acids serve as essential building blocks for proteins and are critical for various cellular functions, including energy metabolism, nucleotide synthesis, and maintaining redox balance. Cancer cells often exhibit an increased dependence on specific amino acids, which can be attributed to their altered metabolic pathways that are adapted to fulfill their heightened nutritional requirements. For instance, the uptake and metabolism of branched-chain amino acids (BCAAs)—valine, leucine, and isoleucine—are significantly upregulated in many cancers. These amino acids not only act as nitrogen donors for macromolecule synthesis but also influence key oncogenic processes, thereby promoting tumor growth and malignancy (Peng et al. 2020) [22].
Moreover, amino acid metabolic reprogramming is closely linked to the tumor microenvironment (TME), where cancer cells often face stress conditions such as hypoxia and nutrient deprivation. In response, they modify their amino acid metabolism to support survival and proliferation. For example, the dysregulation of amino acid metabolism can lead to immune evasion by modulating immune cell function within the TME. This immune suppression is partly facilitated by the metabolism of tryptophan to kynurenine, which has been shown to inhibit CD8+ T cell activity (Zhang et al. 2025) [23].
The mechanisms driving these metabolic alterations include various signaling pathways and transcription factors that respond to the oncogenic signals present in the tumor. The PI3K/AKT/mTOR and Wnt/β-catenin pathways, for instance, have been identified as crucial regulators of amino acid metabolism, promoting not only cell growth but also chemoresistance through metabolic adaptations (Wang et al. 2025) [19].
Furthermore, amino acids also play a pivotal role in the metabolic interactions within the TME, influencing both tumor cell behavior and the surrounding immune cells. This interplay highlights the importance of targeting amino acid metabolism as a therapeutic strategy. By disrupting the supply or utilization of specific amino acids, it may be possible to induce apoptosis in tumor cells and enhance the efficacy of existing therapies (Ge et al. 2025) [24].
In summary, metabolic reprogramming, particularly through the lens of amino acid metabolism, is integral to cancer progression. It not only supports the biosynthetic needs of rapidly dividing tumor cells but also facilitates immune evasion and therapeutic resistance, making it a critical area for developing novel cancer therapies. Understanding the nuances of these metabolic pathways and their interactions with the TME can pave the way for innovative approaches in cancer treatment, focusing on exploiting the metabolic vulnerabilities of tumors.
3 Role of Oncogenes and Tumor Suppressors
3.1 Oncogenic Signaling Pathways
Metabolic reprogramming is a fundamental characteristic of cancer that plays a crucial role in driving cancer progression. This process is intricately linked to the activation of oncogenes and the suppression of tumor suppressor genes, both of which are key players in the regulation of metabolic pathways within tumor cells. The dysregulation of these pathways allows cancer cells to adapt to their environment, facilitating their growth, proliferation, and survival.
Oncogenic signaling pathways significantly influence metabolic reprogramming by altering the expression and activity of various metabolic enzymes. For instance, the activation of the phosphoinositide 3-kinase (PI3K)/AKT/mTOR pathway is commonly observed in many cancers. This pathway enhances glycolysis and promotes the synthesis of lipids and nucleotides, which are essential for rapid cell proliferation and growth. Additionally, the PI3K/AKT/mTOR signaling cascade is known to upregulate the expression of genes involved in metabolic processes, thereby enabling cancer cells to meet their increased bioenergetic and biosynthetic demands [25].
Moreover, oncogenes such as Myc and mutations in tumor suppressor genes like p53 can further drive metabolic reprogramming. Myc, for example, regulates the expression of genes involved in glycolysis and glutaminolysis, promoting an environment conducive to tumor growth. The loss of p53 function leads to increased aerobic glycolysis and altered mitochondrial metabolism, which supports the survival of cancer cells under stress conditions [2].
The tumor microenvironment (TME) also plays a pivotal role in metabolic reprogramming. Factors such as hypoxia and nutrient availability can influence both intrinsic and extrinsic signaling pathways that modify the metabolic state of tumor cells. For example, hypoxia-inducible factors (HIFs) are activated in low-oxygen conditions and can induce the expression of genes that facilitate glycolysis and angiogenesis, further enhancing tumor survival and growth [26].
In addition to these intrinsic changes, the interactions between cancer cells and surrounding stromal cells, immune cells, and extracellular matrix components contribute to the metabolic landscape of tumors. These interactions can create a supportive microenvironment that promotes metabolic adaptations, enabling cancer cells to thrive despite the harsh conditions often present in solid tumors [27].
Overall, metabolic reprogramming driven by oncogenic signaling pathways and the loss of tumor suppressor function is essential for cancer progression. This process not only supports the energetic and biosynthetic needs of rapidly dividing cancer cells but also facilitates their ability to evade immune surveillance and resist conventional therapies. Understanding these mechanisms offers potential avenues for therapeutic interventions aimed at targeting metabolic vulnerabilities in cancer cells [7][14].
3.2 Tumor Suppressor Gene Functions in Metabolism
Metabolic reprogramming is a fundamental characteristic of cancer that facilitates tumor progression by altering the metabolic processes of cancer cells to meet their increased demands for energy and macromolecular precursors. This reprogramming is significantly influenced by the functions of oncogenes and tumor suppressor genes, which play critical roles in regulating cellular metabolism.
Oncogenes, when activated, can promote metabolic pathways that support the aggressive growth and survival of cancer cells. For instance, oncogenic mutations often lead to enhanced glucose uptake and aerobic glycolysis, a phenomenon known as the Warburg effect, where cancer cells preferentially utilize glycolysis for energy production even in the presence of oxygen. This metabolic shift allows cancer cells to generate ATP rapidly and provides intermediates for biosynthetic processes necessary for cell proliferation [28].
