Next Article in Journal
Cell Proliferation, Chondrogenic Differentiation, and Cartilaginous Tissue Formation in Recombinant Silk Fibroin with Basic Fibroblast Growth Factor Binding Peptide
Previous Article in Journal
Comparison of Optical Properties and Fracture Loads of Multilayer Monolithic Zirconia Crowns with Different Yttria Levels
Previous Article in Special Issue
Multifunctional Iron Oxide Nanoparticles as Promising Magnetic Biomaterials in Drug Delivery: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mannan-Decorated Lipid Calcium Phosphate Nanoparticle Vaccine Increased the Antitumor Immune Response by Modulating the Tumor Microenvironment

1
Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
2
Yong Loo Lin School of Medicine, National University of Singapore, Singapore 19077, Singapore
*
Author to whom correspondence should be addressed.
These authors contributed equally to this study.
J. Funct. Biomater. 2024, 15(8), 229; https://doi.org/10.3390/jfb15080229
Submission received: 23 May 2024 / Revised: 13 August 2024 / Accepted: 14 August 2024 / Published: 16 August 2024
(This article belongs to the Special Issue Nanomaterials for Drug Targeting and Drug Delivery)

Abstract

:
With the rapid development of tumor immunotherapy, nanoparticle vaccines have attracted much attention as potential therapeutic strategies. A systematic review and analysis must be carried out to investigate the effect of mannose modification on the immune response to nanoparticles in regulating the tumor microenvironment, as well as to explore its potential clinical application in tumor therapy. Despite the potential advantages of nanoparticle vaccines in immunotherapy, achieving an effective immune response in the tumor microenvironment remains a challenge. Tumor immune escape and the overexpression of immunosuppressive factors limit its clinical application. Therefore, our review explored how to intervene in the immunosuppressive mechanism in the tumor microenvironment through the use of mannan-decorated lipid calcium phosphate nanoparticle vaccines to improve the efficacy of immunotherapy in patients with tumors and to provide new ideas and strategies for the field of tumor therapy.

1. Introduction

Recently, tumor immunotherapy, as a revolutionary treatment, has brought new hope for patients with tumors [1,2,3]. However, despite some success, it still faces a number of challenges and limitations [4].
The core idea of tumor immunotherapy is to activate the body’s own immune system to attack and eliminate tumor cells [5]. However, the presence of the tumor microenvironment seriously affects the activity and function of immune cells, thus weakening the effectiveness of immunotherapy [6,7,8,9,10]. This microenvironment includes tumor cells, immune cells, blood vessels, interstitial cells, and other components, which interact with each other in a complex manner, thus resulting in immunosuppression [11]. The overexpression of immunosuppressive factors, the existence of immune escape mechanisms, and the immunosuppressive effect of tumor cells are some of the main challenges facing tumor immunotherapy [12]. To overcome these challenges, in recent years, scientists have focused on identifying new strategies and methods to improve the effectiveness of tumor immunotherapy [13,14,15,16]. As a new therapeutic strategy, nanoparticle vaccines have attracted much attention [17,18,19,20]. As a new nanoparticle carrier, the mannan-decorated lipid calcium phosphate nanoparticle vaccine has unique advantages and potential application prospects [21,22,23,24,25]. Mannose modification can make it easier for nanoparticles and tumor cells to be recognized and taken up [26]. This effect can increase the amount of vaccine that is enriched in tumor tissues, which improves the effectiveness of tumor immunotherapy [27,28,29,30]. In addition, mannose modification can also regulate the expression of immunosuppressive factors in the tumor microenvironment, destroy the interaction between tumor cells and immune cells, and further enhance the effect of immunotherapy [31]. We found that this nanoparticle vaccine can precisely target tumor cells and significantly improve the immune system’s ability to recognize and clear tumors by enhancing antigen delivery and immune cell activation. This study revealed how the vaccine, through a mannan-decorated strategy, regulates inflammation in the tumor microenvironment, inhibits immune escape mechanisms, and promotes the infiltration and activation of immune cells, thereby enhancing tumor-specific T-cell responses and cytotoxic activity [32]. The specific design of nanoparticles can optimize the efficiency of antigen delivery and improve the stability and biocompatibility of vaccines [33]. This reveals the potential of mannan-decorated lipid calcium–phosphorus nanoparticle vaccines as a novel immunotherapy strategy, providing an important theoretical and experimental basis for the development of more effective cancer immunotherapy protocols.
Our review systematically analyzes the research progress of tumor vaccines in enhancing the antitumor immune response and regulating the tumor microenvironment to provide a theoretical basis and practical guidance for further research in this field.

