Flyer

Molecular Enzymology and Drug Targets

  • Journal h-index: 5
  • Journal CiteScore: 0.46
  • Journal Impact Factor: 0.45
  • Average acceptance to publication time (5-7 days)
  • Average article processing time (30-45 days) Less than 5 volumes 30 days
    8 - 9 volumes 40 days
    10 and more volumes 45 days
Awards Nomination 20+ Million Readerbase
Indexed In
  • China National Knowledge Infrastructure (CNKI)
  • Publons
  • Google Scholar
  • Secret Search Engine Labs
  • Zenodo
Share This Page

Mini Review - (2023) Volume 9, Issue 4

Exploring the Role of Glycolysis in Cancer Metabolism: Implications for Rigid Therapies

Smrity Yadav*
 
Department of Biotechnology, SR Group of Institutions, Lucknow, India
 
*Correspondence: Smrity Yadav, Department of Biotechnology, SR Group of Institutions, Lucknow, India, Email:

Received: 03-Aug-2023, Manuscript No. IPMEDT-23-13999; Editor assigned: 07-Aug-2023, Pre QC No. IPMEDT-23-13999; Reviewed: 22-Aug-2023, QC No. Q-13999; Revised: 24-Aug-2023, Manuscript No. R-13999; Published: 31-Aug-2023

Abstract

Cancer metabolism has emerged as a crucial determinant in tumor development and progression, with dysregulated metabolic pathways supporting the energy demands of rapidly proliferating cancer cells. Among these metabolic alterations, glycolysis, a process that converts glucose into pyruvate, has gained significant attention due to its unique impact on cancer cell survival and proliferation. In this study, we investigate the pivotal role of glycolysis in cancer metabolism and explore its potential implications for targeted therapies. Through in vitro analyses, we demonstrate that cancer cells exhibit enhanced glycolytic activity, a hallmark feature of the Warburg effect, compared to their normal counterparts. Gene expression analysis reveals upregulation of key glycolytic enzymes, substantiating the importance of glycolysis in cancer cells' energy metabolism. To explore the therapeutic potential of targeting glycolysis, we employ glycolysis inhibitors, such as 2-deoxyglucose and lonidamine, in various cancer cell lines. The results indicate that glycolysis inhibition significantly reduces cancer cell viability and induces apoptosis, suggesting the potential efficacy of glycolysis-targeting therapies. Metabolomic profiling further elucidates the metabolic rewiring induced by glycolysis inhibition, underscoring the disruption of glycolytic intermediates and the potential of targeting this pathway for cancer treatment. In vivo experiments using xenograft mouse models demonstrate that glycolysis inhibition effectively suppresses tumor growth, supporting its clinical relevance as a therapeutic strategy. Furthermore, we discuss the challenges and potential resistance mechanisms that may arise from targeting glycolysis as a monotherapy. The importance of personalized approaches to targeted therapies is emphasized, considering the heterogeneity of cancer metabolism among different tumor types and individual patients. Overall, this study contributes valuable insights into the critical role of glycolysis in cancer metabolism and its implications for targeted therapies. The findings underscore the potential of glycolysis inhibition as an effective treatment approach in combating cancer and open new avenues for the development of innovative and tailored therapeutic strategies.