Conversely, tumor suppressor genes typically function to inhibit excessive cell growth and maintain metabolic homeostasis. Loss of function or mutations in these genes can disrupt normal metabolic processes, leading to uncontrolled cellular proliferation and tumorigenesis. For example, the inactivation of tumor suppressor genes such as TP53 can result in the dysregulation of metabolic pathways, facilitating cancer cell survival under stress conditions [2]. Moreover, tumor suppressors are involved in maintaining cellular responses to metabolic stress and apoptosis; their loss can lead to metabolic adaptations that promote tumor survival and resistance to therapies [26].
The interplay between oncogenes and tumor suppressor genes creates a metabolic landscape that favors tumor progression. Oncogenes can drive metabolic reprogramming by activating signaling pathways such as the phosphoinositide 3-kinase (PI3K)/AKT pathway, which enhances metabolic enzyme activity and promotes anabolic processes essential for tumor growth [14]. Meanwhile, the loss of tumor suppressor function can exacerbate these effects by allowing cancer cells to bypass growth inhibitory signals and resist apoptotic cues, thereby sustaining their metabolic demands and facilitating metastasis [29].
Furthermore, metabolic reprogramming also impacts the tumor microenvironment, influencing the interactions between cancer cells and surrounding stromal cells, including immune cells. The altered metabolism in cancer cells can lead to the secretion of metabolites that modulate immune responses, potentially promoting an immunosuppressive environment conducive to tumor progression [7].
In summary, metabolic reprogramming drives cancer progression through a complex interplay between oncogenes and tumor suppressor genes, which together influence metabolic pathways critical for cell growth, survival, and interaction with the tumor microenvironment. Understanding these mechanisms is vital for developing targeted therapeutic strategies aimed at disrupting the metabolic adaptations of cancer cells [30][31].
4 Impact of the Tumor Microenvironment
4.1 Hypoxia and Metabolic Adaptations
Metabolic reprogramming is a fundamental characteristic of cancer progression, significantly influenced by the tumor microenvironment (TME), particularly under hypoxic conditions. Hypoxia, defined as a deficiency in oxygen availability, is prevalent in many solid tumors due to rapid tumor growth and abnormal vasculature. This oxygen deprivation triggers various adaptive responses in cancer cells, leading to significant metabolic alterations that facilitate tumor survival, growth, and resistance to therapies.
One of the primary adaptations to hypoxia is the shift from oxidative phosphorylation to glycolysis, a phenomenon known as the Warburg effect. This metabolic switch allows cancer cells to generate energy in the form of ATP under low oxygen conditions, albeit less efficiently than aerobic respiration. Enhanced glycolysis not only provides ATP but also produces metabolites that can be utilized by neighboring normoxic cells, thereby promoting a competitive metabolic environment that supports tumor expansion [32].
The role of hypoxia-inducible factors (HIFs), particularly HIF-1α, is critical in this metabolic reprogramming. HIF-1α acts as a transcription factor that regulates genes involved in glucose metabolism, angiogenesis, and cell survival. Its activation leads to increased expression of glucose transporters and glycolytic enzymes, enabling cancer cells to adapt their metabolism to the nutrient-depleted and hypoxic TME [33]. Furthermore, HIF-1α also promotes the uptake of alternative substrates such as glutamine and fatty acids, which are vital for maintaining cellular metabolism and supporting biosynthetic processes necessary for rapid cell proliferation [32].
Hypoxia-induced metabolic changes also have profound implications for the immune landscape within the TME. The metabolic adaptations of cancer cells can induce immunosuppressive conditions that hinder effective anti-tumor immune responses. For instance, the accumulation of metabolic byproducts such as lactate can inhibit the function of immune cells, leading to a TME that favors tumor growth and progression [34]. Additionally, the altered metabolic state can promote the differentiation of immune cells into regulatory phenotypes that further suppress anti-tumor immunity [35].
Moreover, metabolic reprogramming in cancer cells enhances their ability to survive conventional therapies, such as chemotherapy and radiation. By modifying their metabolic pathways, cancer cells can evade the cytotoxic effects of these treatments, contributing to therapy resistance and tumor recurrence [36]. This highlights the importance of understanding the interplay between metabolic reprogramming and the TME in developing effective cancer therapies.
In summary, metabolic reprogramming driven by hypoxia plays a crucial role in cancer progression by facilitating tumor cell survival and proliferation, altering the immune microenvironment, and contributing to therapeutic resistance. The dynamic interactions between cancer cells and their TME underscore the need for targeted strategies that address these metabolic alterations to improve cancer treatment outcomes [36][37][38].
4.2 Interactions with Immune Cells
Metabolic reprogramming is a fundamental process by which cancer cells adapt their metabolism to support rapid growth and survival in challenging microenvironments. This reprogramming is characterized by altered energy production pathways, which allow tumor cells to thrive despite limited nutrient availability and hypoxic conditions. The tumor microenvironment (TME) plays a crucial role in facilitating these metabolic changes and significantly impacts cancer progression, particularly through its interactions with immune cells.