2. Regulation and Influence of the Tumor Microenvironment

The tumor microenvironment is an important aspect of tumor growth and development, and its characteristics are closely related to immunosuppressive mechanisms [34,35,36]. There are many immunosuppressive factors, such as transforming growth factor β (TGF-β) and interleukin-10 (IL-10), in the area around the tumor [35]. These factors can prevent immune cells from performing their functions and hinder their ability to find and kill tumor cells. Many immunosuppressant molecules, such as programmed death ligand-1 (PD-L1) and acidic extracellular matrix protein (TSP), are produced by tumor cells and surrounding cells [36,37,38,39,40]. These molecules interact with ligands on the surface of immune cells to allow the immune system to tolerate and escape. In addition, the highly acidified and hypoxic environment in the tumor microenvironment is also an important factor in immunosuppression, which not only affects the activity and function of immune cells but also induces apoptosis and functional abnormalities in immune cells [41,42,43,44]. The inflammatory response and immune cell infiltration in the tumor microenvironment are also closely related to immunosuppression [45,46,47,48]. The inflammatory response can promote the activation and infiltration of immune cells; however, it can also lead to their functional polarization and immune escape [49]. The tumor microenvironment provides favorable conditions for tumor escape by regulating the activity, function, and quantity of immune cells and changing the local physiological environment, thereby inhibiting the immune response [50].
The combined application of 3D bioprinting technology and bio-nanocarrier technology has led to the construction of a new tumor treatment platform [51,52,53,54]. Three-dimensional bioprinting can accurately manufacture complex three-dimensional structures, whereas bio-nanocarrier technology can effectively deliver drugs or genes [55]. This combined application platform can enable customized tumor treatment programs, thus targeting drugs or gene carriers to the tumor site to improve treatment effectiveness [56]. In addition, the combination of these two technologies can improve the tumor immune microenvironment [57,58,59,60]. The immunosuppressive tumor microenvironment can be controlled by the release of nanocarriers carrying specific immunomodulators. It can also boost the activity of immune cells, help tumor cells die and immune cells invade, and improve the immune response of patients [61]. This combined application platform provides a new method for personalized and precise tumor therapy and has important clinical application prospects (Figure 1).
Cell interactions in the tumor microenvironment are closely related to those in the tumor mesenchyme and have important effects on the immune response [62,63,64]. The tumor stroma is composed of tumor cells, stromal cells, and stroma, and its complex cellular interactions affect the characteristics of the tumor microenvironment and the mechanism of immunosuppression [65]. Tumor cells influence the behavior of surrounding cells by secreting cytokines and chemokines, such as vascular endothelial growth factor (VEGF) and tumor necrosis factor (TNF), and by regulating tumor stromal formation and function [66]. Mesenchymal cells, including tumor-associated macrophages (TAMs) and cancer-associated fibrocytes (CAFs), interact with tumor cells by secreting cytokines and molecules, such as TGF-β and IL-6, to promote tumor growth, invasion, and metastasis and inhibit the activity of immune cells [67,68,69,70]. In a mouse model of brain metastases, studies have shown that the mannan-decorated lipid calcium–phosphorus nanoparticle vaccine has a significant targeted penetration ability and can successfully penetrate the meninges and accurately locate tumor cells.
This nanoparticle carrier achieves effective drug delivery through specific binding to tumor cells, thus significantly killing tumor cells. More importantly, in this process, the nanoparticles not only directly act on the tumor cells but also regulate the interaction between the cells and the tumor matrix in the tumor microenvironment. The tumor microenvironment is composed of tumor cells, stromal cells, immune cells, and extracellular matrix, and its complex dynamic relationship plays a key role in the genesis and development of tumors [71]. By regulating this microenvironment, the mannan-decorated nanoparticles effectively destroy the interaction between tumor cells and stromal cells and weaken the viability and aggressiveness of tumor cells [72]. The nanoparticle vaccine not only works by directly killing tumor cells but also further enhances the antitumor immune response by modulating cell interactions in the tumor microenvironment (Figure 2).
Immune escape and tumor suppressor cells in the tumor microenvironment are important reasons for the hindered immune response [73,74,75]. Tumor cells and their surrounding cells and molecules work together in the tumor microenvironment to form a pattern of immune escape [76]. Tumor cells express excessive immunosuppressive molecules, such as PD-L1 and PD-L2, and immunosuppressive factors, such as TGF-β and IL-10. These factors prevent immune cells from functioning and hinder their ability to find and kill tumor cells [77]. In addition, tumor suppressor cells in the area around the tumor (such as TAMs and Tregs) control the immune response and help the tumor grow and spread by releasing immunosuppressive substances such as IL-10 and TGF-β [78,79,80].
Lipid calcium phosphate nanoparticles are usually prepared by the thin-film solution method, in which phospholipids and calcium phosphate are mixed in a certain proportion to form a lipid–calcium ion complex. Mannose-modified lipid calcium phosphate nanoparticles were formed by combining mannose with a lipid–calcium complex via the addition of an appropriate amount of mannose modifier.
In the tumor microenvironment, immune escape and tumor suppressor cells are key factors in tumor development and treatment difficulties. Tumor cells evade the surveillance and attack of the immune system through a variety of mechanisms, including regulating the activity of immunosuppressive cells such as regulatory T cells and myeloid suppressor cells, thereby suppressing the antitumor immune response. In addition, tumor suppressor cells in the tumor microenvironment promote tumor cell growth and metastasis by secreting a variety of cytokines and growth factors. It was found that the mannan-decorated lipid calcium–phosphorus nanoparticle vaccine can effectively regulate the tumor microenvironment and enhance the antitumor immune response, thus overcoming immune escape and inhibiting the function of tumor suppressor cells. Studies [81,82] in mouse models showed that the mannan-decorated lipid calcium–phosphorus nanoparticle vaccine was detected and monitored by photoacoustic imaging (PAI), demonstrating its distribution and biological distribution characteristics in vivo. Photoacoustic imaging technology combines the high contrast of optical imaging with the high resolution of acoustic imaging to monitor the distribution of nanoparticles in the body in real time without damaging tissues. This imaging technique demonstrated that mannan-decorated nanoparticles can effectively target tumor tissue and accumulate in the tumor microenvironment, thereby exerting their antitumor effects.
The accumulation of mannan-decorated nanoparticles at tumor sites can be clearly observed by photoacoustic imaging, which provides important support for the further understanding of its mechanism of action. These nanoparticles were not only able to directly kill tumor cells but also enhanced the antitumor immune response by modulating immune escape and tumor suppressor cells in the tumor microenvironment (Figure 3).

