GET THE APP

Metabolic Interactions in the Tumor Microenvironment
..

Journal of Immunobiology

ISSN: 2476-1966

Open Access

Mini Review - (2023) Volume 8, Issue 3

Metabolic Interactions in the Tumor Microenvironment

Ehsan Dean*
*Correspondence: Ehsan Dean, Department of Computational Bioscience, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK, Email:
Department of Computational Bioscience, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK

Received: 17-Aug-2023, Manuscript No. jib-23-116368; Editor assigned: 19-Aug-2023, Pre QC No. P-116368; Reviewed: 02-Sep-2023, QC No. Q-116368; Revised: 07-Sep-2023, Manuscript No. R-116368; Published: 14-Sep-2023 , DOI: 10.37421/2476-1966.2023.8.203
Citation: Dean, Ehsan. “Metabolic Interactions in the Tumor Microenvironment.” J Immuno Biol 8 (2023): 203.
Copyright: © 2023 Dean E. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

The Tumor Microenvironment (TME) is a complex and dynamic ecosystem in which cancer cells interact with various stromal cells, immune cells and extracellular matrix components. One crucial aspect of these interactions is metabolic reprogramming. This article explores the intricate metabolic interactions that occur within the TME, shedding light on how these interactions drive tumor progression, immune evasion and potential therapeutic targets. Understanding the metabolic intricacies of the TME is crucial for developing novel cancer treatments and improving patient outcomes.

Keywords

Tumor microenvironment • Metabolism • Metabolic reprogramming • Cancer progression • Immune evasion • Therapeutic targets • Cancer treatment

Introduction

Cancer is a multifaceted disease, characterized not only by uncontrolled cell growth but also by its ability to manipulate the surrounding environment to support its survival and growth. This surrounding environment, known as the Tumor Microenvironment (TME), is a complex ecosystem comprising various cell types, extracellular matrix components and signaling molecules. One of the critical aspects of TME is the metabolic interactions that take place within it. Metabolic reprogramming in both cancer cells and stromal cells profoundly influences tumor progression, immune evasion and response to therapy.

Cancer cells display a distinct metabolic profile compared to normal cells. They often rely on glycolysis, even in the presence of oxygen, a phenomenon known as the Warburg effect. This glycolytic switch provides cancer cells with rapid ATP production and necessary intermediates for biomass synthesis, enabling their uncontrolled growth. In addition to glycolysis, cancer cells also upregulate other metabolic pathways such as the pentose phosphate pathway and lipid synthesis to meet their energy and biosynthetic demands. Metabolic interactions in the tumor microenvironment are a critical determinant of cancer progression, immune evasion and response to therapy. The intricate metabolic adaptations of cancer cells and stromal cells within the TME create a challenging environment for therapeutic intervention. Nevertheless, ongoing research into the metabolic dependencies of cancer cells and the dynamic interactions within the TME holds promise for the development of innovative treatments that can improve patient outcomes in the fight against cancer [1].

Literature Review

In addition to cancer cells, the TME hosts various stromal cells, including Cancer-Associated Fibroblasts (CAFs), endothelial cells and immune cells. These stromal cells play essential roles in tumor progression and exhibit their metabolic adaptations. Cancer-Associated Fibroblasts are a prominent component of the TME. They often undergo metabolic reprogramming, shifting towards aerobic glycolysis, which supports the synthesis of extracellular matrix components and the release of growth factors that nourish cancer cells. CAFs can also promote angiogenesis by secreting factors like Vascular Endothelial Growth Factor (VEGF), thereby facilitating tumor growth [2].

Immune cells within the TME must adapt to the nutrient-deprived and immunosuppressive environment created by cancer cells. Immune cells like T cells and macrophages may undergo metabolic reprogramming to survive and function in the TME. Some T cells switch to glycolysis to sustain their effector functions, while Tumor-Associated Macrophages (TAMs) can display a spectrum of metabolic profiles, ranging from glycolysis to oxidative phosphorylation, depending on their phenotype. Understanding the metabolic interactions in the TME has significant therapeutic implications. Targeting these metabolic adaptations could provide new avenues for cancer treatment [3].