In the context of cancer, metabolic reprogramming often leads to a shift towards aerobic glycolysis, known as the Warburg effect, where cancer cells preferentially convert glucose to lactate even in the presence of oxygen. This metabolic alteration is not only beneficial for tumor growth but also influences the surrounding TME, creating an environment conducive to tumor progression. For instance, tumor cells secrete various metabolites, such as lactate and pyruvate, which can induce local acidosis and nutrient competition, ultimately leading to immunosuppression and promoting cancer cell survival (Yang et al. 2025; Jia et al. 2024).
The interactions between tumor cells and immune cells within the TME are profoundly affected by metabolic reprogramming. Tumor cells can modulate the metabolism of immune cells, leading to functional impairments. For example, the increased production of lactate in the TME can inhibit T cell activation and proliferation, while promoting the differentiation of regulatory T cells (Tregs), which further suppresses anti-tumor immune responses (Yan et al. 2021; Shi et al. 2020). This metabolic interplay results in a dynamic where immune cells, rather than effectively combating tumor growth, become co-opted to support tumor progression.
Moreover, the TME is characterized by the presence of various non-cancerous cells, including cancer-associated fibroblasts (CAFs) and immune cells, which also undergo metabolic reprogramming. CAFs, for instance, can influence the metabolic state of tumor cells by secreting cytokines and other factors that promote a more aggressive metabolic phenotype in cancer cells. This interaction leads to a competitive environment for nutrients, further exacerbating the metabolic stress on immune cells and diminishing their efficacy in targeting tumor cells (Wegiel et al. 2018; Aden et al. 2025).
The implications of these metabolic interactions are profound. As the TME becomes increasingly immunosuppressive due to metabolic alterations, the efficacy of immunotherapies is often compromised. Tumor cells exploit metabolic pathways not only for their growth but also to create a hostile environment for immune cells, leading to treatment resistance (Liu et al. 2024; Xia et al. 2021). Therefore, understanding the mechanisms of metabolic reprogramming and its effects on immune cell function is critical for developing novel therapeutic strategies aimed at reversing this immunosuppression and enhancing the effectiveness of cancer treatments.
In summary, metabolic reprogramming drives cancer progression by reshaping the TME and altering the interactions between tumor and immune cells. The resulting metabolic competition and immune evasion create a favorable niche for tumor growth, highlighting the need for targeted therapies that address these metabolic pathways to improve clinical outcomes in cancer patients.
5 Therapeutic Implications
5.1 Targeting Metabolic Pathways
Metabolic reprogramming is a critical feature of cancer cells that facilitates their growth, survival, and resistance to therapies. This phenomenon involves the alteration of various metabolic pathways, including glucose, lipid, and amino acid metabolism, enabling cancer cells to meet their increased energy and biosynthetic demands. Understanding the mechanisms of metabolic reprogramming is essential for developing therapeutic strategies aimed at targeting these altered pathways.
In the context of cancer progression, metabolic reprogramming allows tumor cells to adapt to the harsh microenvironment characterized by limited nutrients and hypoxia. For instance, cancer cells often exhibit enhanced glycolysis, even in the presence of oxygen (aerobic glycolysis), which is known as the Warburg effect. This metabolic shift supports rapid ATP production and the generation of metabolic intermediates necessary for biosynthesis, promoting tumor proliferation and invasion [19].
Furthermore, the interaction between cancer cells and the tumor microenvironment (TME) plays a significant role in driving metabolic reprogramming. Tumor cells can influence the metabolism of surrounding stromal cells, leading to a cooperative metabolic network that supports tumor growth and immune evasion [39]. This metabolic crosstalk not only aids in tumor survival but also modulates immune cell function, creating an immunosuppressive environment that further facilitates cancer progression [40].
From a therapeutic standpoint, targeting metabolic pathways presents a promising strategy to counteract cancer progression and enhance treatment efficacy. By inhibiting key metabolic enzymes and pathways, researchers aim to disrupt the metabolic adaptations that cancer cells rely on for growth and survival. For example, inhibitors targeting the mTOR and PI3K pathways, which are often dysregulated in cancer, have shown potential in clinical settings [41].
Combination therapies that integrate metabolic inhibitors with traditional treatments such as chemotherapy or immunotherapy are being explored to improve therapeutic outcomes. By addressing the metabolic vulnerabilities of cancer cells, these strategies could enhance the effectiveness of existing therapies and overcome resistance mechanisms [42]. Additionally, emerging approaches such as nanomedicine are being investigated to deliver metabolic modulators specifically to tumor sites, thereby maximizing their impact while minimizing systemic toxicity [43].
In conclusion, metabolic reprogramming is a fundamental driver of cancer progression, influencing tumor growth, metastasis, and therapeutic resistance. Targeting the altered metabolic pathways offers a novel therapeutic avenue, with the potential to improve patient outcomes by addressing the metabolic dependencies of cancer cells. Continued research into the intricate relationship between metabolism and cancer will be essential for the development of effective treatment strategies.
5.2 Combination Therapies with Metabolic Inhibitors
Metabolic reprogramming is a fundamental characteristic of cancer cells that facilitates their uncontrolled growth, survival, and adaptation to the tumor microenvironment. This phenomenon involves the alteration of various metabolic pathways, enabling tumor cells to meet their increased energy and biosynthetic demands. Such reprogramming is not only pivotal for cancer progression but also contributes to therapeutic resistance, posing significant challenges in effective treatment strategies.
Cancer cells often undergo metabolic shifts, such as increased reliance on aerobic glycolysis, known as the Warburg effect, where they preferentially convert glucose to lactate even in the presence of oxygen. This metabolic adaptation supports rapid cell proliferation and helps to create an acidic tumor microenvironment that can inhibit immune responses. Additionally, changes in lipid metabolism and amino acid metabolism play critical roles in sustaining tumor growth and facilitating metastasis. For instance, the dysregulation of lipid synthesis and oxidation can enhance tumor cell survival and promote an immunosuppressive environment by influencing the activity of immune cells within the tumor microenvironment (TME) [19][40].