2.1. Role of Lipid Calcium Phosphate Nanoparticles in the Immune System

Lipid Calcium Phosphate Nanoparticles in the Immune System

Lipid calcium phosphate nanoparticles (100–200 nanometers, spherical) are important nanocarriers that have the potential to modulate antitumor immune responses in the immune system [83]. These nanoparticles are structurally designed to improve vaccine stability, biocompatibility, and immunogenicity. These nanoparticles can also mimic the structure and appearance of the virus. This strongly affects the immune system, which improves the body’s ability to find and destroy tumor cells [84,85,86,87]. In general, as an effective vaccine carrier, lipid calcium phosphate nanoparticles play an important role in the immune system, thus enhancing the antitumor immune response by promoting antigen presentation and immune cell activation and providing new strategies and hope for tumor treatment. mRNA LNPS (mRNA lipid nanoparticle) and calcium phosphate nanoparticles play different roles in vaccine delivery systems. mRNA LNPs are primarily used to deliver mRNA vaccines, and at their core are lipid nanoparticles that enclose mRNA molecules. This nanoparticle has a small particle size and good biocompatibility, which can effectively protect mRNA from degradation, improve its stability, and promote mRNA uptake in cells. In contrast, lipid calcium phosphate nanoparticles, with a calcium phosphate core and a surface modified with mannose to enhance targeting, are more commonly used to deliver proteins or antigens. In addition, the two have different application fields and characteristics. mRNA LNPs are widely used in the delivery of mRNA vaccines. Due to the easy degradation and instability of mRNA, LNPs can effectively wrap and protect mRNA and release it in the body to trigger an immune response. However, lipid calcium phosphate nanoparticles are more commonly used to deliver protein antigens, enhance targeting through mannan-decorated pathways, enter the tumor microenvironment, activate immune cells, and enhance the antitumor immune response.
Our study used photoacoustic imaging (PA) to measure oxidative stress in lipid calcium phosphate nanoparticles. Lipid calcium phosphate nanoparticles were injected into the tumor site to locate the targeted organs and tumor sites in vivo, and the distribution and signal intensity of the lipid calcium phosphate nanoparticles were monitored in real time by using photoacoustic imaging technology; moreover, the intensity of the PA signal reflected the degree of oxidative stress [88]. During the observation process, we can infer the degree of oxidative stress in the tumor microenvironment from changes in signal intensity and further evaluate the role of lipid calcium phosphate nanoparticles in modulating the tumor immune response [89]. This process effectively combines lipid calcium phosphate, nanoparticle technology, and photoacoustic imaging technology to provide a feasible, noninvasive measurement method for the study of oxidative stress in the tumor microenvironment and provides an important reference for the optimal design of antitumor immunotherapy (Figure 4).

