Journal of Molecular Oncology Research

All submissions of the EM system will be redirected to Online Manuscript Submission System. Authors are requested to submit articles directly to Online Manuscript Submission System of respective journal.
Reach Us +1 (202) 780-3397

Perspective - Journal of Molecular Oncology Research (2025) Volume 9, Issue 1

Immunometabolism in the tumor microenvironment and neoantigen-based cancer vaccines: A new era in cancer therapy

Jackie Wong *

Institute of Molecular Oncology, University of Milan, Italy

*Corresponding Author:
Jackie Wong
Institute of Molecular Oncology, University of Milan, Italy
E-mail: jackie@wong.it

Received: 01-Jan-2025, Manuscript No. AAMOR -25-161672; Editor assigned: 02-Jan-2025, PreQC No. AAMOR -25-161672(PQ); Reviewed: 18-Jan-2025, QC No. AAMOR -25-161672; Revised: 22-Jan-2025, Manuscript No. AAMOR -25-161672(R); Published: 29-Jan-2025, DOI: 10.35841/ aamor-9.1.274

Citation: : Wong J. Immunometabolism in the tumor microenvironment and neoantigen-based cancer vaccines: A new era in cancer therapy. J Mol Oncol Res. 2025;9(1):274.

Visit for more related articles at Journal of Molecular Oncology Research

Introduction

Cancer remains one of the most complex diseases, characterized by its ability to evade immune responses and adapt to various metabolic conditions. The tumor microenvironment (TME) plays a crucial role in modulating immune cell function and metabolic pathways, which in turn influence tumor progression. Immunometabolism, the intersection of immune cell function and metabolic processes, has emerged as a vital area of cancer research. Understanding how metabolic reprogramming within the TME affects immune responses can lead to more effective therapeutic strategies [1].

Simultaneously, advancements in cancer immunotherapy have led to the development of personalized treatment approaches such as neoantigen-based cancer vaccines. These vaccines harness the body's immune system to recognize and destroy tumor-specific antigens, offering a promising strategy for long-term cancer control. This article explores the relationship between immunometabolism in the TME and the potential of neoantigen-based vaccines to revolutionize cancer treatment [2].

The TME consists of tumor cells, immune cells, fibroblasts, and endothelial cells, all of which interact dynamically to influence tumor growth. Within this environment, metabolic alterations occur that support tumor survival while suppressing immune responses. For instance, cancer cells shift towards glycolysis, even in oxygen-rich conditions, a phenomenon known as the Warburg effect. This metabolic shift leads to the depletion of nutrients, such as glucose and amino acids, which are essential for immune cell activation [3].

Furthermore, the accumulation of immunosuppressive metabolites, such as lactic acid and kynurenine, impairs T-cell function and promotes the expansion of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). These immunosuppressive elements create a hostile environment that limits the effectiveness of anti-tumor immunity, highlighting the need for strategies that can counteract metabolic suppression [4].

Recent research has focused on modulating immunometabolism to enhance anti-cancer immune responses. Targeting metabolic pathways, such as glycolysis inhibitors and amino acid deprivation strategies, can help reprogram immune cells within the TME. Additionally, interventions like immune checkpoint inhibitors (ICIs) have been shown to improve T-cell metabolism, restoring their ability to attack cancer cells [5].

Another promising approach involves using metabolic adjuvants in combination with immunotherapies. For example, drugs that inhibit lactate production or enhance oxidative phosphorylation in T cells can create a more favorable metabolic landscape, increasing the effectiveness of cancer treatments. These metabolic interventions pave the way for novel immunotherapeutic strategies, including cancer vaccines [6].

Neoantigen-based cancer vaccines represent a groundbreaking approach in precision medicine. Unlike traditional cancer vaccines, which target shared tumor-associated antigens, neoantigens are unique to individual tumors and arise from somatic mutations. This makes them highly immunogenic and less likely to trigger tolerance mechanisms that hinder immune responses [7].

