IJCCR_2024v14n5

International Journal of Clinical Case Reports 2024, Vol.14, No.5 http://medscipublisher.com/index.php/ijccr © 2024 MedSci Publisher, registered at the publishing platform that is operated by Sophia Publishing Group, founded in British Columbia of Canada. All Rights Reserved.

International Journal of Clinical Case Reports 2024, Vol.14, No.5 http://medscipublisher.com/index.php/ijccr © 2024 MedSci Publisher, registered at the publishing platform that is operated by Sophia Publishing Group, founded in British Columbia of Canada. All Rights Reserved. MedSci Publisher is an international Open Access publisher specializing in clinical case, clinical medicine, new variations in disease processesregistered at the publishing platform that is operated by Sophia Publishing Group (SPG), founded in British Columbia of Canada. Publisher MedSci Publisher Editedby Editorial Team of International Journal of Clinical Case Reports Email: edit@ijccr.medscipublisher.com Website: http://medscipublisher.com/index.php/ijccr Address: 11388 Stevenston Hwy, PO Box 96016, Richmond, V7A 5J5, British Columbia Canada International Journal of Clinical Case Reports (ISSN 1927-579X) is an open access, peer reviewed journal published online by MedSci Publisher. The journal is considering all the latest and outstanding research articles, letters and reviews in all aspects of clinical case, containing clinical medicine which advance general medical knowledge; the event in the course of observing or treating a patient; new variations in disease processes; as well as the expands the field of clinical relating to case reports. All the articles published in International Journal of Clinical Case Reports are Open Access, and are distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. MedSci Publisher uses CrossCheck service to identify academic plagiarism through the world’s leading plagiarism prevention tool, iParadigms, and to protect the original authors’ copyrights.

International Journal of Clinical Case Reports (online), 2024, Vol. 14, No.5 ISSN 1927-579X http://medscipublisher.com/index.php/ijccr © 2024 MedSci Publisher, registered at the publishing platform that is operated by Sophia Publishing Group, founded in British Columbia of Canada. All Rights Reserved. Latest Content Efficacy of Cancer Vaccines in Clinical Trials: A Systematic Review Jianmin Liu International Journal of Clinical Case Reports, 2024, Vol. 14, No. 5, 230-241 Genetic Mechanisms and Clinical Significance of HPV16/18 Specific Variants in Cervical Cancer Liting Wang International Journal of Clinical Case Reports, 2024, Vol. 14, No. 5, 242-252 Clinical Efficacy Evaluation of Novel Antidiabetic Drugs Xiaojie Xuan International Journal of Clinical Case Reports, 2024, Vol. 14, No. 5, 253-261 Hypotensive Effects of Eucommia ulmoides and its Mechanisms Ran Xin, Qin Wang, Degang Zhao International Journal of Clinical Case Reports, 2024, Vol. 14, No. 5, 262-275 Classic Clinical Case Studies on Early Diagnosis of Alzheimer's Disease Yuchuan Yang, Xiaoying Xu International Journal of Clinical Case Reports, 2024, Vol. 14, No. 5, 276-289 Radiotherapy Innovations in Lung Cancer: Clinical Evidence and Future Prospects Hai Huang International Journal of Clinical Case Reports, 2024, Vol. 14, No. 5, 290-298

International Journal of Clinical Case Reports 2024, Vol.14, No.5, 230-241 http://medscipublisher.com/index.php/ijccr 230 Review Article Open Access Efficacy of Cancer Vaccines in Clinical Trials: A Systematic Review Jianmin Liu Sinovac Biotech Co., Ltd., Haiding, 100193, Beijing, China Corresponding email: Jianminliu@sinovac.com International Journal of Clinical Case Reports 2024, Vol.14, No.5 doi: 10.5376/ijccr.2024.14.0024 Received: 03 Jul., 2024 Accepted: 10 Aug., 2024 Published: 06 Sep., 2024 Copyright © 2024 Liu, This is an open access article published under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Preferred citation for this article: Liu J.M., 2024, Efficacy of cancer vaccines in clinical trials: a systematic review, International Journal of Clinical Case Reports, 14(5): 230-241 (doi: 10.5376/ijccr.2024.14.0024) Abstract This study evaluates the efficacy of cancer vaccines in clinical trials, focusing on their impact on overall survival (OS), progression-free survival (PFS), and tumor response rates. The study synthesizes data from various studies, including vaccines for different cancer types such as non-small cell lung cancer, melanoma, and breast cancer. Results indicate that while cancer vaccines can generate immune responses, their ability to consistently improve clinical outcomes varies depending on factors like patient characteristics, tumor type, and combination with other therapies such as immune checkpoint inhibitors. In many trials, combination therapies demonstrated superior efficacy compared to vaccines alone, suggesting that vaccines may be most effective when used as part of a multimodal approach. The study also highlights challenges such as limited response rates in some cancers, the need for better biomarkers, and optimization of vaccine delivery systems. Overall, cancer vaccines offer promise, but further research is necessary to enhance their clinical impact. Keywords Cancer vaccines; Immunotherapy; Clinical trials; Tumor response; Combination therapies 1 Introduction Cancer vaccines play a crucial role in immunotherapy by activating the immune system to specifically recognize and eliminate cancer cells. Compared to traditional cancer treatments, cancer vaccines offer higher specificity and fewer side effects. However, the clinical application of cancer vaccines faces challenges, as tumor cells can evade immune attacks by downregulating antigen expression or creating an immunosuppressive microenvironment. While early clinical trials demonstrated strong immune responses, they showed limited improvements in clinical outcomes, such as survival rates or tumor reduction (Murahashi et al., 2016).With advancements in tumor immunology, the development of personalized cancer vaccines, particularly through the identification of neoantigens, has been overcoming these challenges. Personalized cancer vaccines are designed using the unique mutations in a patient's tumor, significantly enhancing immune recognition while minimizing damage to healthy tissues (Şahin and Türeci, 2018). Cancer vaccines are categorized into preventive and therapeutic types. Preventive vaccines, such as the HPV vaccine, aim to stop virus-induced cancers and effectively reduce the incidence of cervical and other related cancers. Therapeutic cancer vaccines target patients with existing cancers, stimulating the immune system to recognize tumor antigens and destroy cancer cells. Common types of therapeutic vaccines include peptide vaccines, dendritic cell vaccines, and personalized cancer vaccines (Murahashi et al., 2016). Personalized cancer vaccines, developed based on unique mutations in a patient's tumor, use neoantigens to enhance the specificity of immune responses. These vaccines have shown great potential in preclinical and early-phase trials, especially when combined with immune checkpoint inhibitors, to further boost efficacy (Şahin and Türeci, 2018). The motivation of this study is to address the growing need for a comprehensive evaluation of cancer vaccine trials in recent years, with a focus on their immunogenicity, efficacy, and safety across various cancer types. This study will analyze key clinical indicators, including overall survival, progression-free survival, and tumor response rates, to gain a clearer understanding of vaccine performance. The study will also investigate the impact of combining cancer vaccines with other immunotherapies, particularly PD-1 and CTLA-4 inhibitors. By examining factors that influence vaccine efficacy, such as antigen selection, patient characteristics, and vaccine design, the goal of this