3. Design and Preparation of a Mannan-Decorated Lipid Calcium Phosphate Nanoparticle Vaccine

3.1. Effect of Mannose Modification on Vaccines

Mannose modification can confer good biocompatibility and immunological activity on nanoparticle vaccines [90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121]. Mannose modification can improve the stability of a vaccine and increase its circulation time in the body.
Mannose modification plays an important role in improving the stability of vaccines by covalently linking mannose to the surface of lipid calcium phosphate nanoparticles to form a protective film, which effectively prevents vaccines from being affected by the external environment in vivo. This protective film can resist adverse factors, such as enzyme degradation, pH changes, and temperature fluctuations, increasing the stability and reliability of the vaccine during storage and delivery. In addition, the addition of mannose can increase the duration of the vaccine cycle in the body. The modified nanoparticles have better biocompatibility and anti-clearance and can evade clearance by the liver or spleen, extending their residence time in the blood circulation.
In addition, mannan-decorated nanoparticles can bind specifically to immune cells to improve the cellular uptake rate and antigen delivery efficiency of the vaccine [122,123,124]. Mannose modification can also activate certain immune signaling pathways and improve the ability of antigen-presenting cells to express antigens, which strengthens the immune response of antigen-specific T cells [125,126,127,128]. During the design and preparation of the mannan-decorated lipid calcium phosphate nanoparticle vaccine, the influence of the mannose modification on the vaccine is reflected in its ability to improve its stability, enhance its immune activity, and promote antigen presentation, which provides strong technical support for tumor immunotherapy [129].
Mannan-decorated lipid calcium phosphate nanoparticles have demonstrated a potentially revolutionary role in cancer therapy, and their ability to target cancer-causing long noncoding RNAs (ARAs) has brought new hope for cancer therapy [130]. By regulating the tumor microenvironment, nanoparticles can not only inhibit the growth and spread of tumor cells but also enhance the body’s antitumor immune response [131]. Moreover, combined with the research progress in tumor immunity, the results demonstrated that the use of mannan-decorated lipid calcium phosphate nanoparticles is not only a method of direct attack against tumor cells but also an innovative strategy for promoting the body’s immune system to participate in antitumor processes (Figure 5).

3.2. Design and Preparation of Lipid Calcium Phosphate Nanoparticles

3.2.1. Preparation Method and Structural Advantages

Lipid calcium phosphate (CaP) nanoparticles have attracted much attention due to their unique advantages in vaccine delivery systems. Their design and preparation are essential for improving the bioavailability and immunological efficacy of vaccines [132,133,134,135]. Typically, the preparation process includes the solvent precipitation method and the coprecipitation method [136]. During solvent precipitation, the addition of phosphate and calcium ions causes the formation of calcium phosphate nanoparticles in solution. The coprecipitation of the drug and calcium phosphate is typically how the coprecipitation method produces the drug’s carrier [137,138,139,140,141,142,143,144,145,146,147,148,149,150]. The structural advantages provide a good platform for vaccine delivery and provide a foundation for regulating the tumor microenvironment and enhancing the antitumor immune response.

3.2.2. Stability and Biocompatibility of Nanoparticles

Lipid calcium phosphate (CaP) nanoparticles are an important vaccine delivery system and have potential applications in antitumor immunotherapy [151]. In the design and preparation of these materials, we need to consider the stability and biocompatibility of the nanoparticles, which are essential for improving vaccine effectiveness and safety [152,153,154]. The stability of nanoparticles can be achieved by adjusting the preparation methods and adding surface modifiers. During the preparation process, the size, morphology, and dispersion of nanoparticles can be controlled via solvent precipitation or coprecipitation to ensure their stability [155]. Additionally, the use of appropriate surface modifiers, such as polyvinylpyrrolidone (PVP), can increase the stability of nanoparticles and prevent them from being cleared from the bloodstream and breaking down in living organisms [156].
Biocompatibility is an important indicator for evaluating the application of nanoparticles [157]. Mannan-decorated lipid calcium phosphate nanoparticles have received much attention due to their good biocompatibility [158]. Mannose, a natural sugar in the human body, has good biocompatibility and biodegradability and can reduce the immune response and toxic side-effects on the body [159]. Mannan-decorated nanoparticles can effectively avoid the clearance and decomposition of nanoparticles caused by immune responses, thus extending their circulation time in the body and increasing their accumulation in tumor tissues [160]. Additionally, changing the mannose concentration can improve the specific binding between nanoparticles and tumor cells, thus allowing for more precise targeted delivery and a better immune response against the tumor in the vaccine [161,162,163,164,165].
In general, the stability and biocompatibility of lipid calcium phosphate nanoparticles are problems that need to be considered and solved. Through rational design and preparation methods, as well as the introduction of biocompatible modifications such as mannose, the application of nanoparticles in antitumor immunotherapy can be effectively improved, thus providing strong support for regulating the tumor microenvironment and enhancing the antitumor immune response.

4. Immunomodulatory Mechanism of Mannan-Decorated Lipid Calcium Phosphate Nanoparticle Vaccine

4.1. Tumor Antigen Presentation and T-Cell Activation

4.1.1. Mannan-Decorated Lipid Calcium Phosphate Nanoparticle Vaccine in Tumor Immunity

The mannan-decorated lipid calcium phosphate nanoparticle vaccine plays an important role in enhancing the antitumor immune response, and its immune regulatory mechanism involves several mechanisms [166]. As carriers, these nanoparticles can effectively load tumor antigens and their related immune stimulators (such as proteins and nucleic acids) in a stable manner on their surface or on the inside. Mannan-decorated nanoparticles can achieve precise, targeted delivery through specific binding to tumor cell surfaces [167,168,169,170]. This targeted loading allows the nanoparticles to be more efficiently sought out in tumor tissue and consumed by tumor cells [171]. NPs release tumor antigens that are loaded on themselves. This makes it easier for antigen-presenting cells, such as dendritic cells, to take in and process these antigens, which then causes immune cells to recognize and respond to the tumor antigens. In addition, mannose can interact with specific receptors on the surface of tumor cells to promote intracellular phagocytosis and the internal presentation of nanoparticles [172,173,174,175,176,177,178,179,180]. Finally, the release of these immune stimulators and the presentation of tumor antigens activate the body’s immune system, especially by promoting the activation and proliferation of antigen-specific T cells and B cells, thus strengthening the immune response to tumors [181,182,183,184].
The mannan-decorated lipid calcium phosphate nanoparticle vaccine regulates the tumor microenvironment and enhances the antitumor immune response by targeting tumor antigen delivery, promoting antigen presentation, and activating immune cells, thus providing new ideas and methods for tumor therapy.