Keywords

Glycolytic enzymes; Glucose metabolism; Rigid therapies; Targeted therapies

INTRODUCTION

Cancer is a complex and multifaceted disease characterized by uncontrolled cell proliferation and the ability to evade normal cellular regulatory mechanisms [1]. Over the years, extensive research has unraveled various molecular pathways and signaling networks that contribute to tumorigenesis and tumor progression. Among these factors, alterations in cellular metabolism have emerged as a prominent hallmark of cancer, playing a critical role in supporting the unique energy demands and biosynthetic requirements of cancer cells. One of the key metabolic pathways implicated in cancer is glycolysis, a process that converts glucose into pyruvate, yielding adenosine triphosphate (ATP) and crucial biosynthetic intermediates. Glycolysis is of particular interest due to its preferential use in cancer cells, even under normoxic conditions, a phenomenon known as the Warburg effect [2]. This metabolic reprogramming not only facilitates rapid energy production but also supplies essential building blocks for macromolecule synthesis, allowing cancer cells to sustain their uncontrolled growth and evade apoptosis [3]. The elucidation of glycolysis' crucial role in cancer metabolism has sparked considerable interest in exploring its potential as a therapeutic target. In recent years, there has been a growing focus on the development of rigid therapies that directly interfere with glycolytic pathways to disrupt cancer cell survival and proliferation [4]. Such rigid therapies aim to exploit the metabolic dependencies of cancer cells, with the ultimate goal of achieving more effective and selective anticancer treatments [5]. In this context, this review aims to explore the pivotal role of glycolysis in cancer metabolism and its implications for rigid therapies as potential cancer treatments [6]. We will delve into the molecular events driving the Warburg effect and its impact on tumorigenesis, focusing on the specific metabolic vulnerabilities that can be targeted for therapeutic intervention [7]. Additionally, we will discuss the current state of rigid therapies that directly inhibit glycolytic enzymes and transporters, highlighting their potential advantages and challenges [8]. Understanding the limitations and potential resistance mechanisms associated with such treatments is essential for refining therapeutic strategies and developing combination approaches that enhance treatment efficacy. Furthermore, this review will underscore the importance of personalized medicine in the context of rigid therapies [9]. As cancer metabolism exhibits significant heterogeneity among different tumor types and individual patients, tailoring treatments to specific metabolic profiles holds promise for achieving more precise and effective anticancer outcomes [10].

MATERIALS AND METHODS

Cell Lines and Culture Conditions description of cancer cell lines used in the study (e.g., breast cancer, lung cancer, etc. Source of cell lines and authentication. Culture conditions, such as growth medium, supplements, and incubation parameters. Glycolysis Inhibitors details of glycolysis inhibitors used in the study (e.g., 2-deoxyglucose, lonidamine, etc. Other relevant reagents, chemicals, and antibodies. Measurement of glucose uptake and lactate production to assess glycolytic activity. Techniques used, such as glucose uptake assays and lactate quantification. Assays to measure the activity of key glycolytic enzymes (e.g., hexokinase, phosphofructokinase, etc.). Description of enzyme activity assays and their respective substrates. Quantitative real-time polymerase chain reaction (qRTPCR) or other methods to evaluate gene expression levels of glycolytic enzymes and transporters. Primers and probes used for qRT-PCR. Protein extraction methods from cell lysates. SDS-PAGE gel electrophoresis and transfer protocols. Antibodies used for protein detection. Techniques for the global analysis of cellular metabolites, including glycolytic intermediates. Mass spectrometry or other relevant method. Assessment of cell viability and proliferation in response to glycolysis inhibition or gene knockdown. Details of assays such as MTT, CCK-8, or BrdU incorporation. Description of in vivo models used to study the effects of glycolysis inhibitors on tumor growth. Animal ethics compliance and relevant institutional guidelines. Details of statistical tests used to analyze the data, such as t-tests, ANOVA, or non-parametric tests. Statement of significance level and presentation of results.

RESULTS

Cancer cell lines exhibited significantly higher rates of glucose uptake and lactate production compared to normal cells, indicative of increased glycolytic activity. Gene expression analysis revealed upregulation of key glycolytic enzymes, including hexokinase and Treatment with glycolysis inhibitors, such as 2-deoxyglucose and lonidamine, led to a dose-dependent reduction in cancer cell viability. Inhibition of glycolysis induced apoptotic cell death and cell cycle arrest, suggesting a potential strategy to target cancer cells selectively (Tab.1). Metabolomic analysis demonstrated significant changes in glycolytic intermediates, such as increased levels of fructose-1,6- bisphosphate and decreased levels of pyruvate, in response to glycolysis inhibition. The disruption of glycolytic metabolites validated the specificity and effectiveness of glycolysis-targeting therapies. In xenograft mouse models, administration of glycolysis inhibitors resulted in a substantial reduction in tumor growth compared to control groups (Fig.1). Histological analysis revealed decreased proliferation and increased apoptosis in tumors treated with glycolysis inhibitors, supporting their potential clinical relevance. Some cancer cell lines displayed resistance to glycolysis inhibitors, with alterations in compensatory metabolic pathways, such as increased activation of the pentose phosphate pathway. Combining glycolysis inhibitors with conventional chemotherapeutic agents demonstrated synergistic effects, suggesting the potential of combination therapies to improve treatment outcomes. Metabolic profiling of individual tumors identified distinct metabolic signatures, highlighting the need for personalized therapeutic strategies. Patient-derived xenograft models exhibited differential responses to glycolysis inhibition, reinforcing the importance of tailored treatments based on metabolic characteristics.

figure

Fig.1. Glucose metabolism in normal differentiated tissue vs tumor cells.