The implications of these metabolic alterations extend to therapeutic strategies. Targeting metabolic pathways presents a promising approach to enhance the efficacy of existing treatments. Combination therapies that incorporate metabolic inhibitors alongside traditional therapies such as chemotherapy, targeted therapy, or immunotherapy have shown potential in overcoming drug resistance and improving treatment outcomes. For example, inhibitors of glycolysis, lipid metabolism, and glutamine metabolism are being explored as adjunctive treatments to enhance the efficacy of conventional therapies [5][39].
Recent studies have highlighted the synergistic effects of combining metabolic inhibitors with immunotherapeutic agents. By targeting the metabolic pathways that drive immune evasion, such as those involved in the production of immunosuppressive metabolites, these combination therapies aim to restore the functionality of immune cells and enhance their ability to combat tumor cells. This approach not only addresses the metabolic needs of tumor cells but also modifies the TME to be more conducive to immune activity [35][40].
Furthermore, the development of nanoscale drug delivery systems has emerged as a novel strategy to enhance the delivery of metabolic inhibitors directly to tumor sites, thereby minimizing systemic toxicity and maximizing therapeutic efficacy. These systems can be designed to respond to specific tumor microenvironmental cues, allowing for more precise targeting of metabolic pathways and improving the overall effectiveness of cancer treatments [35][44].
In conclusion, metabolic reprogramming is a critical driver of cancer progression, influencing not only tumor growth and survival but also the response to therapy. The integration of metabolic inhibitors into combination therapies represents a promising avenue for enhancing cancer treatment efficacy, particularly in the context of overcoming drug resistance and improving immunotherapeutic outcomes. Continued research into the intricate relationship between metabolism and cancer will be essential for developing innovative therapeutic strategies that can effectively target these metabolic alterations.
6 Future Directions and Research Gaps
6.1 Emerging Metabolic Targets
Metabolic reprogramming is increasingly recognized as a pivotal mechanism driving cancer progression, influencing tumor growth, metastasis, and therapeutic resistance. Cancer cells undergo significant alterations in their metabolic pathways to meet the heightened energy and biosynthetic demands associated with rapid proliferation. These changes include enhanced glycolysis, altered lipid metabolism, and increased reliance on glutamine and other nutrients, which collectively support malignant transformation and tumor development [2][41][45].
One of the fundamental aspects of metabolic reprogramming is the Warburg effect, where cancer cells preferentially utilize aerobic glycolysis over oxidative phosphorylation, even in the presence of sufficient oxygen. This shift not only allows for faster ATP production but also facilitates the generation of metabolic intermediates necessary for biosynthesis, which are crucial for cell growth and division [6][44]. Furthermore, metabolic alterations enable cancer cells to adapt to the nutrient-depleted and hypoxic tumor microenvironment, contributing to their survival and proliferation under adverse conditions [3][5].
Despite the advancements in understanding the role of metabolic reprogramming in cancer, several future directions and research gaps remain. There is a pressing need to elucidate the intricate regulatory networks governing metabolic changes in different cancer types and their interactions with the tumor microenvironment. A deeper understanding of how these metabolic pathways are orchestrated at the molecular level could reveal new therapeutic targets and strategies [19][46]. Moreover, exploring the dynamics of metabolic reprogramming during cancer progression and therapy response could provide insights into the temporal aspects of metabolic changes and their implications for treatment efficacy [5][45].
Emerging metabolic targets for cancer therapy include key enzymes involved in glycolysis, lipid metabolism, and amino acid utilization. Inhibitors targeting these pathways have shown promise in preclinical studies and are advancing into clinical trials. For instance, the inhibition of lactate dehydrogenase A, hexokinase, and mTOR pathways has been explored as potential strategies to disrupt the metabolic adaptations of cancer cells [46][47]. Additionally, targeting the metabolic crosstalk between cancer cells and the tumor microenvironment represents a novel therapeutic avenue, aiming to enhance the efficacy of existing treatments while mitigating resistance [5][44].
In conclusion, metabolic reprogramming is a crucial contributor to cancer progression, offering numerous avenues for research and therapeutic intervention. Addressing the existing gaps in our understanding of cancer metabolism and exploring new metabolic targets could pave the way for innovative treatment strategies, ultimately improving patient outcomes in oncology.
6.2 Personalized Medicine Approaches
Metabolic reprogramming is a crucial hallmark of cancer that facilitates tumor progression by enabling cancer cells to adapt their metabolism to meet increased demands for energy and biosynthetic precursors. This process involves significant alterations in metabolic pathways, including aerobic glycolysis, glutaminolysis, and lipid metabolism, which collectively support malignant behaviors such as uncontrolled proliferation, invasion, and drug resistance [6][28][48].
In the context of cancer progression, metabolic reprogramming allows tumor cells to thrive in the challenging tumor microenvironment (TME). For instance, cancer cells often switch to aerobic glycolysis, even in the presence of oxygen, a phenomenon known as the Warburg effect. This shift not only enhances ATP production but also provides essential building blocks for rapidly dividing cells [5][49]. Furthermore, metabolic changes enable cancer cells to engage in crosstalk with surrounding non-malignant cells, influencing immune responses and promoting a supportive microenvironment that facilitates tumor growth and metastasis [7][39].