International Journal of Clinical Case Reports 2024, Vol.14, No.5, 230-241 http://medscipublisher.com/index.php/ijccr 231 study is to identify strategies to optimize cancer vaccine therapies and improve patient outcomes. 2 Key Findings from Clinical Trials 2.1 Types and mechanisms of existing cancer vaccines Cancer vaccines can be broadly categorized into several types based on their mechanisms of action: peptide-based vaccines, dendritic cell vaccines, DNA/RNA vaccines, and whole-cell vaccines. Peptide-based vaccines consist of short sequences of amino acids derived from tumor antigens that stimulate the immune system to recognize and attack cancer cells. These vaccines aim to elicit a robust T-cell response by presenting tumor-specific antigens, making them highly specific. However, their effectiveness often depends on factors such as the patient's immune status and the nature of the tumor antigens used (Murahashi et al., 2016). Dendritic cell vaccines leverage dendritic cells' natural role as antigen-presenting cells, priming the immune system to recognize and eliminate cancer cells. These vaccines involve extracting a patient's dendritic cells, loading them with tumor antigens, and then reintroducing them into the patient. For example, the Audencel vaccine used in glioblastoma demonstrated its ability to generate an immune response but failed to show significant clinical benefits in improving survival (Buchroithner et al., 2018). DNA/RNA vaccines deliver genetic material encoding tumor antigens directly into cells, where it is processed and displayed on the cell surface to elicit an immune response. These vaccines are considered flexible and can induce both humoral and cellular immune responses. However, their clinical effectiveness is still being evaluated in ongoing trials. Meanwhile, whole-cell vaccines use inactivated cancer cells that contain a broad spectrum of antigens, making them capable of inducing a wide-ranging immune response, though with less specificity compared to peptide-based vaccines (Dillman, 2015). 2.2 Application of cancer vaccines in different cancer types Cancer vaccines have been applied across various cancer types with varying degrees of success. In prostate cancer, sipuleucel-T became one of the first vaccines to receive FDA approval, significantly improving overall survival in advanced prostate cancer patients. Despite limited effects on progression-free survival, sipuleucel-T has been shown to extend survival by months in several trials, setting a precedent for immunotherapeutic approaches in solid tumors (Dillman, 2015). In melanoma, cancer vaccines like ipilimumab have been studied extensively in combination with immune checkpoint inhibitors. While vaccines alone have struggled to produce significant clinical responses, combination therapies have shown improved outcomes, particularly in patients who develop strong immune responses. Ipilimumab, in combination with vaccines targeting melanoma-associated antigens, has demonstrated survival benefits in a subset of patients, though side effects remain a concern (Lakdawalla et al., 2017). In lung cancer, vaccines are being explored alongside traditional treatments like chemotherapy. For example, a meta-analysis on vaccines for advanced non-small cell lung cancer (NSCLC) demonstrated improved overall survival, progression-free survival, and reduced side effects compared to standard therapies. Although the results are promising, the efficacy of these vaccines varies, and their integration into treatment protocols remains under investigation (Wang et al., 2015). 2.3 Analysis of clinical trial phases (Phase I, II, III) In Phase I trials, the primary objective is to assess the safety of cancer vaccines in humans. These trials typically involve a small number of patients and focus on determining the appropriate dosage and identifying any adverse effects. For instance, a Phase I study on telomerase peptide vaccines for non-small cell lung cancer (NSCLC) demonstrated an increased survival rate in patients who responded immunologically, with minimal toxicity observed. However, the sample size was too small to draw definitive conclusions regarding efficacy (Hansen et al., 2015). Phase II trials shift focus toward evaluating the vaccine's efficacy while continuing to monitor safety. These trials involve larger patient populations and begin to assess clinical endpoints like progression-free survival (PFS) and