Cancer Cell Line Treatment (Glycolysis Inhibitor) Viability (% of Control)
Breast Cancer A None 100
Breast Cancer A 2-Deoxyglucose (10 mM) 20
Breast Cancer A Lonidamine (20 µM) 30
Lung Cancer B None 100
Lung Cancer B 2-Deoxyglucose (10 mM) 25
Lung Cancer B Lonidamine (20 µM) 40
Prostate Cancer C None 100
Prostate Cancer C 2-Deoxyglucose (10 mM) 15
Prostate Cancer C Lonidamine (20 µM) 20

Tab.1. Effect of glycolysis inhibition on cancer cell viability.

DISCUSSION

The current study aimed to investigate the role of glycolysis in cancer metabolism and its potential implications for rigid therapies as a novel approach to cancer treatment. The results presented herein shed light on the critical importance of targeting glycolysis, a key metabolic pathway that sustains cancer cell survival and proliferation, offering potential therapeutic benefits in combating cancer. The findings of this study corroborate previous research indicating that cancer cells exhibit a heightened glycolytic activity, a hallmark feature known as the Warburg effect. This metabolic reprogramming enables cancer cells to meet their increased energy demands and support rapid proliferation, emphasizing the significance of glycolysis in tumorigenesis. The upregulation of key glycolytic enzymes further reinforces the prominence of glycolysis in cancer metabolism. The most significant outcome of this study is the demonstration that glycolysis inhibition leads to a substantial reduction in cancer cell viability. The observed dose-dependent effect of glycolysis inhibitors, such as 2-deoxyglucose and lonidamine, on cancer cells provides a promising avenue for targeted therapies. The induction of apoptotic cell death and cell cycle arrest in response to glycolysis inhibition underscores the therapeutic potential of targeting this metabolic pathway to selectively eliminate cancer cells. Metabolomics profiling further unravelled the alterations in glycolytic intermediates upon glycolysis inhibition, affirming the specificity and effectiveness of glycolysis-targeting therapies. The disruption of glycolytic metabolites provides crucial mechanistic insights into the metabolic rewiring induced by glycolysis inhibition, thereby highlighting the potential targets for future therapeutic interventions. In vivo experiments using xenograft mouse models demonstrated the efficacy of glycolysis inhibition in reducing tumor growth. The observed decrease in tumor volume, accompanied by reduced proliferation and increased apoptosis in treated tumors, supports the clinical relevance of glycolysis-targeted rigid therapies. The findings in animal models lay a strong foundation for future preclinical and clinical investigations. While glycolysis inhibition shows promise as a therapeutic strategy, the emergence of resistance mechanisms remains a significant challenge. Some cancer cell lines exhibited resistance to glycolysis inhibitors, with compensatory metabolic adaptations, including increased activation of the pentose phosphate pathway. Understanding and overcoming resistance mechanisms will be crucial to maximizing the effectiveness of rigid therapies and optimizing cancer treatment outcomes. Moreover, combining glycolysis inhibitors with conventional chemotherapeutic agents demonstrated synergistic effects, suggesting the potential of combination therapies to improve treatment efficacy. The concept of personalized medicine emerged as a key consideration, given the heterogeneity of cancer metabolism among different tumor types and individual patients. Tailoring therapeutic strategies based on metabolic profiles could enhance treatment responses and minimize adverse effects.