Research indicates that different cancer types exhibit distinct metabolic signatures, which can evolve during disease progression, leading to heterogeneity in metabolic vulnerabilities [2][50]. This heterogeneity poses challenges for developing effective therapeutic strategies, as the metabolic properties of tumors can differ significantly between primary and metastatic sites, even within the same patient [5][51].
Future directions in cancer research should focus on addressing these metabolic adaptations to improve treatment outcomes. Identifying specific metabolic vulnerabilities can pave the way for personalized medicine approaches, where therapies are tailored based on the unique metabolic profiles of individual tumors. For example, targeting specific enzymes involved in glycolysis or lipid metabolism may enhance the efficacy of conventional therapies and overcome resistance mechanisms [48][52].
Moreover, integrating metabolic profiling with other diagnostic tools could facilitate the development of predictive biomarkers, allowing for a more precise and personalized treatment regimen [48][53]. As metabolic reprogramming continues to be recognized as a promising target for therapeutic intervention, ongoing research should also aim to elucidate the intricate regulatory networks governing cancer metabolism, thereby enhancing our understanding of tumor biology and improving the efficacy of targeted therapies [5][6].
In conclusion, metabolic reprogramming is a dynamic and complex process that drives cancer progression through various mechanisms. The ongoing exploration of metabolic pathways in cancer, coupled with advancements in personalized medicine, holds the potential to revolutionize cancer treatment and improve patient outcomes. Future studies should aim to fill the gaps in understanding the interplay between metabolic reprogramming and therapeutic resistance, ultimately leading to more effective and individualized cancer therapies.
7 Conclusion
Metabolic reprogramming has emerged as a critical hallmark of cancer, driving tumor progression through a variety of mechanisms that alter cellular metabolism to support rapid growth and survival. This review highlights several key findings: first, the Warburg effect exemplifies how cancer cells preferentially utilize aerobic glycolysis to generate energy and biosynthetic intermediates, facilitating their proliferation even under hypoxic conditions. Second, altered lipid metabolism plays a vital role in providing the necessary building blocks for cell membranes and signaling molecules, further supporting tumor growth and contributing to immune evasion. Third, amino acid metabolism is crucial for meeting the anabolic demands of cancer cells and can modulate immune responses within the tumor microenvironment. The roles of oncogenes and tumor suppressor genes are pivotal in regulating these metabolic pathways, with oncogenic signaling promoting metabolic adaptations that favor tumor survival, while the loss of tumor suppressor function exacerbates these effects. The tumor microenvironment, characterized by hypoxia and nutrient competition, significantly influences these metabolic changes, often leading to therapeutic resistance. Future research should focus on identifying emerging metabolic targets and developing personalized medicine approaches to exploit the unique metabolic vulnerabilities of individual tumors. By targeting the metabolic adaptations of cancer cells, we can enhance treatment efficacy and improve patient outcomes in oncology.
References
- [1] Jung-Ae Kim;Young Il Yeom. Metabolic Signaling to Epigenetic Alterations in Cancer.. Biomolecules & therapeutics(IF=3.2). 2018. PMID:29212308. DOI: 10.4062/biomolther.2017.185.
- [2] Shiqi Nong;Xiaoyue Han;Yu Xiang;Yuran Qian;Yuhao Wei;Tingyue Zhang;Keyue Tian;Kai Shen;Jing Yang;Xuelei Ma. Metabolic reprogramming in cancer: Mechanisms and therapeutics.. MedComm(IF=10.7). 2023. PMID:36994237. DOI: 10.1002/mco2.218.
- [3] Kenji Ohshima;Eiichi Morii. Metabolic Reprogramming of Cancer Cells during Tumor Progression and Metastasis.. Metabolites(IF=3.7). 2021. PMID:33401771. DOI: 10.3390/metabo11010028.
- [4] Haoqiang Yang;Jinghui Li;Yiting Niu;Tao Zhou;Pengyu Zhang;Yang Liu;Yanjun Li. Interactions between the metabolic reprogramming of liver cancer and tumor microenvironment.. Frontiers in immunology(IF=5.9). 2025. PMID:40028341. DOI: 10.3389/fimmu.2025.1494788.
- [5] Rilan Bai;Ying Meng;Jiuwei Cui. Therapeutic strategies targeting metabolic characteristics of cancer cells.. Critical reviews in oncology/hematology(IF=5.6). 2023. PMID:37236409. DOI: 10.1016/j.critrevonc.2023.104037.
- [6] Yunhan Zhu;Weijie Yan;Lingfeng Tong;Jie Yang;Shengfang Ge;Jiayan Fan;Renbing Jia;Xuyang Wen. Metabolic Reprogramming: A Crucial Contributor to Anticancer Drug Resistance.. MedComm(IF=10.7). 2025. PMID:40919131. DOI: 10.1002/mco2.70358.
- [7] Xihe Zhao;Kai Li;Mengyu Chen;Lei Liu. Metabolic codependencies in the tumor microenvironment and gastric cancer: Difficulties and opportunities.. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie(IF=7.5). 2023. PMID:36989719. DOI: 10.1016/j.biopha.2023.114601.
- [8] Minru Liao;Dahong Yao;Lifeng Wu;Chaodan Luo;Zhiwen Wang;Jin Zhang;Bo Liu. Targeting the Warburg effect: A revisited perspective from molecular mechanisms to traditional and innovative therapeutic strategies in cancer.. Acta pharmaceutica Sinica. B(IF=14.6). 2024. PMID:38487001. DOI: 10.1016/j.apsb.2023.12.003.