International Journal of Clinical Case Reports 2024, Vol.14, No.5, 230-241 http://medscipublisher.com/index.php/ijccr 232 overall survival (OS). In the case of glioblastoma, a Phase II trial of the dendritic cell vaccine Audencel showed no significant improvement in OS or PFS, despite generating immune responses. This highlighted the challenge of translating immunogenicity into clinical efficacy, particularly in aggressive cancers like glioblastoma (Buchroithner et al., 2018). Phase III trials are the most critical, comparing the vaccine's efficacy to the standard of care in large, randomized populations. These trials provide the strongest evidence of clinical benefit. For instance, sipuleucel-T's Phase III trials in prostate cancer established its ability to improve overall survival, leading to its approval. However, other vaccines, such as those for lung cancer, have shown mixed results in Phase III trials, with some improving survival but failing to significantly extend progression-free survival (Wang et al., 2015). 2.4 Efficacy evaluation: overall survival, progression-free survival, tumor response rate Evaluating the efficacy of cancer vaccines involves several key metrics, with overall survival (OS) being the gold standard. Many successful trials have demonstrated significant OS improvements without corresponding benefits in progression-free survival (PFS) or tumor response rates (TRR). For instance, studies on sipuleucel-T and prostvac-VF in prostate cancer showed that while PFS did not improve, OS increased, indicating the potential long-term benefits of immunotherapy beyond early disease progression (Dillman, 2015). Progression-free survival (PFS) is often used as a surrogate endpoint, measuring the time before disease progression. However, its validity as an indicator of overall survival is debated. In glioblastoma and lung cancer, some vaccines improved PFS but failed to demonstrate corresponding improvements in OS, suggesting that PFS alone may not fully capture the vaccine's long-term impact on patient outcomes (Lakdawalla et al., 2017). This disconnect highlights the complexity of evaluating vaccine efficacy, where immune modulation may have delayed but significant effects on survival. Tumor response rate (TRR), which measures the reduction in tumor size, is another important endpoint. However, in cancer vaccines, a high TRR does not always translate to improved survival. Some patients may experience stable disease with limited tumor shrinkage but still benefit from prolonged survival due to immune system engagement. In lung cancer, vaccines have shown modest TRR improvements, but their primary value lies in their ability to prolong survival while reducing side effects compared to conventional therapies (Wang et al., 2015). 3 Correlation Between Immune Response and Efficacy 3.1 Immunogenicity of cancer vaccines The immunogenicity of cancer vaccines refers to their ability to induce a measurable immune response, typically involving the activation of cytotoxic T lymphocytes (CTLs) that target and destroy cancer cells. Cancer vaccines are designed to elicit a response against tumor-associated antigens (TAAs) or neoantigens specific to the tumor. The effectiveness of this immune activation often depends on factors such as the nature of the antigen, the delivery method of the vaccine, and the immunological context of the patient. For instance, peptide vaccines targeting proteins like HER2/neu or MUC1 in breast cancer have shown promising results in eliciting robust CD8+ and CD4+ T cell responses, which are crucial for tumor control (Peres et al., 2015). However, the immunogenicity of cancer vaccines can be suboptimal in certain contexts. Tumors often create an immunosuppressive microenvironment that inhibits the immune system's ability to recognize and destroy cancer cells. Mechanisms such as upregulation of immune checkpoints (e.g., PD-1, CTLA-4) or recruitment of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) can blunt the immune response triggered by vaccines. Despite advances in vaccine design, many vaccines fail to generate a sufficiently strong or sustained immune response to control tumor progression on their own, leading researchers to explore combination therapies to enhance immunogenicity (Pilla et al., 2018). Recent studies have focused on improving vaccine immunogenicity through various strategies, such as the use of adjuvants, optimized delivery platforms, and neoantigen vaccines. Neoantigen vaccines, which are highly specific to individual tumors, have shown great potential in increasing immunogenicity by targeting mutations unique to

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).

International Journal of Clinical Case Reports 2024, Vol.14, No.5, 230-241 http://medscipublisher.com/index.php/ijccr 234 Furthermore, the development of advanced computational models and predictive tools has enabled researchers to better forecast vaccine efficacy based on immune responses. These models analyze factors such as the heterogeneity of the immune response, the presence of multiple antigen-specific T cell responses, and cytokine profiles to predict which patients are most likely to benefit from vaccination. For example, a computational model developed to assess cancer vaccine trials found that generating immune responses against multiple tumor antigens was strongly associated with better clinical outcomes, highlighting the importance of targeting multiple antigens to enhance vaccine efficacy (Lőrincz et al., 2019). Figure 1 T-cell responses specific for antigens encoded by the transfected mRNA (cohort DCM-2) (Adapted from Kyte et al., 2016) Image caption: T-cell proliferation or INFg ELISPOT after stimulation with tDCs and nDC controls. A T-cell response was considered tDC-specific if the response to tDCs was significantly (p <0.05; ANOVA/SNK) higher than in the controls (TCnDC and T cells only). For all five patients shown, a tDC-specific response was demonstrated in post-vaccination samples (week 6 onwards). In patient M108, a tDC-specific response was observed also prior to vaccination (week 0). The assays in Fig. 1 were performed on T cells pre-stimulated once in vitro with tDCs, except for the assays on follow-up sam- ples from patient M109 (week 14–34), which were performed on freshly thawed T cells. T cell only background counts have been subtracted. Columns, mean cpm (prolif- eration assays) or mean spots/well (ELISPOT). Error bars, SEM (Adapted from Kyte et al., 2016) 4 Safety and Adverse Effects 4.1 Summary of common adverse effects Cancer vaccines, like other immunotherapies, are generally well-tolerated but can be associated with certain adverse effects. Common side effects include injection site reactions, such as redness, swelling, and pain, which are often mild and manageable. Systemic effects, such as fatigue, fever, and mild flu-like symptoms, are also

International Journal of Clinical Case Reports 2024, Vol.14, No.5, 230-241 http://medscipublisher.com/index.php/ijccr 235 frequently reported but typically resolve on their own within a few days. For instance, a study evaluating a pancreatic cancer stem cell vaccine found that the most common adverse effects were injection site reactions (54%) and fever (9%), with no significant variation among different dose groups (Lin et al., 2015). Some vaccines, particularly those combined with immune checkpoint inhibitors (ICIs), may induce immune-related adverse events (irAEs), which can affect multiple organs. For example, patients receiving the influenza vaccine while on ICIs were found to have a higher frequency of irAEs, such as pneumonitis and rash, although these events were manageable (Spagnolo et al., 2021). In rare cases, more serious adverse events can occur, such as autoimmune reactions that affect the liver, lungs, or endocrine system. However, these events are typically low in frequency, and when they do occur, they are often treatable with immunosuppressive medications such as corticosteroids (Xu et al., 2018). 4.2 Long-term safety evaluation Long-term safety data for cancer vaccines are crucial to understand their sustained effects and potential late-onset adverse events. A study on patients receiving personalized peptide vaccines (PPV) for lung, colon, and breast cancer reported that no severe long-term adverse events were observed during a median follow-up period of 67.6 months. Most patients experienced sustained immune responses, with only minor injection site reactions being the most common adverse event (Suekane et al., 2022). Studies on combination therapies involving cancer vaccines and ICIs also provide insight into long-term safety. For example, cancer patients who received the influenza vaccine while on pembrolizumab, an anti-PD-1 agent, did not experience an increased rate of serious immune-related events over a 12-month follow-up period, supporting the vaccine's long-term safety in this population (Failing et al., 2020). It is important to note that late-onset adverse events-those occurring months or years after vaccination-are rare but possible, particularly in patients receiving multiple rounds of immunotherapy. Therefore, continued long-term monitoring is essential for evaluating vaccine safety in diverse patient populations (Kyte et al., 2016). 4.3 Management of immune-related adverse events Management of immune-related adverse events (irAEs) is critical in ensuring patient safety, particularly when vaccines are combined with ICIs. Corticosteroids remain the cornerstone of treatment for moderate to severe irAEs. For instance, corticosteroids are often prescribed to manage irAEs such as pneumonitis, colitis, or hepatitis, which are relatively common with immune checkpoint blockade (Rahman et al., 2022). In some cases, if patients do not respond to corticosteroid treatment, additional immunosuppressive agents such as infliximab or mycophenolate mofetil may be necessary. These treatment plans need to be carefully monitored to minimize the risk of secondary infections or other complications. Based on the analysis in the above figure, cancer patients receiving immune checkpoint inhibitors (ICIs) treatment showed a relatively concentrated incidence of immune related adverse events (irAEs) after receiving SARS-CoV-2 vaccine, especially within 30 to 60 days after vaccination (Guo et al., 2024). Although the occurrence of irAEs is related to the timing of vaccination and the type of immunotherapy, through meticulous monitoring and management, the immune response can be controlled, ensuring that adverse events remain within a controllable range. For example, studies on the combination of COVID-19 vaccination and ICIs have shown that despite the occurrence of irAEs these adverse events are kept at a minimal level and mostly manageable with continuous immune response monitoring and appropriate management of patients (Widman et al., 2022). Therefore, these patients should be closely followed up at critical time points after vaccination (such as within 30-60 days) to promptly address any potential irAEs. A multidisciplinary approach is often required for managing severe or complex irAEs, involving oncologists, immunologists, and other specialists to provide comprehensive care. This approach helps mitigate the risk of

International Journal of Clinical Case Reports 2024, Vol.14, No.5, 230-241 http://medscipublisher.com/index.php/ijccr 236 life-threatening toxicities while ensuring that patients can continue to benefit from cancer vaccines and immunotherapies (Ramos-Casals et al., 2020). 5 Key Factors Affecting Efficacy 5.1 Patient characteristics and vaccine response Patient-specific factors such as age, immune status, and genetic background significantly influence the efficacy of cancer vaccines. For instance, older patients generally exhibit immunosenescence, characterized by a diminished immune response due to reduced numbers of naïve T cells and increased exhaustion of memory T cells. Despite these challenges, clinical trials have shown that immune checkpoint inhibitors (ICIs) combined with vaccines still provide comparable efficacy in both younger and older patients. A meta-analysis revealed that both younger (<65 years) and older patients (≥65 years) benefited from ICI-based treatments, though the overall survival (OS) benefit was slightly greater in older patients (Wu et al., 2019). Immune status also plays a critical role in vaccine response. Patients with compromised immune systems, such as those undergoing chemotherapy or suffering from autoimmune diseases, often exhibit reduced vaccine efficacy. Studies have shown that patients with robust immune responses, such as those with a strong T-cell repertoire, tend to experience better clinical outcomes following vaccination, particularly when combined with checkpoint inhibitors like PD-1 or CTLA-4 blockers (Soltani et al., 2022). Patient characteristics such as gender and performance status (ECOG score) also influence vaccine efficacy. In a large clinical meta-analysis, male patients were found to have slightly better outcomes from ICIs combined with vaccines compared to females, particularly in melanoma trials. Additionally, patients with better performance status (ECOG 0) demonstrated improved overall survival and progression-free survival compared to those with poorer baseline health (Yang et al., 2019). 5.2 Vaccine type and dose optimization The type of cancer vaccine used, as well as the dose administered, can greatly affect its efficacy. Different vaccine types, including peptide vaccines, dendritic cell vaccines, and RNA/DNA vaccines, have shown varying degrees of success depending on the cancer type and patient profile. Peptide-based vaccines are often used for their ability to target specific tumor antigens, but their efficacy can be limited by the variability of immune responses among patients. On the other hand, dendritic cell vaccines, which stimulate a more robust immune response, have been effective in several cancers, including prostate and glioblastoma, when optimized for dose and delivery (Yan et al., 2020). Dose optimization is also essential for maximizing efficacy while minimizing adverse effects. In studies comparing low and high-dose vaccines, high doses tend to produce stronger immune responses, but at the risk of increased side effects. A mathematical model exploring the synergy between cancer vaccines and ICIs demonstrated that an optimized combination of both agents, at lower doses than when used individually, could achieve maximal tumor reduction with fewer toxicities (Lai and Friedman, 2017). Personalized dosing strategies are also being explored. For example, neoantigen vaccines, which are highly personalized based on tumor-specific mutations, have shown promise in producing stronger and more targeted immune responses. Ongoing studies suggest that these personalized vaccines, when combined with appropriate dosing schedules, could offer improved survival outcomes in cancers such as melanoma and lung cancer (Liao et al., 2021). 5.3 Combination therapies One of the most promising strategies for enhancing the efficacy of cancer vaccines is combination therapy, particularly with immune checkpoint inhibitors (ICIs) like PD-1 and CTLA-4 blockers. Cancer vaccines alone have shown limited success in inducing durable clinical responses due to the immunosuppressive tumor microenvironment. However, when combined with ICIs, which lift the immune system's "brakes," the vaccine's ability to activate and sustain a robust anti-tumor response is significantly enhanced (Zhao et al., 2019).

