JVR_2024v14n5

Journal of Vaccine Research 2024, Vol.14, No.5, 255-268 http://medscipublisher.com/index.php/jvr 265 promising strategies that are being actively explored to achieve this goal. 8 Future Directions in Cancer Vaccines for Breast Cancer 8.1 Next-generation vaccines: personalized and multi-antigen vaccines The advent of next-generation sequencing and bioinformatics has revolutionized the development of personalized cancer vaccines. These vaccines are designed to target neoantigens, which are unique to each patient's tumor, thereby eliciting a robust and specific immune response. Personalized neoantigen-based vaccines have shown promise in early clinical trials, demonstrating safety, immunogenicity, and preliminary evidence of antitumor activity in various cancers, including melanoma and glioblastoma. The process involves sequencing the tumor's DNA to identify mutations, predicting which neoantigens will be most immunogenic, and then creating a vaccine tailored to these targets. This approach aims to broaden the endogenous repertoire of tumor-specific T cells, thereby enhancing the immune system's ability to recognize and destroy cancer cells (Blass and Ott, 2021). Multi-antigen vaccines, which target several tumor-associated antigens simultaneously, are another promising avenue. These vaccines aim to overcome tumor heterogeneity and reduce the likelihood of immune escape by targeting multiple pathways involved in tumor growth and survival. Combining personalized and multi-antigen approaches could potentially offer a more comprehensive and effective immunotherapy strategy for breast cancer patients. The integration of these next-generation vaccines with other immunotherapeutic modalities, such as immune checkpoint inhibitors, could further enhance their efficacy by overcoming the immunosuppressive tumor microenvironment (Kim et al., 2021). 8.2 Combining vaccines with checkpoint inhibitors Combining cancer vaccines with immune checkpoint inhibitors (ICIs) represents a promising strategy to enhance antitumor immunity. ICIs, such as anti-PD-1 and anti-CTLA-4 antibodies, have revolutionized cancer treatment by unleashing the immune system to attack tumors. However, their efficacy is often limited by the lack of pre-existing T-cell responses in many cancers, including breast cancer (Collins et al., 2018; Thomas et al., 2021). Cancer vaccines can prime and expand tumor-specific T cells, turning "cold" tumors into "hot" ones, thereby making them more susceptible to ICIs (Collins et al., 2018; Wang, 2024). Preclinical and early clinical studies have shown that combining vaccines with ICIs can lead to synergistic antitumor effects. For instance, a study combining a MUC1 mRNA nano-vaccine with CTLA-4 blockade demonstrated significant inhibition of tumor growth in triple-negative breast cancer (TNBC) models (Liu et al., 2018). This combination therapy enhanced the activation and expansion of tumor-specific T cells, leading to a more robust and durable immune response compared to either treatment alone (Liu et al., 2018). Additionally, personalized cancer vaccines combined with ICIs have shown acceptable safety profiles and minimal additional toxicity, making them a viable option for enhancing clinical outcomes in breast cancer patients (Zhao et al., 2019). 8.3 Vaccine development in metastatic breast cancer Metastatic breast cancer (MBC) remains a significant clinical challenge due to its aggressive nature and poor prognosis. Developing effective vaccines for MBC requires overcoming several hurdles, including the immunosuppressive tumor microenvironment and the heterogeneity of metastatic lesions. Recent advances in vaccine technology, such as the use of dendritic cell (DC)-based vaccines and nanoparticle delivery systems, offer new avenues for targeting metastatic disease. DC-based vaccines have shown potential in inducing strong antitumor immune responses by presenting tumor antigens to T cells and activating them. Combining DC vaccines with ICIs, such as PD-1 blockade, has been proposed as a strategy to enhance their efficacy by overcoming immune evasion mechanisms. Additionally, nanoparticle-based mRNA vaccines targeting specific tumor antigens have demonstrated promising results in preclinical models of TNBC, suggesting their potential for treating metastatic disease (Liu et al., 2018). Ongoing clinical trials are exploring the efficacy of various vaccine platforms in combination with standard therapies and ICIs in MBC patients. These studies aim to identify optimal combinations and treatment regimens

RkJQdWJsaXNoZXIy MjQ4ODYzNQ==