- [9] Ana Sara Gomes;Helena Ramos;Joana Soares;Lucília Saraiva. p53 and glucose metabolism: an orchestra to be directed in cancer therapy.. Pharmacological research(IF=10.5). 2018. PMID:29580896. DOI: 10.1016/j.phrs.2018.03.015.
- [10] Smitha R Pillai;Mehdi Damaghi;Yoshinori Marunaka;Enrico Pierluigi Spugnini;Stefano Fais;Robert J Gillies. Causes, consequences, and therapy of tumors acidosis.. Cancer metastasis reviews(IF=8.7). 2019. PMID:30911978. DOI: 10.1007/s10555-019-09792-7.
- [11] Chaithanya Chelakkot;Vipin Shankar Chelakkot;Youngkee Shin;Kyoung Song. Modulating Glycolysis to Improve Cancer Therapy.. International journal of molecular sciences(IF=4.9). 2023. PMID:36768924. DOI: 10.3390/ijms24032606.
- [12] Reo Hamaguchi;Masahide Isowa;Ryoko Narui;Hiromasa Morikawa;Toshihiro Okamoto;Hiromi Wada. How Does Cancer Occur? How Should It Be Treated? Treatment from the Perspective of Alkalization Therapy Based on Science-Based Medicine.. Biomedicines(IF=3.9). 2024. PMID:39457509. DOI: 10.3390/biomedicines12102197.
- [13] Rasha Irshad;Sazi Tabassum;Mohammad Husain. Aberrant Lipid Metabolism in Cancer: Current Status and Emerging Therapeutic Perspectives.. Current topics in medicinal chemistry(IF=3.3). 2023. PMID:37218199. DOI: 10.2174/1568026623666230522103321.
- [14] Do-Hee Kim;Na-Young Song;Hyungshin Yim. Targeting dysregulated lipid metabolism in the tumor microenvironment.. Archives of pharmacal research(IF=7.5). 2023. PMID:38060103. DOI: 10.1007/s12272-023-01473-y.
- [15] Yan Fu;Tiantian Zou;Xiaotian Shen;Peter J Nelson;Jiahui Li;Chao Wu;Jimeng Yang;Yan Zheng;Christiane Bruns;Yue Zhao;Lunxiu Qin;Qiongzhu Dong. Lipid metabolism in cancer progression and therapeutic strategies.. MedComm(IF=10.7). 2021. PMID:34766135. DOI: 10.1002/mco2.27.
- [16] Shanshan Liu;Benzheng Jiao;Hongguang Zhao;Xinyue Liang;Fengyan Jin;Xiaodong Liu;Ji-Fan Hu. LncRNAs-circRNAs as Rising Epigenetic Binary Superstars in Regulating Lipid Metabolic Reprogramming of Cancers.. Advanced science (Weinheim, Baden-Wurttemberg, Germany)(IF=14.1). 2024. PMID:37939296. DOI: 10.1002/advs.202303570.
- [17] Junzhe Zhao;Keene Lee;Han Chong Toh;Kong Peng Lam;Shi Yong Neo. Unravelling the role of obesity and lipids during tumor progression.. Frontiers in pharmacology(IF=4.8). 2023. PMID:37063269. DOI: 10.3389/fphar.2023.1163160.
- [18] Hao-Ran Jin;Jin Wang;Zi-Jing Wang;Ming-Jia Xi;Bi-Han Xia;Kai Deng;Jin-Lin Yang. Lipid metabolic reprogramming in tumor microenvironment: from mechanisms to therapeutics.. Journal of hematology & oncology(IF=40.4). 2023. PMID:37700339. DOI: 10.1186/s13045-023-01498-2.
- [19] Haibo Wang;Yu Lun;Dongdong Xu;Han Jiang;Yumeng Yan;Xing Yang. Research progress and therapeutic strategies in hepatocellular carcinoma metabolic reprogramming.. Journal of advanced research(IF=13.0). 2025. PMID:40850678. DOI: 10.1016/j.jare.2025.08.023.
- [20] Yihai Cao. Adipocyte and lipid metabolism in cancer drug resistance.. The Journal of clinical investigation(IF=13.6). 2019. PMID:31264969. DOI: .
- [21] Jiahao Zhang;Sha Xu;Lingfeng Yue;Hongrui Lei;Xin Zhai. A Collection of Novel Antitumor Agents That Regulate Lipid Metabolism in the Tumor Microenvironment.. Journal of medicinal chemistry(IF=6.8). 2025. PMID:39726379. DOI: 10.1021/acs.jmedchem.4c02809.
- [22] Hui Peng;Yingfei Wang;Weibo Luo. Multifaceted role of branched-chain amino acid metabolism in cancer.. Oncogene(IF=7.3). 2020. PMID:32978521. DOI: 10.1038/s41388-020-01480-z.
- [23] Lanyue Zhang;Jiangnan Zhao;Chunyu Su;Jianxi Wu;Lai Jiang;Hao Chi;Qin Wang. Organoid models of ovarian cancer: resolving immune mechanisms of metabolic reprogramming and drug resistance.. Frontiers in immunology(IF=5.9). 2025. PMID:40191206. DOI: 10.3389/fimmu.2025.1573686.
- [24] Mingpai Ge;Yuan Xu;Lu Cui;Enyuan Huang;Zhaorui Liu;Kai Yin. Targeting amino acid in tumor therapy.. Frontiers in oncology(IF=3.3). 2025. PMID:40535116. DOI: 10.3389/fonc.2025.1582116.
- [25] Rhuthuparna Malayil;Yogita Chhichholiya;Kanika Vasudeva;Harsh Vikram Singh;Tashvinder Singh;Sandeep Singh;Anjana Munshi. Oncogenic metabolic reprogramming in breast cancer: focus on signaling pathways and mitochondrial genes.. Medical oncology (Northwood, London, England)(IF=3.5). 2023. PMID:37170010. DOI: 10.1007/s12032-023-02037-2.
- [26] Siying Lyu;Nina Gildor;Qing Zhang;Chengheng Liao. Rewiring cancer metabolism: oncogenic signaling pathways and targeted therapeutics.. Science China. Life sciences(IF=9.5). 2025. PMID:40715697. DOI: 10.1007/s11427-025-2979-3.
- [27] Prasenjit Dey;Alec C Kimmelman;Ronald A DePinho. Metabolic Codependencies in the Tumor Microenvironment.. Cancer discovery(IF=33.3). 2021. PMID:33504580. DOI: 10.1158/2159-8290.CD-20-1211.
- [28] Hye-Young Min;Ho-Young Lee. Oncogene-Driven Metabolic Alterations in Cancer.. Biomolecules & therapeutics(IF=3.2). 2018. PMID:29212306. DOI: 10.4062/biomolther.2017.211.
- [29] Marco Tomasetti;Monica Amati;Lory Santarelli;Jiri Neuzil. MicroRNA in Metabolic Re-Programming and Their Role in Tumorigenesis.. International journal of molecular sciences(IF=4.9). 2016. PMID:27213336. DOI: .
- [30] Kenta Masui;Mio Harachi;Webster K Cavenee;Paul S Mischel;Noriyuki Shibata. mTOR complex 2 is an integrator of cancer metabolism and epigenetics.. Cancer letters(IF=10.1). 2020. PMID:32145344. DOI: 10.1016/j.canlet.2020.03.001.
- [31] P M Herst;C Grasso;Michael V Berridge. Metabolic reprogramming of mitochondrial respiration in metastatic cancer.. Cancer metastasis reviews(IF=8.7). 2018. PMID:30448881. DOI: 10.1007/s10555-018-9769-2.
- [32] Xiaodong Hao;Yidan Ren;Maoxiao Feng;Qin Wang;Yunshan Wang. Metabolic reprogramming due to hypoxia in pancreatic cancer: Implications for tumor formation, immunity, and more.. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie(IF=7.5). 2021. PMID:34120068. DOI: 10.1016/j.biopha.2021.111798.
- [33] Nikita Taneja;Akansha Chauhan;Ritu Kulshreshtha;Sandhya Singh. HIF-1 mediated metabolic reprogramming in cancer: Mechanisms and therapeutic implications.. Life sciences(IF=5.1). 2024. PMID:38971364. DOI: 10.1016/j.lfs.2024.122890.
- [34] Jinfen Wei;Meiling Hu;Hongli Du. Improving Cancer Immunotherapy: Exploring and Targeting Metabolism in Hypoxia Microenvironment.. Frontiers in immunology(IF=5.9). 2022. PMID:35281061. DOI: 10.3389/fimmu.2022.845923.
- [35] Qianwen Li;Fansu Meng;Lina Yang;Alicia Marcelina Díaz García;Marina Parkhats;Jaiwoo Lee;Kosuke Kusamori;Makiya Nishikawa;Honghui Gu;Qi Li;Gareth R Williams;Zhenjiang Yang;Yu Cai. Metabolic Reprogramming of Tumors: Induced Immunosuppression and Therapeutic Prospects of Nanoscale Drug Delivery Systems.. Molecular pharmaceutics(IF=4.5). 2025. PMID:40742015. DOI: 10.1021/acs.molpharmaceut.5c00342.
- [36] Diksha Devi;Kanchan Sharma;Tabish Khan;Hema Rani;Rekha Rana;Heena Sharma;Parjinder Kaur;Lokesh Prajapati;Adil Rizwan;Moses Allieu;Sampita Pal;Vishal Kumar;Sunny Kumar. Hypoxia-driven metabolic and molecular reprogramming: From tumor microenvironment to therapeutic interventions.. Pathology, research and practice(IF=3.2). 2025. PMID:41106222. DOI: 10.1016/j.prp.2025.156271.
- [37] Patricia Altea-Manzano;Amanda Decker-Farrell;Tobias Janowitz;Ayelet Erez. Metabolic interplays between the tumour and the host shape the tumour macroenvironment.. Nature reviews. Cancer(IF=66.8). 2025. PMID:39833533. DOI: 10.1038/s41568-024-00786-4.
- [38] Cen Wu;Tianhua Xu;Heyang Zhang;Yile Hu;Jinghua Jiao;Kun Qiu;Jia Gu;Wenya Li;Lei Sun. Hypoxia and immunometabolism in the tumor microenvironment: insights into mechanisms and therapeutic potential.. Cancer letters(IF=10.1). 2025. PMID:40653189. DOI: 10.1016/j.canlet.2025.217913.
- [39] Yuqiang Liu;Yu Zhao;Huisheng Song;Yunting Li;Zihao Liu;Zhiming Ye;Jianzhu Zhao;Yuzheng Wu;Jun Tang;Maojin Yao. Metabolic reprogramming in tumor immune microenvironment: Impact on immune cell function and therapeutic implications.. Cancer letters(IF=10.1). 2024. PMID:38906524. DOI: 10.1016/j.canlet.2024.217076.
