Journal of Cancer Immunology & Therapy

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.

Rapid Communication - Journal of Cancer Immunology & Therapy (2024) Volume 7, Issue 5

Neoantigen-based vaccines: A new era in cancer immunotherapy.

Youra Gyga *

Department of Medicine, University of British Columbia, Canada

*Corresponding Author:
Youra Gyga
Department of Medicine, University of British Columbia, Canada
E-mail: gygi.y@dal.ca

Received: 27-Sep -2024, Manuscript No. AAJCIT-24-150273; Editor assigned: 28-Sep-2024, PreQC No. AAJCIT-24-150273 (PQ); Reviewed:14-Oct-2024, QC No. AAJCIT-24-150273; Revised:19-Oct-2024, Manuscript No. AAJCIT-24-150273 (R); Published:28-Oct-2024, DOI:10.35841/aara-7.5.233

Citation: Gyga Y., Neoantigen-based vaccines: A new era in cancer immunotherapy. J Cancer Immunol Ther. 2024;7(5):233

Visit for more related articles at Journal of Cancer Immunology & Therapy

Introduction

Cancer immunotherapy has revolutionized the treatment landscape for various malignancies, offering new hope to patients. Among the innovative strategies being explored, neoantigen-based vaccines stand out for their potential to harness the power of the immune system against tumors. Neoantigens are unique peptides formed due to mutations in tumor cells, distinguishing them from normal cells. This article discusses the principles of neoantigen-based vaccines, their development, clinical applications, and the challenges ahead in this promising field [1].

Neoantigens are peptide fragments derived from mutated proteins that are not present in normal tissues, making them attractive targets for immune recognition. They arise from nonsynonymous mutations, which lead to changes in amino acid sequences and generate novel epitopes that can be presented by major histocompatibility complex (MHC) molecules to T cells. This specificity allows the immune system to identify and attack tumor cells while sparing healthy tissues [2].

The immune system’s ability to recognize neoantigens relies on the activation of cytotoxic T lymphocytes (CTLs), which can effectively kill tumor cells. Research has demonstrated that a higher mutation burden in tumors correlates with improved responses to immunotherapy, underscoring the importance of neoantigens in driving anti-tumor immunity [3].

The process of developing neoantigen-based vaccines involves several key steps: The first step is to sequence the tumor DNA to identify mutations specific to the tumor. This genomic profiling is typically performed using next-generation sequencing (NGS) technologies, which allow for the detection of somatic mutations at a high resolution [4].

Some neoantigens may be less immunogenic or may induce tolerance rather than an active immune response. Understanding the factors influencing immunogenicity will be essential for optimizing vaccine design. The personalized nature of neoantigen vaccines presents challenges in manufacturing and scalability. Developing efficient and standardized processes will be crucial for broader clinical application. Once mutations are identified, bioinformatics tools predict which of these mutations will generate neoantigens that can bind to MHC molecules. Algorithms such as NetMHC and MHCflurry are commonly used to predict peptide binding affinities [5].

The selected neoantigens are synthesized as peptides and formulated into a vaccine. Various delivery platforms, including lipid nanoparticles, dendritic cell-based vaccines, and viral vectors, are being explored to enhance immune responses [6].

Neoantigen vaccines undergo rigorous clinical testing to evaluate their safety and efficacy. Early-phase clinical trials often focus on assessing immune responses and tumor-specific T cell activation before examining clinical outcomes such as tumor shrinkage and overall survival [7].

Several clinical trials have demonstrated the potential of neoantigen-based vaccines in treating various cancers, including melanoma, lung cancer, and glioblastoma. Notable studies include: In a groundbreaking trial by Sahin et al. (2017), patients with melanoma received personalized vaccines targeting neoantigens identified from their tumors. The results showed a robust T cell response and significant tumor regression in some patients [8].

Accurately identifying immunogenic neoantigens is critical for the success of these vaccines. Ongoing advancements in genomics and bioinformatics will improve neoantigen prediction algorithms and enhance our understanding of tumor-specific mutations. Combining neoantigen vaccines with immune checkpoint inhibitors has shown promise in enhancing anti-tumor immunity. For instance, a trial investigating the combination of a neoantigen vaccine with anti-PD-1 therapy demonstrated improved responses compared to either treatment alone [9].

While early clinical trials have shown promise, long-term outcomes and durability of responses need to be assessed. Ongoing studies will provide insights into the sustainability of immune responses induced by neoantigen vaccines. Neoantigen vaccines are being explored across various malignancies, including colorectal cancer, bladder cancer, and pancreatic cancer, with encouraging results. The ability to personalize vaccines based on individual tumor profiles is a significant advantage [10].

Conclusion

Neoantigen-based vaccines represent a new frontier in cancer immunotherapy, offering the potential for personalized treatment strategies that harness the immune system’s ability to recognize and eliminate tumor cells. With continued advancements in genomic profiling, bioinformatics, and vaccine development, neoantigen vaccines could significantly impact cancer treatment and improve patient outcomes. As the field progresses, addressing the existing challenges will be crucial in realizing the full potential of this innovative approach to cancer immunotherapy.

References

  1. Shen H, Yang ES, Conry M, Fiveash J, Contreras C, Bonner JA, Shi LZ. Predictive biomarkers for immune checkpoint blockade and opportunities for combination therapies. Gen Dis. 2019;6(3):232-46.
  2. Indexed at, Google Scholar

  3. Grizzi G, Caccese M, Gkountakos A, Carbognin L, Tortora G, Bria E, Pilotto S. Putative predictors of efficacy for immune checkpoint inhibitors in non-small-cell lung cancer: facing the complexity of the immune system. Expert Rev Mol Diagn. 2017;17(12):1055-69.
  4. Google Scholar

  5. Kreiter S, Vormehr M, Van de Roemer N, Diken M, Löwer M, Diekmann J, Boegel S, Schrörs B, Vascotto F, Castle JC, Tadmor AD. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nat. 2015;520(7549):692-6.
  6. Google Scholar

  7. Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nat. 2011;480(7378):480-9.
  8. Indexed at, Google Scholar

  9. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS, Miller ML. Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Sci. 2015;348(6230):124-8.
  10. Google Scholar

  11. Sahin U, Derhovanessian E, Miller M, Kloke BP, Simon P, Löwer M, Bukur V, Tadmor AD, Luxemburger U, Schrörs B, Omokoko T. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nat. 2017;547(7662):222-6.
  12. Google Scholar

  13. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Sci. 2015;348(6230):69-74.
  14. Indexed at, Google Scholar

  15. Nebhan CA, Johnson DB. Predictive biomarkers of response to immune checkpoint inhibitors in melanoma. Expert Rev Anticancer Ther. 2020;20(2):137-45.
  16. Google Scholar

  17. Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, Sucker A, Hillen U, Geukes Foppen MH, Goldinger SM, Utikal J. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Sci. 2015;350(6257):207-11.
  18. Indexed at, Google Scholar

  19. Nguyen HM, Guz-Montgomery K, Lowe DB, Saha D. Pathogenetic features and current management of glioblastoma. Cancers. 2021;13(4):856.
  20. Indexed at, Google Scholar

Get the App