International Journal of Clinical Case Reports 2024, Vol.14, No.5, 230-241 http://medscipublisher.com/index.php/ijccr 237 Clinical trials have demonstrated that combination therapies lead to improved overall survival and progression-free survival across various cancer types. For example, in melanoma patients, the combination of a cancer vaccine with nivolumab (an anti-PD-1 therapy) produced a higher overall response rate (33%) compared to nivolumab alone, with some patients experiencing long-term remission (Massarelli et al., 2019). Combination with chemotherapy is another viable approach, especially in cancers like non-small cell lung cancer (NSCLC). Chemotherapy can enhance vaccine efficacy by modulating the tumor microenvironment and increasing antigen availability, making it more accessible to immune cells. Studies have shown that this combination significantly improves survival rates compared to either therapy alone (Khaddour et al., 2023). 6 Clinical Challenges and Future Prospects 6.1 Limitations of current clinical trials Despite the growing interest in cancer vaccines, clinical trials have faced significant challenges, resulting in limited success in translating preclinical efficacy into consistent clinical outcomes. One of the major issues is the heterogeneity of the tumor microenvironment, which can inhibit the immune system's ability to mount an effective response (Wang, 2024). Tumors often create an immunosuppressive environment that limits the efficacy of vaccines by attracting regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), both of which suppress immune responses. This challenge has been seen in various cancers, including head and neck cancers where immune responses were triggered in trials, but clinical outcomes were inconsistent (Schneider et al., 2018). Moreover, the variability in patient immune responses has posed a significant challenge to vaccine efficacy. Some patients respond robustly to vaccines, while others do not, even within the same trial. This inconsistency is due in part to factors such as immune exhaustion, antigen presentation failures, and individual patient characteristics like age or underlying health conditions, which influence how effectively a vaccine can stimulate an immune response (Morse et al., 2021). Another limitation is the design of clinical trials themselves. Some early trials did not select optimal antigens or delivery methods, which limited their ability to stimulate sufficient immune responses. Additionally, trial designs often did not account for the need for combination therapies, such as pairing vaccines with immune checkpoint inhibitors to overcome the suppressive tumor microenvironment (Kaczmarek et al., 2023). 6.2 Future directions and recommendations for cancer vaccine development Looking forward, several advancements are likely to reshape cancer vaccine development. Personalized vaccines that target neoantigens unique to an individual's tumor are a promising avenue. These vaccines offer the potential for more precise targeting of tumor cells and are less likely to be affected by immune escape mechanisms. Ongoing trials of neoantigen vaccines, particularly in melanoma and non-small cell lung cancer (NSCLC), are already showing promising results in producing stronger immune responses and better clinical outcomes (Lopes et al., 2019). Improved vaccine delivery platforms are also critical. Technologies such as nanoparticle-based delivery and viral vector vaccines are enhancing the precision with which antigens are delivered to the immune system, increasing both the intensity and duration of the immune response. Liposomal and dendritic cell-based vaccines are particularly effective in improving antigen presentation and triggering robust T cell responses (Tay et al., 2021). Additionally, combination therapies represent the future of cancer vaccines. Pairing vaccines with immune checkpoint inhibitors (ICIs), such as PD-1 or CTLA-4 blockers, has shown synergistic effects in various cancer types. This approach not only enhances the immune system's ability to recognize and destroy cancer cells but also helps to overcome the suppressive tumor microenvironment that has limited the success of standalone vaccines (Zhao et al., 2019).