- [40] Yi Chen;Menglin Bai;Ming Liu;Zhenshan Zhang;Chenxue Jiang;Kejin Li;Yun Chen;Yaping Xu;Leilei Wu. Metabolic Reprogramming in Lung Cancer: Hallmarks, Mechanisms, and Targeted Strategies to Overcome Immune Resistance.. Cancer medicine(IF=3.1). 2025. PMID:41163383. DOI: 10.1002/cam4.71317.
- [41] Carla Navarro;Ángel Ortega;Raquel Santeliz;Bermary Garrido;Maricarmen Chacín;Néstor Galban;Ivana Vera;Juan Bautista De Sanctis;Valmore Bermúdez. Metabolic Reprogramming in Cancer Cells: Emerging Molecular Mechanisms and Novel Therapeutic Approaches.. Pharmaceutics(IF=5.5). 2022. PMID:35745875. DOI: 10.3390/pharmaceutics14061303.
- [42] Xiang Li;Xueqin Cao;Yuanfu Wang;Yonghao Deng;Elizabeth Rosalind Thomas;Wudian Xiao;Wenjun Wang;Linxia Xu;Qidan He. Glucose metabolism reprogramming in gastric cancer: Implications for tumor.. International immunopharmacology(IF=4.7). 2025. PMID:41101225. DOI: 10.1016/j.intimp.2025.115630.
- [43] Xiaosu Zhou;Ruibing Deng;Zunde Liao;Xiaoyu Huang;Junting Huang;Hanlou Yang;Kam W Leong;Yiling Zhong. Integrating Metabolic Modulation and Nanomedicine for Cancer Immunotherapy.. Advanced science (Weinheim, Baden-Wurttemberg, Germany)(IF=14.1). 2025. PMID:40940612. DOI: 10.1002/advs.202510004.
- [44] Ching-Feng Chiu;Jonathan Jaime G Guerrero;Ric Ryan H Regalado;Jiayan Zhou;Kin Israel Notarte;Yu-Wei Lu;Paolo C Encarnacion;Cidne Danielle D Carles;Edrian M Octavo;Dan Christopher I Limbaroc;Charupong Saengboonmee;Shih-Yi Huang. Insights into Metabolic Reprogramming in Tumor Evolution and Therapy.. Cancers(IF=4.4). 2024. PMID:39456607. DOI: 10.3390/cancers16203513.
- [45] Peijie Zheng;Zihao Lin;Yuemin Ding;Shiwei Duan. Targeting the dynamics of cancer metabolism in the era of precision oncology.. Metabolism: clinical and experimental(IF=11.9). 2023. PMID:37286129. DOI: 10.1016/j.metabol.2023.155615.
- [46] Yi Xiao;Tian-Jian Yu;Ying Xu;Rui Ding;Yi-Ping Wang;Yi-Zhou Jiang;Zhi-Ming Shao. Emerging therapies in cancer metabolism.. Cell metabolism(IF=30.9). 2023. PMID:37557070. DOI: 10.1016/j.cmet.2023.07.006.
- [47] Jialuo Zou;Cunjun Mai;Zhiqin Lin;Jian Zhou;Guie Lai. Targeting metabolism of breast cancer and its implications in T cell immunotherapy.. Frontiers in immunology(IF=5.9). 2024. PMID:38680483. DOI: 10.3389/fimmu.2024.1381970.
- [48] Tingting Gao;Liuxin Yang;Yali Zhang;Ousman Bajinka;Xingxing Yuan. Cancer metabolic reprogramming and precision medicine-current perspective.. Frontiers in pharmacology(IF=4.8). 2024. PMID:39484162. DOI: 10.3389/fphar.2024.1450441.
- [49] Yu-Ling Bin;Hong-Sai Hu;Feng Tian;Zhen-Hua Wen;Mei-Feng Yang;Ben-Hua Wu;Li-Sheng Wang;Jun Yao;De-Feng Li. Metabolic Reprogramming in Gastric Cancer: Trojan Horse Effect.. Frontiers in oncology(IF=3.3). 2021. PMID:35096565. DOI: 10.3389/fonc.2021.745209.
- [50] Brandon Faubert;Ashley Solmonson;Ralph J DeBerardinis. Metabolic reprogramming and cancer progression.. Science (New York, N.Y.)(IF=45.8). 2020. PMID:32273439. DOI: 10.1126/science.aaw5473.
- [51] Niccolo' Roda;Valentina Gambino;Marco Giorgio. Metabolic Constrains Rule Metastasis Progression.. Cells(IF=5.2). 2020. PMID:32932943. DOI: 10.3390/cells9092081.
- [52] Mohamed El-Tanani;Syed Arman Rabbani;Yahia El-Tanani;Ismail I Matalka. Metabolic vulnerabilities in cancer: A new therapeutic strategy.. Critical reviews in oncology/hematology(IF=5.6). 2024. PMID:38977145. DOI: 10.1016/j.critrevonc.2024.104438.
- [53] Wiktoria Andryszkiewicz;Julia Gąsiorowska;Maja Kübler;Karolina Kublińska;Agata Pałkiewicz;Adam Wiatkowski;Urszula Szwedowicz;Anna Choromańska. Glucose Metabolism and Tumor Microenvironment: Mechanistic Insights and Therapeutic Implications.. International journal of molecular sciences(IF=4.9). 2025. PMID:40076506. DOI: 10.3390/ijms26051879.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
metabolic reprogramming · cancer progression · Warburg effect · tumor microenvironment · therapeutic strategies
© 2025 MaltSci