4.1.2. Activation of T Cells by a Mannan-Decorated Lipid Calcium Phosphate Nanoparticle Vaccine

The mannan-decorated lipid calcium phosphate nanoparticle vaccine changes the microenvironment of the tumor, which boosts the immune response against it [185]. One method of achieving this effect is by activating T cells; specifically, T cells are an important part of the immune system and play a key role in recognizing and eliminating tumor cells [186,187,188]. Mannan-decorated nanoparticles can enhance the immune response by promoting the activation and proliferation of T cells in a variety of ways [189].
Mannan-decorated nanoparticles can effectively improve the delivery efficiency of tumor antigens. These nanoparticles act as carriers that can stably load tumor antigens and release them into the tumor microenvironment [190]. Antigen-presenting cells (such as dendritic cells) take up and process these tumor antigens before presenting them to T cells and inducing an immune response to the tumor antigen. Mannan-decorated nanoparticles modulate immunosuppressive factors in the tumor microenvironment, thereby reducing T-cell suppression [191,192,193,194,195]. In the tumor microenvironment, the presence of immunosuppressive factors (such as PD-L1 and TGF-β) can inhibit the activation and function of T cells [196]. NPs modified with mannose can control the production and release of these immune-suppressing substances by interacting with specific receptors on the surface of tumor cells. This makes T cells less inhibited and more active, thus leading to increased cell growth and activation [197]. The mannan-decorated nanoparticles also activated T-cell costimulatory signaling pathways. Costimulatory signaling is a key factor in T-cell activation and proliferation, and the CD28/B7 and CD40/CD40L signaling pathways play important roles in T-cell activation and function [198,199,200]. NPs modified with mannose can activate these costimulatory signaling pathways by attaching to the correct receptors on the surface of T cells. This makes the T-cell immune response stronger.
The use of a mannan-decorated lipid calcium phosphate nanoparticle vaccine, which is an innovative immunotherapy method, has received extensive attention and research in recent years [201]. By modulating the tumor microenvironment, this vaccine can significantly enhance the antitumor immune response, thus providing new possibilities for tumor treatment. Several studies [202,203,204,205] have explored the treatment of this nanoparticle nucleic acid vaccine through clinical trials. These clinical trials typically involve the treatment of tumor patients in groups, with one receiving the mannan-decorated lipid calcium phosphate nanoparticle vaccine and the other receiving either standard treatment or a placebo. The main purpose of the trial was to assess the effect of the vaccine on tumor growth in patients and the extent to which it activated the immune system [206]. By comparing the effects of treatment on different groups of patients, researchers can assess the effectiveness and safety of the vaccine. In clinical trials [207,208,209,210,211,212], researchers typically examine data on several aspects, including changes in tumor size, longer patient survival, and increased immune cell activity. These data can not only help determine the therapeutic effect of the vaccine but also provide an important basis for further optimization of the vaccine design and treatment plan (Figure 6).

4.2. Enhancement in the Tumor Immune Response and Establishment of Immune Memory

The use of a mannan-decorated lipid calcium phosphate nanoparticle vaccine is a novel tumor immunotherapy method that can enhance the immune response to tumors by regulating the tumor microenvironment [213]. Previous studies [214,215,216,217,218,219,220] have shown that vaccines can activate the body’s immune system, promote the expression and recognition of tumor-associated antigens, and trigger a specific immune response against tumor cells. Through mannose modification, the vaccine can be more effectively taken up by antigen-presenting cells and improve the efficiency of antigen delivery in the lymph nodes, thus further activating immune cells such as dendritic cells and T cells and enhancing the potential of the immune response [221].
In the establishment of immune memory, the application of a vaccine has also shown remarkable results [222,223,224,225]. After inoculation with mannan-decorated lipid calcium phosphate nanoparticles, the body can form a long-term memory of tumor antigens [226]. This immune memory allows the body to recognize and clear tumor cells quickly and efficiently during subsequent tumor invasion, thereby reducing the risk of tumor recurrence and metastasis. In addition, the establishment of immune memory also provides a solid foundation for subsequent immunotherapy, thus enabling the body to produce a more durable and powerful response to further treatment with tumor vaccines or other immunomodulators [227,228,229,230].
As a new method to treat tumors with immunotherapy, a mannan-decorated lipid calcium phosphate nanoparticle vaccine has shown great promise in improving the immune response to tumors and building immune memory [231,232,233,234,235]. This provides new ideas and strategies for the future treatment of cancer and is expected to play an important role in clinical practice, thus resulting in more effective treatments and a better quality of life for patients [236,237,238,239,240]. The main determinants of drug resistance include tumor microenvironment heterogeneity, immunosuppressive mechanisms, and inefficient drug delivery [241]. A mannan-decorated lipid calcium phosphate nanoparticle vaccine can improve the immunogenicity of tumor cells, regulate the tumor microenvironment, and promote an antitumor immune response by stimulating natural antigen presentation (Figure 7).

4.3. Analysis of Immune Cell Infiltration in Tumor Tissue

Tumor tissue immune cell infiltration is an important indicator for evaluating the ability of the mannan-decorated lipid calcium phosphate nanoparticle vaccine to enhance the antitumor immune response by regulating the tumor microenvironment [242]. The infiltration of different types of immune cells (such as CD8+ T cells, CD4+ T cells, and natural killer cells) in tumor tissues can be quantitatively analyzed via immunohistochemical staining, flow cytometry, and other techniques. It was found that the mannan-decorated lipid calcium phosphate nanoparticle vaccine can significantly increase the amount of CD8+ T-cell infiltration in tumor tissues, improve the ratio of CD8+/CD4+ T cells, and promote the transformation of the tumor immune microenvironment [243,244,245]. In addition, the vaccine can also effectively increase the degree of invasion of natural killer cells, thereby enhancing the clearance of tumor cells [246]. An analysis of tumor immune cell infiltration showed that a mannan-decorated lipid calcium phosphate nanoparticle vaccine could significantly regulate the tumor microenvironment and enhance the antitumor immune response.

5. Promising Research Prospects for Preclinical Research

As a novel tumor immunotherapy strategy, the use of a mannan-decorated lipid calcium phosphate nanoparticle vaccine has shown great potential in preclinical studies [247,248,249,250]. Through in-depth investigation of its mechanism of action, we found that the vaccine can effectively regulate the tumor microenvironment and enhance the antitumor immune response of the body. Previous studies [251,252,253] have shown that mannan-decorated nanoparticles can promote the uptake and endocytosis of tumor cells through specific targeting, thereby improving the efficiency of antigen delivery and activating the activity of tumor-associated antigen-specific T cells. In addition, the vaccine can also induce immune cells in the tumor microenvironment, such as plasma cells and dendritic cells, to release proinflammatory factors and inhibit the function of immunosuppressive cells, thereby promoting the activation and expansion of T cells, enhancing the killing ability of cytotoxic T lymphocytes and ultimately realizing the effective elimination of tumors.

6. Discussion

In future studies, we can further optimize the formulation and preparation process of a mannan-decorated lipid calcium phosphate nanoparticle vaccine to improve its stability and bioavailability in vivo, thereby enhancing its antitumor immunotherapy effect [254]. In addition, vaccines could be explored in combination with other tumor therapies, such as chemotherapy, radiotherapy, and immune checkpoint inhibitors, to achieve better therapeutic outcomes. In addition, it is possible to design personalized treatment regimens for different types and stages of tumors and verify their safety and efficacy through preclinical and clinical studies. In general, mannan-decorated lipid calcium phosphate nanoparticle vaccines have broad application prospects in the field of tumor immunotherapy and are expected to become an important strategy for tumor therapy in the future.
In comparison with existing therapies and the literature, mannan-decorated lipid calcium–phosphorus nanoparticle vaccines show significant advantages and unique mechanisms of action [255]. Traditional cancer treatments such as surgery, radiation, and chemotherapy, although effective in some cases, are often accompanied by a higher risk of side-effects and tumor recurrence. In addition, although many immunotherapies have demonstrated effectiveness against specific cancer types, their universality and targeting are still insufficient, and they are prone to immune escape and adverse reactions. In contrast, mannan-decorated nanoparticle vaccines achieve more efficient antigen delivery and specific immune responses by precisely regulating the tumor microenvironment, resulting in more durable and intense antitumor effects in mouse models. A variety of nanoparticle vaccines in the existing literature have shown some antitumor potential, but most of them lack the regulatory ability to target the tumor microenvironment, which plays a key role in tumor growth and immune escape. The nanoparticle vaccine in this review not only improves tumor targeting and immune cell recognition through mannose modification but also significantly enhances the effect of the antitumor immune response by regulating the activity of immune cells in the tumor microenvironment, inhibiting the tumor-related inflammatory response and reducing the immunosuppressive mechanism.

7. Conclusions

MK-modified NPs can effectively regulate the tumor microenvironment, inhibit tumor growth, and enhance the infiltration of immune cells. The mannan-decorated lipid calcium phosphate nanoparticle vaccine showed good potential for regulating the tumor microenvironment, promoting immune cell infiltration, and inducing antibody and T-cell responses, thus providing new ideas and strategies for tumor immunotherapy.

