International Journal of Clinical Case Reports 2024, Vol.14, No.5, 230-241 http://medscipublisher.com/index.php/ijccr 233 the patient's cancer. In melanoma, for example, dendritic cell vaccines loaded with neoantigen RNA have demonstrated the ability to induce a broad immune response with a polyfunctional cytokine profile, improving patient outcomes in clinical trials (Kyte et al., 2016). 3.2 Relationship between immune response and clinical outcomes The relationship between immune response and clinical efficacy is a cornerstone of cancer vaccine development. Clinical outcomes, such as overall survival (OS), progression-free survival (PFS), and tumor response rates, are closely tied to the strength and durability of the immune response elicited by the vaccine. In multiple studies, patients who generate a strong immune response to the vaccine show significantly improved clinical outcomes compared to non-responders. For instance, in non-small cell lung cancer (NSCLC), patients who developed an immune response to a telomerase peptide vaccine experienced a marked increase in both OS and PFS. Immune responders lived an average of 54 months, compared to just 13 months for non-responders, highlighting the importance of effective immunogenicity for clinical benefit (Hansen et al., 2015). The effectiveness of vaccines is often negatively affected by immune suppressive cells, such as regulatory T cells (Tregs) and myeloid derived suppressor cells (MDSCs). The high levels of these cells in the tumor microenvironment are often associated with poorer clinical outcomes as they suppress immune responses to vaccines. In vaccinated patients, if the level of Tregs can be reduced and a favorable cytokine environment can be established, especially a higher IFN γ/IL-10 ratio, better survival outcomes are usually observed. The T cell response shown in Figure 1 confirms that the effectiveness of the vaccine is closely related to the intensity of the immune response. Patients with strong immune responses, such as those exhibiting high T cell proliferation and IFN γ production, often have better prognosis. On the contrary, the presence of immunosuppressive microenvironments such as Tregs or MDSCs activity may limit the efficacy of vaccines, leading to poorer clinical outcomes. This further supports the strategy of enhancing vaccine efficacy by modulating immune responses or in combination with other therapies such as immune checkpoint inhibitors. (Kyte et al., 2016). In addition to T cell responses, the quality of the cytokine response is also critical. Polyfunctional T cells that can produce multiple cytokines (e.g., IFNγ, IL-2, TNF-α) are more effective in controlling tumors. Studies have shown that patients with polyfunctional immune responses tend to have better clinical outcomes, as these cells are more capable of sustaining an anti-tumor attack. This correlation between immune response and clinical efficacy underscores the need for immune monitoring to optimize vaccine strategies and identify the most responsive patient populations (Burg, 2018). 3.3 Role of immune monitoring in vaccine efficacy assessment Immune monitoring plays a pivotal role in assessing the efficacy of cancer vaccines, as it allows researchers to track the immune response over time and adjust treatment strategies accordingly. Techniques such as flow cytometry, ELISPOT assays, and cytokine profiling are commonly used to measure the activation of immune cells and the production of cytokines in response to vaccination. These tools provide valuable insights into whether a vaccine is effectively stimulating the immune system, which is essential for predicting clinical outcomes. For example, delayed-type hypersensitivity (DTH) testing and T cell proliferation assays were used in a melanoma trial to assess the immune response to tumor-mRNA-loaded dendritic cell vaccines, demonstrating that immune responders had significantly better survival outcomes than non-responders (Kyte et al., 2016). In addition to measuring immune responses, immune monitoring can help identify biomarkers that predict which patients are most likely to benefit from cancer vaccines. For instance, high levels of PD-1 or CTLA-4 expression on T cells may indicate an exhausted immune state, which could be overcome by combining vaccines with immune checkpoint inhibitors. Monitoring these markers can guide clinicians in tailoring treatment plans, such as adding checkpoint inhibitors to enhance vaccine efficacy in patients with high levels of immune suppression. This approach has been shown to improve outcomes in several cancers, including melanoma and lung cancer (Zhao et al., 2019).
RkJQdWJsaXNoZXIy MjQ4ODYzNQ==