Small molecules that target specific metabolic enzymes or pathways in cancer cells or stromal cells are under development. For example, inhibitors of glycolytic enzymes or glutamine metabolism have shown promise in preclinical studies. Combining immunotherapies with metabolic interventions may enhance their efficacy. Strategies such as checkpoint inhibitors and adoptive T-cell therapy can be potentiated by modulating the TME's metabolic landscape. Strategies aimed at disrupting the nutrient competition between cancer cells and immune cells are being explored. This involves enhancing nutrient availability to immune cells while limiting cancer cells' access to key metabolites [4].

Developing personalized approaches to target the specific metabolic vulnerabilities of individual tumours is a major goal. Biomarker discovery and patient stratification based on metabolic profiles will be essential for tailoring treatment strategies. The complexity of the TME necessitates a multifaceted approach to cancer treatment. Combinations of metabolic inhibitors, immunotherapies and conventional treatments may be required to effectively target both cancer cells and the surrounding stromal cells. Just as cancer cells can develop resistance to traditional therapies, they can also adapt metabolically to overcome metabolic inhibitors. Understanding the mechanisms underlying metabolic resistance is crucial for the long-term success of metabolic-targeted therapies [5].

Discussion

Developing non-invasive imaging techniques that can assess metabolic changes within tumors in real-time will be invaluable for monitoring treatment response and adapting therapy as needed. Moving promising preclinical findings into clinical trials and eventually into routine clinical practice is a significant challenge. Clinical trials that focus on metabolic interventions and combinations with established therapies are essential for evaluating their safety and efficacy in humans. Targeting metabolic pathways can have systemic effects on normal tissues and metabolic homeostasis. Minimizing off-target effects and understanding potential toxicities are critical considerations in the development of metabolic therapies. Understanding the potential long-term consequences of metabolic interventions is crucial. These therapies may have lasting effects on patients' metabolic health and their impact on survivorship and quality of life must be carefully studied [6].

Conclusion

The intricate metabolic interactions within the tumor microenvironment represent a promising frontier in cancer research and therapy. The dynamic interplay between cancer cells and stromal cells, coupled with their metabolic adaptations, offers numerous opportunities for therapeutic intervention. While challenges remain, ongoing research in this field holds the potential to revolutionize cancer treatment and improve outcomes for patients. As our understanding of these metabolic intricacies deepens, we can expect innovative strategies to emerge that target cancer at its metabolic core, bringing us closer to the goal of effective and personalized cancer care. However, the metabolic reprogramming of cancer cells extends beyond energy production and biomass synthesis. It plays a pivotal role in immune evasion and tumor progression. Furthermore, cancer cells may compete with immune cells for nutrients, depleting critical metabolites required for immune cell function and proliferation.

Acknowledgement

We thank the anonymous reviewers for their constructive criticisms of the manuscript.

Conflict of Interest

The author declares there is no conflict of interest associated with this manuscript.

References

  1. Hanahan, Douglas and Robert A. Weinberg. "Hallmarks of cancer: The next generation." Cell 144 (2011): 646-674.
  2. Google Scholar, Crossref, Indexed at

  3. Zhang, Zhuzhen, Zhenzhen Zi, Eunice E. Lee and Jiawei Zhao, et al. "Differential glucose requirement in skin homeostasis and injury identifies a therapeutic target for psoriasis." Nat Med 24 (2018): 617-627.
  4. Google Scholar, Crossref, Indexed at

  5. Chen, Chao, Guixue Hou, Chunwei Zeng and Yan Ren, et al. "Metabolomic profiling reveals amino acid and carnitine alterations as metabolic signatures in psoriasis." Theranostics 11 (2021): 754.
  6. Google Scholar, Crossref, Indexed at

  7. Clark, Susan J. and Peter L. Molloy. "Early insights into cancer epigenetics: Gene promoter hypermethylation emerges as a potential biomarker for cancer detection." Cancer Res 82 (2022): 1461-1463.
  8. Google Scholar, Crossref, Indexed at

  9. Zhou, Qiang, Ulrich Mrowietz and Martin Rostami-Yazdi. "Oxidative stress in the pathogenesis of psoriasis." Free Radic Biol Med 47 (2009): 891-905.
  10. Google Scholar, Crossref, Indexed at

  11. Azarova, Anna M., Yi Lisa Lyu, Chao-Po Lin and Yuan-Chin Tsai, et al. "Roles of DNA topoisomerase II isozymes in chemotherapy and secondary malignancies." Proc Natl Acad Sci 104 (2007): 11014-11019.
  12. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 34

Journal of Immunobiology received 34 citations as per Google Scholar report

Journal of Immunobiology peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward