GET THE APP

Neoantigens: The Cutting-Edge Precision Cancer Vaccine
..

Journal of Molecular Histology & Medical Physiology

ISSN: 2684-494X

Open Access

Mini Review - (2024) Volume 9, Issue 1

Neoantigens: The Cutting-Edge Precision Cancer Vaccine

Huang Hong*
*Correspondence: Huang Hong, Department of Hematology, Beckman Research Institute, Duarte, USA, Email:
Department of Hematology, Beckman Research Institute, Duarte, USA

Received: 13-Dec-2023, Manuscript No. jmhmp-23-117509; Editor assigned: 15-Dec-2023, Pre QC No. P-117509; Reviewed: 27-Dec-2023, QC No. Q-117509; Revised: 01-Jan-2024, Manuscript No. R-117509; Published: 08-Jan-2024 , DOI: 10.37421/2684-494X.2024.9.106
Citation: Hong, Huang. “Neoantigens: The Cutting-Edge Precision Cancer Vaccine.” J Mol Hist Med Phys 9 (2024): 106.
Copyright: © 2024 Hong H. 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

Cancer has long been a formidable adversary in the realm of medical science. It's a disease that can originate from various tissues and affect virtually any part of the body, making it a complex and elusive target. While cancer treatment has evolved significantly over the years, the development of effective and precise therapeutic strategies has remained a major challenge. However, recent advancements in the field of immunotherapy have given rise to a promising approach known as neoantigen-based cancer vaccines. This cutting-edge technology holds the potential to revolutionize cancer treatment, offering personalized therapies that target the specific mutations in a patient's cancer cells. In this article, we will explore the concept of neoantigens, their significance in the realm of cancer immunotherapy, and the development of precision cancer vaccines. Neoantigens, a term derived from "new antigens," refer to a class of antigens that are unique to an individual's tumor cells. These antigens arise from mutations in a patient's DNA, either due to genetic predisposition or environmental factors, such as exposure to carcinogens. Neoantigens play a crucial role in the immune system's ability to distinguish between healthy cells and cancerous ones.

Keywords

Cancer • Neoantigens • Antigens

Introduction

Unlike traditional antigens that are commonly expressed in healthy tissues, neoantigens are specific to the tumor, making them ideal targets for immunotherapy. These mutations generate protein sequences that are not recognized by the immune system as self, allowing the body to mount a targeted immune response against the tumor. The specificity of neoantigens makes them an attractive option for developing precision cancer vaccines. The human immune system is a remarkably intricate network of cells and molecules designed to recognize and eliminate foreign invaders, including pathogens like bacteria and viruses. It can also identify and destroy abnormal cells, such as cancer cells. Central to this function are T cells and B cells, which are part of the adaptive immune system. T cells are the foot soldiers of the immune system and play a critical role in recognizing and attacking cells displaying foreign or mutated antigens. This is where neoantigens come into play. When a tumor cell presents neoantigens on its surface, it acts as a red flag, alerting the immune system to the presence of a potential threat. The process begins when antigen-presenting cells, such as dendritic cells, capture antigens from the tumor and present them to T cells. T cells then recognize the neoantigens as foreign and initiate an immune response. The response can include the recruitment of other immune cells and the activation of cytotoxic T cells, which are responsible for directly killing the tumor cells. This orchestrated immune response can lead to the eradication of the tumor [1].

Neoantigen discovery and vaccine development can be time-consuming and expensive. This can pose challenges, particularly in cases where patients require immediate treatment. While neoantigens are less likely to be subject to immune tolerance than self-antigens, some level of tolerance can still exist. It is essential to ensure that the immune response to neoantigens is robust enough to overcome these tolerance mechanisms. The first step involves sequencing the tumor's DNA to identify the mutations that give rise to neoantigens. This is typically done using next-generation sequencing technologies. Computational tools are employed to predict potential neoantigens from the identified mutations. These tools consider factors like the stability of the mutant peptides, their likelihood to be presented on Major Histocompatibility Complex (MHC) molecules, and their immunogenicity. Not all predicted neoantigens are equally effective at eliciting an immune response. Researchers prioritize the most promising candidates for vaccine development. Once neoantigens are selected, the next step is to create a vaccine. These vaccines can consist of synthetic neoantigen peptides, neoantigen-encoding RNA, or even dendritic cells pulsed with neoantigens. The choice of vaccine format depends on the specific characteristics of the tumor and the patient. The vaccine is then administered to the patient, typically via subcutaneous or intramuscular injection. The vaccine activates the patient's immune system, training it to recognize and target the neoantigens present in the tumor. Patients are closely monitored to assess the immune response and the vaccine's effectiveness. This may involve various imaging techniques, such as CT scans, to track changes in the tumor size and overall disease progression [2,3].

Literature Review

Neoantigen-based vaccines are often used in combination with other immunotherapies or checkpoint inhibitors to enhance their effectiveness. Neoantigen-based cancer vaccines have shown remarkable promise in earlyphase clinical trials. In a groundbreaking study published in the journal Nature in 2017, researchers developed personalized neoantigen vaccines for patients with advanced melanoma. The study demonstrated that neoantigen vaccines induced strong immune responses and significantly improved progressionfree survival. Patients with glioblastoma, a highly aggressive form of brain cancer, have also benefited from neoantigen-based vaccines. A clinical trial published in Nature in 2019 reported encouraging results, with some patients experiencing prolonged survival. Neoantigen vaccines have shown promise in treating colorectal cancer. A study published in Science in 2015 described the successful use of a neoantigen vaccine in a patient with metastatic colorectal cancer.