Author Contributions

L.W. analyzed the data and wrote this paper; J.Y. designed and guided the research; X.Q., W.H., L.Y. and S.W. collected and downloaded the data used in our research. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the China Scholarship Council (CSC NO. 202406210298), the Scientific Research Project of the Education Department of Anhui Province (YJS20210324), the Research and Development of Intelligent Surgical Navigation and Operating System for Precise Liver Resection (2022ZLA006), and the Start-up Fund for Talent Researchers of Tsinghua University (10001020507).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Huang, J.L.; Chen, H.Z.; Gao, X.L. Lipid-coated calcium phosphate nanoparticle and beyond: A versatile platform for drug delivery. J. Drug Target. 2018, 26, 398–406. [Google Scholar] [CrossRef]
  2. Haynes, M.T.; Huang, L. Lipid-coated calcium phosphate nanoparticles for nonviral gene therapy. Adv. Genet. 2014, 88, 205–229. [Google Scholar] [CrossRef] [PubMed]
  3. Shen, Y.; Ma, H. Oridonin-loaded lipid-coated calcium phosphate nanoparticles: Preparation, characterization, and application in A549 lung cancer. Pharm. Dev. Technol. 2022, 27, 598–605. [Google Scholar] [CrossRef]
  4. Favarin, B.Z.; Bolean, M.; Ramos, A.P.; Magrini, A.; Rosato, N.; Millán, J.L.; Bottini, M.; Costa-Filho, A.J.; Ciancaglini, P. Lipid composition modulates ATP hydrolysis and calcium phosphate mineral propagation by TNAP-harboring proteoliposomes. Arch. Biochem. Biophys. 2020, 691, 108482. [Google Scholar] [CrossRef]
  5. Satterlee, A.B.; Huang, L. Current and Future Theranostic Applications of the Lipid-Calcium-Phosphate Nanoparticle Platform. Theranostics 2016, 6, 918–929. [Google Scholar] [CrossRef] [PubMed]
  6. Zhou, C.; Yu, B.; Yang, X.; Huo, T.; Lee, L.J.; Barth, R.F.; Lee, R.J. Lipid-coated nano-calcium-phosphate (LNCP) for gene delivery. Int. J. Pharm. 2010, 392, 201–208. [Google Scholar] [CrossRef] [PubMed]
  7. Dong, K.; Zhang, Y.; Ji, H.R.; Guan, Z.L.; Wang, D.Y.; Guo, Z.Y.; Deng, S.J.; He, B.Y.; Xing, J.F.; You, C.Y. Dexamethasone-Loaded Lipid Calcium Phosphate Nanoparticles Treat Experimental Colitis by Regulating Macrophage Polarization in Inflammatory Sites. Int. J. Nanomed. 2024, 19, 993–1016. [Google Scholar] [CrossRef]
  8. Cruz MA, E.; Ferreira, C.R.; Tovani, C.B.; de Oliveira, F.A.; Bolean, M.; Caseli, L.; Mebarek, S.; Millán, J.L.; Buchet, R.; Bottini, M.; et al. Phosphatidylserine controls calcium phosphate nucleation and growth on lipid monolayers: A physicochemical understanding of matrix vesicle-driven biomineralization. J. Struct. Biol. 2020, 212, 107607. [Google Scholar] [CrossRef] [PubMed]
  9. Tang, J.; Li, B.; Howard, C.B.; Mahler, S.M.; Thurecht, K.J.; Wu, Y.; Huang, L.; Xu, Z.P. Multifunctional lipid-coated calcium phosphate nanoplatforms for complete inhibition of large triple negative breast cancer via targeted combined therapy. Biomaterials 2019, 216, 119232. [Google Scholar] [CrossRef] [PubMed]
  10. Li, J.; Chen, Y.C.; Tseng, Y.C.; Mozumdar, S.; Huang, L. Biodegradable calcium phosphate nanoparticle with lipid coating for systemic siRNA delivery. J. Control. Release Off. J. Control. Release Soc. 2010, 142, 416–421. [Google Scholar] [CrossRef]
  11. Zhang, J.; Zhang, H.; Jiang, J.; Cui, N.; Xue, X.; Wang, T.; Wang, X.; He, Y.; Wang, D. Doxorubicin-Loaded Carbon Dots Lipid-Coated Calcium Phosphate Nanoparticles for Visual Targeted Delivery and Therapy of Tumor. Int. J. Nanomed. 2020, 15, 433–444. [Google Scholar] [CrossRef] [PubMed]
  12. Wu, C.; Xu, J.; Hao, Y.; Zhao, Y.; Qiu, Y.; Jiang, J.; Yu, T.; Ji, P.; Liu, Y. Application of a lipid-coated hollow calcium phosphate nanoparticle in synergistic co-delivery of doxorubicin and paclitaxel for the treatment of human lung cancer A549 cells. Int. J. Nanomed. 2017, 12, 7979–7992. [Google Scholar] [CrossRef] [PubMed]
  13. Oyane, A.; Wang, X.; Sogo, Y.; Ito, A.; Tsurushima, H. Calcium phosphate composite layers for surface-mediated gene transfer. Acta Biomaterialia 2012, 8, 2034–2046. [Google Scholar] [CrossRef] [PubMed]