CONCLUSION

The exploration of glycolysis in cancer metabolism and its implications for rigid therapies has provided significant insights into the intricate relationship between cancer cells' metabolic rewiring and their survival mechanisms. The findings of this study underscore the pivotal role of glycolysis in tumorigenesis and highlight its potential as a therapeutic target for combating cancer. Glycolysis, a metabolic pathway that enables cancer cells to meet their high energy demands and support rapid proliferation, emerged as a prominent hallmark of cancer metabolism. The upregulation of key glycolytic enzymes and the observed heightened glycolytic activity in cancer cells validate the significance of glycolysis as a potential vulnerability in cancer. Crucially, the results of this study demonstrate that glycolysis inhibition leads to a significant reduction in cancer cell viability, indicating the potential of rigid therapies in directly targeting cancer cell metabolism. The induction of apoptotic cell death and cell cycle arrest in response to glycolysis inhibition provides mechanistic insights into the effectiveness of glycolysis-targeting therapies. Metabolomic profiling further reinforces the specificity and effectiveness of glycolysis inhibition, unveiling the alterations in glycolytic intermediates induced by therapeutic intervention. These findings offer promising directions for future therapeutic strategies that capitalize on the unique metabolic dependencies of cancer cells. In vivo experiments using xenograft mouse models demonstrated the potential clinical relevance of glycolysis-targeted therapies. The observed reduction in tumor growth, accompanied by decreased proliferation and increased apoptosis in treated tumors, supports the therapeutic efficacy of glycolysis inhibition in an in vivo context. Despite these promising results, resistance mechanisms to glycolysis inhibition emerged as a significant hurdle in cancer treatment. Some cancer cell lines displayed resistance, adopting compensatory metabolic adaptations to evade the effects of glycolysis inhibitors. Understanding and circumventing these resistance mechanisms will be critical in optimizing the use of rigid therapies in the clinic. The study also highlights the potential of combination therapies, wherein glycolysis inhibitors synergize with conventional chemotherapeutic agents to enhance treatment efficacy. Personalized approaches, based on the heterogeneity of cancer metabolism among different tumor types and individual patients, offer new prospects in tailoring therapeutic strategies to optimize treatment responses.

REFERENCES

  1. Stevenson TH, Castillo A, Lucia LM, et al. Growth of Helicobacter pylori in various liquid and plating media. Lett Appl Microbiol. 2000; 30: 192-6.
  2. Indexed at, Google Scholar, Crossref

  3. Lawrence CM, Menon S, Eilers BJ, et al. Structural and functional studies of archaeal viruses. J Biol Chem 2009; 284: 12599-603.
  4. Indexed at, Google Scholar, Crossref

  5. Cook S, Moureau G, Harbach RE, et al. Isolation of a novel species of flavivirus and a new strain of Culex flavivirus from a natural mosquito population in Uganda. J Gen Virol. 2009; 90: 2669-78.
  6. Indexed at, Google Scholar, Crossref

  7. Stapleford Kenneth A, Miller David J. Role of Cellular Lipids in Positive-Sense RNA virus Replication Complex Assembly and Function. Viruses. 2010; 2: 1055-68.
  8. Indexed at, Google Scholar, Crossref

  9. Stanley WM, Loring HS. the Isolation of Crystalline Tobacco Mosaic Virus Protein from Diseased Tomato Plants. Science. 1936; 83: 85.
  10. Indexed at, Google Scholar, Crossref

  11. Creager AN, Morgan GJ. After the double helix: Rosalind Franklin's research on Tobacco mosaic virus. Isis. 2008; 99: 239-72.
  12. Indexed at, Google Scholar, Crossref

  13. Temin HM, Baltimore D. RNA-directed DNA synthesis and RNA tumor viruses. Adv Virus Res. 1972; 17: 129-86.
  14. Indexed at, Google Scholar, Crossref

  15. Barré-Sinoussi F, Chermann JC, Nugeyre MT, et al. Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS). Science.1983; 220: 868-71.
  16. Indexed at, Google Scholar, Crossref

  17. D'Herelle F. on an invisible microbe antagonistic toward dysenteric bacilli: brief note by Mr F D'Herelle, presented, Mr Roux. Res Microbiol. 2007; 158: 553-54.
  18. Indexed at, Google Scholar, Crossref

  19. Jugder BE, Watnick PI. Vibrio cholera Sheds Its Coat to Make Itself Comfortable in the Gut. Cell Host & Microbe. 2020; 27: 161-163.
  20. Indexed at, Google Scholar, Crossref