Tumors are often heterogeneous, and identifying all relevant neoantigens can be challenging. Advances in single-cell sequencing and more comprehensive bioinformatics tools may help address this issue. Neoantigen vaccines may benefit from combination therapies with other immunotherapies or traditional treatments. Optimizing these combinations and understanding their safety profiles is a priority for ongoing research. Identifying predictive biomarkers that can help select the patients most likely to respond to neoantigen-based therapies is crucial for their broader adoption. As with any new therapeutic approach, gaining regulatory approval is a complex process that requires extensive clinical testing and data collection. The use of genetic information for neoantigen prediction raises ethical and privacy concerns. Addressing these concerns while ensuring informed consent from patients is essential [4].

Discussion

Neoantigens represent a promising avenue in the quest to develop precise and personalized cancer vaccines. These unique antigens, arising from tumorspecific mutations, have the potential to revolutionize cancer treatment. By harnessing the immune system's power to recognize and target neoantigens, researchers are working towards treatments that are not only highly effective but also tailored to each patient's specific cancer. The development of neoantigen-based cancer vaccines is still in its early stages, and challenges remain. However, the remarkable successes observed in clinical trials suggest that this approach has the potential to significantly improve the prognosis of cancer patients and change the way we approach cancer therapy. As research continues and technology advances, the future of precision cancer vaccines looks brighter than ever, offering hope for individuals and families affected by this devastating disease.

Neoantigens are unique to each patient's tumor. This individualization of treatment is a significant departure from traditional cancer therapies, which often take a one-size-fits-all approach. Since neoantigens are specific to the tumor, targeting them reduces the risk of inadvertently harming healthy tissues, a common issue with traditional chemotherapy and radiation therapy. Many tumors employ various mechanisms to evade immune detection. Neoantigens offer a way to overcome these evasion strategies because they are not subject to the same immune tolerance as self-antigens. Neoantigens activate the adaptive immune response, which is highly specific and can "remember" the target, potentially providing long-lasting protection against cancer recurrence. Neoantigen-based therapy is not limited to a specific type of cancer. It can be applied to a wide range of malignancies, making it a versatile approach. While the potential of neoantigens in cancer immunotherapy is undeniable, are often heterogeneous, meaning they consist of various subpopulations of cancer cells, each with its unique set of neoantigens. Identifying and targeting all relevant neoantigens can be a complex task. The identification of neoantigens relies heavily on advanced bioinformatics and data analysis. This process requires significant computational resources and expertise, which may not be readily available in all clinical settings [5,6].

Conclusion

These successes underscore the potential of neoantigen-based cancer vaccines to transform the landscape of cancer treatment. However, it's important to note that these therapies are still in the early stages of development, and further research is needed to establish their efficacy and safety. While neoantigen-based cancer vaccines hold immense promise, several challenges must be addressed for these therapies to become more widely accessible and effective. The cost of sequencing and vaccine development, as well as the need for specialized computational tools, can be prohibitive for some patients and healthcare systems. Efforts are ongoing to reduce costs and increase accessibility.

Acknowledgement

None.

Conflict of Interest

There is no conflict of interest by author.

References

  1. Fritsch, Edward F., Ute E. Burkhardt, Nir Hacohen and Catherine J. Wu. "Personal neoantigen cancer vaccines: A road not fully paved." Cancer Immunol Res 8 (2020): 1465-1469.
  2. Google Scholar, Crossref, Indexed at

  3. Jiang, Tao, Tao Shi, Henghui Zhang and Jie Hu, et al. "Tumor neoantigens: From basic research to clinical applications." J Hematol Oncol 12 (2019): 1-13.
  4. Google Scholar, Crossref, Indexed at

  5. Richters, Megan M., Huiming Xia, Katie M. Campbell and William E. Gillanders, et al. "Best practices for bioinformatic characterization of neoantigens for clinical utility." Genome Med 11 (2019): 1-21.
  6. Google Scholar, Crossref, Indexed at

  7. Carreno, Beatriz M., Vincent Magrini, Michelle Becker-Hapak and Saghar Kaabinejadian, et al. "A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells." Science 348 (2015): 803-808.
  8. Google Scholar, Crossref, Indexed at

  9. Ramarathinam, Lakshmi, Supria Sarma, Maja Maric and Min Zhao, et al. "Multiple lineages of tumors express a common tumor antigen, P1A, but they are not cross-protected." J Immunol 155 (1995): 5323-5329.
  10. Google Scholar, Crossref, Indexed at

  11. Sarma, Supria, Yong Guo, Yannik Guilloux and Cheng Lee, et al. "Cytotoxic T lymphocytes to an unmutated tumor rejection antigen P1A: Normal development but restrained effector function in vivo." J Exp Med 189 (1999): 811-820.
  12. Google Scholar, Crossref, Indexed at

arrow_upward arrow_upward