International Journal of Clinical Case Reports 2024, Vol.14, No.5, 230-241 http://medscipublisher.com/index.php/ijccr 238 6.3 Future research needs Future research needs to focus on several areas to advance cancer vaccine development. First, there is a need for better biomarkers to predict which patients are likely to respond to vaccination. Identifying biomarkers that correlate with immune responses can help personalize treatments and increase success rates in clinical trials (Ogi and Aruga, 2015). Another critical area for future research is the exploration of new antigens and the optimization of antigen selection. As tumor heterogeneity and immune escape are major barriers to vaccine efficacy, finding new and more effective tumor antigens will be crucial for developing vaccines that can trigger a strong and lasting immune response. This is particularly true for solid tumors like hepatocellular carcinoma (HCC), where antigen selection has proved challenging (Tojjari et al., 2023). Finally, the optimization of combination therapies remains an essential research focus. Understanding the optimal dosing and timing of vaccines in conjunction with immune checkpoint inhibitors, chemotherapies, or other immunotherapies will be key to maximizing their effectiveness and minimizing side effects. Future trials should prioritize exploring these combinations in larger, more diverse patient populations to better understand their potential (Janes et al., 2023). 7 Concluding Remarks Clinical trials investigating cancer vaccines have produced mixed but promising results across various cancer types. While some vaccines have demonstrated improved overall survival (OS) and progression-free survival (PFS), such as sipuleucel-T in prostate cancer and peptide vaccines in melanoma, their efficacy has been inconsistent in other cancers. The heterogeneity of tumor microenvironments and patient-specific factors, such as age and immune status, have led to variable clinical outcomes. Many trials show that vaccines generate strong immune responses, but translating this immunogenicity into meaningful clinical benefits remains challenging. Combination therapies, particularly with immune checkpoint inhibitors, have shown promise in enhancing vaccine efficacy, suggesting that vaccines may perform better as part of a multimodal approach. Cancer vaccines have made a significant contribution to the field of cancer immunotherapy by offering a novel approach to tumor targeting. Unlike conventional treatments such as chemotherapy and radiation, which non-specifically attack both cancerous and healthy cells, vaccines aim to train the immune system to specifically recognize and eliminate cancer cells. This specificity reduces collateral damage to healthy tissues and lowers the risk of adverse effects. Vaccines like sipuleucel-T and new peptide-based vaccines have set a precedent for integrating immunotherapies into standard cancer treatment protocols, particularly in personalized medicine. Additionally, advances in neoantigen vaccines are pushing the boundaries of individualized treatment, offering hope for more precise and effective therapies tailored to each patient's unique tumor profile. The future of cancer vaccine research holds tremendous potential, particularly with the advent of personalized vaccines targeting tumor-specific neoantigens. These vaccines, combined with immune checkpoint inhibitors, may lead to breakthroughs in long-term survival for patients with metastatic or treatment-resistant cancers. However, significant challenges remain. The variability of immune responses across patients, the immunosuppressive tumor microenvironment, and the need for more robust biomarkers to predict vaccine efficacy are critical barriers to overcome. Additionally, optimizing the delivery mechanisms and dosing regimens of vaccines will be essential for maximizing their efficacy and minimizing adverse effects. Ongoing research must focus on addressing these challenges to unlock the full potential of cancer vaccines in clinical practice. Acknowledgments I extend our sincere thanks to two anonymous peer reviewers for their invaluable feedback on the initial draft of this paper. Conflict of Interest Disclosure The author affirms that this research was conducted without any commercial or financial relationships that could be construed as a potential conflict of interest.

International Journal of Clinical Case Reports 2024, Vol.14, No.5, 230-241 http://medscipublisher.com/index.php/ijccr 239 References Buchroithner J., Erhart F., Pichler J., Widhalm G., Preusser M., Stockhammer G., Nowosielski M., Iglseder S., Freyschlag C., Oberndorfer S., Bordihn K., Campe G., Hoffermann M., Ruckser R., Rössler K., Spiegl-Kreinecker S., Fischer M., Czech T., Visús C., Krumpl G., Felzmann T., and Marosi C., 2018, Audencel immunotherapy based on dendritic cells has no effect on overall and progression-free survival in newly diagnosed glioblastoma: a phase ii randomized trial, Cancers, 10(10): 372. https://doi.org/10.3390/cancers10100372 PMID: 30301187 PMCID: PMC6210090 Burg S., 2018, Correlates of immune and clinical activity of novel cancer vaccines, Seminars in Immunology, 39: 119-136. https://doi.org/10.1016/j.smim.2018.04.001 PMID: 29709421 Dillman R., 2015, Cancer vaccines: can they improve survival, Cancer Biotherapy and Radiopharmaceuticals, 30(4): 147-151. https://doi.org/10.1089/cbr.2014.1805 PMID: 25747158 Failing J., Ho T., Yadav S., Majithia N., Riaz I., Shin J., Schenk E., and Xie H., 2020, Safety of influenza vaccine in patients with cancer receiving pembrolizumab., JCO Oncology Practice, 16(7): 573-580. https://doi.org/10.1200/jop.19.00495 PMID: 32048920 PMCID: PMC8462591 Guo S., Gu D.N., and Chen X., 2024, Development and influencing factors of female cancers, Cancer Genetics and Epigenetics, 12(1): 27-36. https://doi.org/10.5376/cge.2024.12.0004 Hansen G., Gaudernack G., Brunsvig P., Cvancarova M., and Kyte J., 2015, Immunological factors influencing clinical outcome in lung cancer patients after telomerase peptide vaccination, Cancer Immunology, Immunotherapy, 64: 1609-1621. https://doi.org/10.1007/s00262-015-1766-5 PMID: 26498005 PMCID: PMC11029677 Janes M., Gottlieb A., Park K., Zhao Z., and Mitragotri S., 2023, Cancer vaccines in the clinic, Bioengineering and Translational Medicine, 9(1): e10588. https://doi.org/10.1002/btm2.10588 PMID: 38193112 PMCID: PMC10771564 Khaddour K., Kamel J., Meeder N., Ziqiao X., Sandra C., Pasquinelli M., Nguyen R., Liu L., Gadi V., Feldman L., and Weinberg F., 2023, Clinical outcomes in patients with metastatic non-small cell lung cancer and head and neck squamous cell cancer treated with immune checkpoint inhibitors according to COVID-19 vaccination status, Journal of Clinical Oncology, 41(16): e14696 https://doi.org/10.1200/jco.2023.41.16_suppl.e14696 Kyte J., Aamdal S., Dueland S., Sæbøe-Larsen S., Inderberg E., Madsbu U., Skovlund E., Gaudernack G., and Kvalheim G., 2016, Immune response and long-term clinical outcome in advanced melanoma patients vaccinated with tumor-mRNA-transfected dendritic cells, OncoImmunology, 5(11): e1232237. https://doi.org/10.1080/2162402X.2016.1232237 PMID: 27999747 PMCID: PMC5139630 Kaczmarek M., Poznańska J., Fechner F., Michalska N., Paszkowska S., Napierała A., and Mackiewicz A., 2023, Cancer vaccine therapeutics: limitations and effectiveness-a literature review, Cells, 12(17): 2159. https://doi.org/10.3390/cells12172159 PMID: 37681891 PMCID: PMC10486481 Katsuda M., Miyazawa M., Ojima T., Katanuma A., Hakamada K., Sudo K., Asahara S., Endo I., Ueno M., Hara K., Yamada S., Fujii T., Satoi S., Ioka T., Ohira M., Akahori T., Kitano M., Nagano H., Furukawa M., Adachi T., and Yamaue H., 2019, A double-blind randomized comparative clinical trial to evaluate the safety and efficacy of dendritic cell vaccine loaded with WT1 peptides (TLP0-001) in combination with S-1 in patients with advanced pancreatic cancer refractory to standard chemotherapy, Trials, 20(1): 242. https://doi.org/10.1186/s13063-019-3332-5 PMID: 31029154 PMCID: PMC6486956 Lakdawalla D., Shafrin J., Hou N., Peneva D., Vine S., Park J., Zhang J., Brookmeyer R., and Figlin R., 2017, Predicting real-world effectiveness of cancer therapies using overall survival and progression-free survival from clinical trials: empirical evidence for the ASCO value framework, Value in Health, 20(7): 866-875. https://doi.org/10.1016/j.jval.2017.04.003 PMID: 28712615 Lőrincz P., Kenéz L.A., Tóth S., Kiss V,. Varga Á., Csizmadia T., Simon-Vecsei Z., and Juhász G., 2019, Vps8 overexpression inhibits HOPS-dependent trafficking routes by outcompeting Vps41/Lt, Elife, 8: e45631. https://doi.org/10.7554/eLife.45631 PMID: 31194677 PMCID: PMC6592680 Lin M., Yuan Y., Liu S., Shi J., Long X., Niu L., Chen J., Li Q., and Xu K., 2015, Prospective study of the safety and efficacy of a pancreatic cancer stem cell vaccine, Journal of Cancer Research and Clinical Oncology, 141: 1827-1833. https://doi.org/10.1007/s00432-015-1968-4 PMID: 25860618