Neoantigen vaccines work by presenting tumor-specific antigens to the immune system, stimulating cytotoxic T lymphocytes (CTLs) to recognize and eliminate cancer cells. Advances in next-generation sequencing (NGS) and bioinformatics have enabled the rapid identification of neoantigens, allowing for the development of personalized vaccines tailored to each patient's tumor profile [8].

Despite the promising potential of neoantigen-based vaccines, several challenges remain. One major hurdle is the tumor's ability to evolve and develop immune evasion mechanisms. Tumors can downregulate antigen presentation or modify their microenvironment to suppress immune responses, reducing vaccine efficacy. Overcoming these challenges requires innovative approaches, such as combination therapies that integrate neoantigen vaccines with ICIs, metabolic modulators, or adoptive T-cell therapies [9].

Additionally, optimizing vaccine delivery platforms, such as lipid nanoparticles or dendritic cell-based approaches, can enhance antigen presentation and improve immune responses. Ongoing clinical trials are investigating various strategies to maximize the effectiveness of neoantigen vaccines, offering hope for more durable and personalized cancer treatments in the future [10].

Conclusion

The interplay between immunometabolism in the TME and the development of neoantigen-based cancer vaccines represents a promising frontier in oncology. By targeting metabolic pathways that suppress immune responses, researchers can enhance the efficacy of cancer immunotherapies. Neoantigen vaccines, with their personalized approach, offer a new level of precision in cancer treatment, potentially leading to long-lasting immune protection. As scientific advancements continue to unravel the complexities of the TME and neoantigen presentation, the integration of immunometabolic interventions with personalized cancer vaccines holds the potential to redefine cancer treatment. This innovative approach not only improves patient outcomes but also paves the way for a more effective and targeted battle against cancer.

References

  1. Leeuwendaal NK, Stanton C, O’toole PW, et al. Fermented foods, health and the gut microbiome. Nutr. 2022;14(7):1527.
  2. Indexed at, Google Scholar, Cross Ref

  3. Valentino V, Magliulo R, Farsi D, et al. Fermented foods, their microbiome and its potential in boosting human health. Micro Biotech. 2024;17(2):e14428.
  4. Indexed at, Google Scholar, Cross Ref

  5. Rul F, Béra-Maillet C, Champomier-Vergès MC, et al. Underlying evidence for the health benefits of fermented foods in humans. Food 2022;13(9):4804-24.
  6. Indexed at, Google Scholar, Cross Ref

  7. Obafemi YD, Oranusi SU, Ajanaku KO, et al. African fermented foods: overview, emerging benefits, and novel approaches to microbiome profiling. npj Sci Food. 2022 18;6(1):15.
  8. Indexed at, Google Scholar, Cross Ref

  9. Walsh AM, Leech J, Huttenhower C, et al. Integrated molecular approaches for fermented food microbiome research. Microbio Rev. 2023;47(2):fuad001.
  10. Indexed at, Google Scholar, Cross Ref

  11. Shah AM, Tarfeen N, Mohamed H, et al. Fermented foods: Their health-promoting components and potential effects on gut microbiota. Fermen. 2023;9(2):118.
  12. Indexed at, Google Scholar, Cross Ref

  13. Xing Y, Huang M, Olovo CV, et al. Traditional fermented foods: Challenges, sources, and health benefits of fatty acids. Fermen. 2023;9(2):110.
  14. Indexed at, Google Scholar, Cross Ref

  15. Rastogi YR, Thakur R, Thakur P, et al. Food fermentation–Significance to public health and sustainability challenges of modern diet and food systems. Intern J Food Microb. 16;371:109666.
  16. Indexed at, Google Scholar, Cross Ref

  17. Mukherjee A, Breselge S, Dimidi E, Marco ML, Cotter PD. Fermented foods and gastrointestinal health: underlying mechanisms. Nat Rev Gastro 2024;21(4):248-66.
  18. Indexed at, Google Scholar, Cross Ref

  19. Kunyeit L, Rao RP, Anu-Appaiah KA. Yeasts originating from fermented foods, their potential as probiotics and therapeutic implication for human health and disease. Crit Rev Food Sci Nnu. 2024 25;64(19):6660-71.
  20. Indexed at, Google Scholar, Cross Ref

Get the App