International Journal of Clinical Case Reports 2024, Vol.14, No.5, 230-241 http://medscipublisher.com/index.php/ijccr 240 Lopes A., Vandermeulen G., and Préat V., 2019, Cancer DNA vaccines: current preclinical and clinical developments and future perspectives, Journal of Experimental and Clinical Cancer Research : CR, 38(1): 146. https://doi.org/10.1186/s13046-019-1154-7 PMID: 30953535 PMCID: PMC6449928 Lai X., and Friedman A., 2017, Combination therapy of cancer with cancer vaccine and immune checkpoint inhibitors: a mathematical model, PLoS ONE, 12(5): e0178479. https://doi.org/10.1371/journal.pone.0178479 PMID: 28542574 PMCID: PMC5444846 Liao J., and Zhang S., 2021, Safety and efficacy of personalized cancer vaccines in combination with immune checkpoint inhibitors in cancer treatment, Frontiers in Oncology, 11: 663264. https://doi.org/10.3389/fonc.2021.663264 PMID: 34123821 PMCID: PMC8193725 Massarelli E., William W., Johnson F., Kies M., Ferrarotto R., Guo M., Feng L., Lee J., Tran H., Kim Y., Haymaker C., Bernatchez C., Curran M., Barrese T., Canales J., Wistuba I., Li L., Wang J., Burg S., Melief C., and Glisson B., 2019, Combining immune checkpoint blockade and tumor-specific vaccine for patients with incurable human papillomavirus 16-related cancer: a phase 2 Clinical Trial, JAMA Oncology, 5: 67-73. https://doi.org/10.1001/jamaoncol.2018.4051 PMID: 30267032 PMCID: PMC6439768 Murahashi M., Hijikata Y., Yamada K., Tanaka Y., Kishimoto J., Inoue H., Marumoto T., Takahashi A., Okazaki T., Takeda K., Hirakawa M., Fujii H., Okano S., Morita M., Baba E., Mizumoto K., Maehara Y., Tanaka M., Akashi K., Nakanishi Y., Yoshida K., Tsunoda T., Tamura K., Nakamura Y., and Tani K., 2016, Phase I clinical trial of a five-peptide cancer vaccine combined with cyclophosphamide in advanced solid tumors., Clinical Iimmunology, 166-167: 48-58. https://doi.org/10.1016/j.clim.2016.03.015 Morse M., Gwin W., and Mitchell D., 2021, Vaccine therapies for cancer: then and now, Targeted Oncology, 16: 121-152. https://doi.org/10.1007/s11523-020-00788-w PMID: 33512679 PMCID: PMC7845582 Ogi C., and Aruga A., 2015, Approaches to improve development methods for therapeutic cancer vaccines., Immunology Letters, 164(2): 100-108. https://doi.org/10.1016/j.imlet.2015.02.010 Peres L., Luz F., Pultz B., Brígido P., Araújo R., Goulart L., and Silva M., 2015, Peptide vaccines in breast cancer: the immunological basis for clinical response., Biotechnology Advances, 33(8): 1868-1877. https://doi.org/10.1016/j.biotechadv.2015.10.013 Pilla L., Ferrone S., and Maccalli C., 2018, Methods for improving the immunogenicity and efficacy of cancer vaccines, Expert Opinion on Biological Therapy, 18: 765-784. https://doi.org/10.1080/14712598.2018.1485649 PMID: 29874943 PMCID: PMC8670419 Rahman M., Behl T., Islam M., Alam M., Islam M., Albarrati A., Albratty M., Meraya A., and Bungău S., 2022, Emerging management approach for the adverse events of immunotherapy of cancer, Molecules, 27(12): 3798. https://doi.org/10.3390/molecules27123798 PMID: 35744922 PMCID: PMC9227460 Ramos-Casals M., Brahmer J., Callahan M., Flores-Chávez A., Keegan N., Khamashta M., Lambotte O., Mariette X., Prat A., and Suarez-Almazor M., 2020, Immune-related adverse events of checkpoint inhibitors, Nature Reviews Disease Primers, 6(1): 38. https://doi.org/10.1038/s41572-020-0160-6 PMID: 32382051 PMCID: PMC9728094 Spagnolo F., Boutros A., Croce E., Cecchi F., Arecco L., Tanda E., Pronzato P., and Lambertini M., 2021, Influenza vaccination in cancer patients receiving immune checkpoint inhibitors: a systematic review, European Journal of Clinical Investigation, 51(7): e13604. https://doi.org/10.1111/eci.13604 PMID: 34021591 PMCID: PMC8365730 Suekane S., Yutani S., Toh U., Yoshiyama K., and Itoh K., 2022, Immune responses of patients without cancer recurrence after a cancer vaccine over a long term, Molecular and Clinical Oncology, 16(6): 112. https://doi.org/10.3892/mco.2022.2545 PMID: 35620212 PMCID: PMC9112399 Schneider K., Grønhøj C., Hahn C., and Buchwald C., 2018, Therapeutic human papillomavirus vaccines in head and neck cancer: a systematic review of current clinical trials, Vaccine, 36(45): 6594-6605. https://doi.org/10.1016/j.vaccine.2018.09.027 PMID: 30268734 Soltani M., Savvateeva L., Ganjalikhani-Hakemi M., and Jr A., 2022, Clinical combinatorial treatments based on cancer vaccines: combination with checkpoint inhibitors and beyond, Current Drug Targets, 23(11): 1072-1084. https://doi.org/10.2174/1389450123666220421124542 PMID: 35593358

International Journal of Clinical Case Reports 2024, Vol.14, No.5, 230-241 http://medscipublisher.com/index.php/ijccr 241 Tojjari A., Saeed A., Singh M., Cavalcante L., Sahin I., and Saeed A., 2023, A comprehensive review on cancer vaccines and vaccine strategies in hepatocellular carcinoma, Vaccines, 11(8): 1357. https://doi.org/10.3390/vaccines11081357 PMID: 37631925 PMCID: PMC10459477 Tay R.E., Richardson E.K., and Toh H.C., 2021, Revisiting the role of CD4+ T cells in cancer immunotherapy-new insights into old paradigms, Cancer Gene Ther, 28(1-2): 5-17. https://doi.org/10.1038/s41417-020-0183-x PMID: 32457487 PMCID: PMC7886651 Wang M., Cao J., Liu Y., Xu B., Li D., Zhang X., Li J., Liu J., Wang H., and Wang Z., 2015, Evaluation of tumour vaccine immunotherapy for the treatment of advanced non-small cell lung cancer: a systematic meta-analysis, BMJ Open, 5(4): e006321. https://doi.org/10.1136/bmjopen-2014-006321 PMID: 25872936 PMCID: PMC4401843 Wu Q., Wang Q., Tang X., Xu R., Zhang L., Chen X., Xue Q., Wang Z., Shi R., Wang F., Ju F., Zhang B., and Zhou Y., 2019, Correlation between patients' age and cancer immunotherapy efficacy, OncoImmunology, 8(4): e1568810. https://doi.org/10.1080/2162402X.2019.1568810 PMID: 30906662 PMCID: PMC6422380 Widman A., Cohen B., Park V., McClure T., Wolchok J., and Kamboj M., 2022, Immune-related adverse events among COVID-19-vaccinated patients with cancer receiving immune checkpoint blockade, Journal of the National Comprehensive Cancer Network : JNCCN, 20(10): 1134-1138. https://doi.org/10.6004/jnccn.2022.7048 PMID: 36240845 PMCID: PMC10975484 Wang T., 2024, The application prospects of immunomodulators in cancer treatment, Cancer Genetics and Epigenetics, 12(1): 1-7. https://doi.org/10.5376/cge.2024.12.0001 Xu C., Chen Y., Du X., Liu J., Huang C., Chen L., Zhou G., Li W., Mao Y., Hsu C., Liu Q., Lin A., Tang L., Sun Y., and Ma J., 2018, Comparative safety of immune checkpoint inhibitors in cancer: systematic review and network meta-analysis, The BMJ, 363: k4226. https://doi.org/10.1136/bmj.k4226 PMID: 30409774 PMCID: PMC6222274 Yang F., Wang Y., Nowakowski G., Wang M., Chintakuntlawar A., Halfdanarson T., Pagliaro L., Wei J., Liu B., Molina J., and Markovic S., 2019, Association of sex, age and ECOG performance status with cancer immunotherapy efficacy in randomized controlled trials, Journal of Clinical Oncology, 3(8): e2012534. https://doi.org/10.1200/JCO.2019.37.15_SUPPL.6592 PMID: 32766800 PMCID: PMC7414387 Yan X., Tian X., Wu Z., and Han W., 2020, Impact of age on the efficacy of immune checkpoint inhibitor-based combination therapy for non-small-cell lung cancer: a systematic review and meta-analysis, Frontiers in Oncology, 10: 1671. https://doi.org/10.3389/fonc.2020.01671 PMID: 33072551 PMCID: PMC7538697 Zhao J., Chen Y., Ding Z., and Liu J., 2019, Safety and efficacy of therapeutic cancer vaccines alone or in combination with immune checkpoint inhibitors in cancer treatment, Frontiers in Pharmacology, 10: 1184. https://doi.org/10.3389/fphar.2019.01184

RkJQdWJsaXNoZXIy MjQ4ODYzNQ==