  14. Liu, H.; Zhang, H.; Yin, N.; Zhang, Y.; Gou, J.; Yin, T.; He, H.; Ding, H.; Zhang, Y.; Tang, X. Sialic acid-modified dexamethasone lipid calcium phosphate gel core nanoparticles for target treatment of kidney injury. Biomater. Sci. 2020, 8, 3871–3884. [Google Scholar] [CrossRef]
  15. Li, J.; Yang, Y.; Huang, L. Calcium phosphate nanoparticles with an asymmetric lipid bilayer coating for siRNA delivery to the tumor. J. Control. Release Off. J. Control. Release Soc. 2012, 158, 108–114. [Google Scholar] [CrossRef] [PubMed]
  16. Tseng, Y.C.; Xu, Z.; Guley, K.; Yuan, H.; Huang, L. Lipid-calcium phosphate nanoparticles for delivery to the lymphatic system and SPECT/CT imaging of lymph node metastases. Biomaterials 2014, 35, 4688–4698. [Google Scholar] [CrossRef]
  17. Sethuraman, V.; Janakiraman, K.; Krishnaswami, V.; Natesan, S.; Kandasamy, R. In vivo synergistic anti-tumor effect of lumefantrine combined with pH responsive behavior of nano calcium phosphate based lipid nanoparticles on lung cancer. Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci. 2021, 158, 105657. [Google Scholar] [CrossRef]
  18. Dolci, L.S.; Panzavolta, S.; Albertini, B.; Campisi, B.; Gandolfi, M.; Bigi, A.; Passerini, N. Spray-congealed solid lipid microparticles as a new tool for the controlled release of bisphosphonates from a calcium phosphate bone cement. Eur. J. Pharm. Biopharm. Off. J. Arbeitsgemeinschaft Fur Pharm. Verfahrenstechnik E.V 2018, 122, 6–16. [Google Scholar] [CrossRef]
  19. Tanaka, Y.; Schroit, A.J. Calcium/phosphate-induced immobilization of fluorescent phosphatidylserine in synthetic bilayer membranes: Inhibition of lipid transfer between vesicles. Biochemistry 1986, 25, 2141–2148. [Google Scholar] [CrossRef]
  20. Wang, X.; Zhang, M.; Zhang, L.; Li, L.; Li, S.; Wang, C.; Su, Z.; Yuan, Y.; Pan, W. Designed Synthesis of Lipid-Coated Polyacrylic Acid/Calcium Phosphate Nanoparticles as Dual pH-Responsive Drug-Delivery Vehicles for Cancer Chemotherapy. Chemistry 2017, 23, 6586–6595. [Google Scholar] [CrossRef]
  21. Genge, B.R.; Wu, L.N.; Wuthier, R.E. Mineralization of annexin-5-containing lipid-calcium-phosphate complexes: Modulation by varying lipid composition and incubation with cartilage collagens. J. Biol. Chem. 2008, 283, 9737–9748. [Google Scholar] [CrossRef]
  22. Guo, W.; Morrisett, J.D.; Lawrie, G.M.; DeBakey, M.E.; Hamilton, J.A. Identification of different lipid phases and calcium phosphate deposits in human carotid artery plaques by MAS NMR spectroscopy. Magn. Reson. Med. 1998, 39, 184–189. [Google Scholar] [CrossRef]
  23. Liu, Y.; Hu, Y.; Huang, L. Influence of polyethylene glycol density and surface lipid on pharmacokinetics and biodistribution of lipid-calcium-phosphate nanoparticles. Biomaterials 2014, 35, 3027–3034. [Google Scholar] [CrossRef]
  24. Tang, J.; Howard, C.B.; Mahler, S.M.; Thurecht, K.J.; Huang, L.; Xu, Z.P. Enhanced delivery of siRNA to triple negative breast cancer cells in vitro and in vivo through functionalizing lipid-coated calcium phosphate nanoparticles with dual target ligands. Nanoscale 2018, 10, 4258–4266. [Google Scholar] [CrossRef] [PubMed]
  25. Skrtic, D.; Eanes, E.D. Membrane-mediated precipitation of calcium phosphate in model liposomes with matrix vesicle-like lipid composition. Bone Miner. 1992, 16, 109–119. [Google Scholar] [CrossRef]
  26. Claudio, T. Stable expression of heterologous multisubunit protein complexes established by calcium phosphate- or lipid-mediated cotransfection. Methods Enzymol. 1992, 207, 391–408. [Google Scholar] [CrossRef] [PubMed]
  27. Dolci, L.S.; Panzavolta, S.; Torricelli, P.; Albertini, B.; Sicuro, L.; Fini, M.; Bigi, A.; Passerini, N. Modulation of Alendronate release from a calcium phosphate bone cement: An in vitro osteoblast-osteoclast co-culture study. Int. J. Pharm. 2019, 554, 245–255. [Google Scholar] [CrossRef]
  28. Chen, J.; Gao, P.; Yuan, S.; Li, R.; Ni, A.; Chu, L.; Ding, L.; Sun, Y.; Liu, X.Y.; Duan, Y. Oncolytic Adenovirus Complexes Coated with Lipids and Calcium Phosphate for Cancer Gene Therapy. ACS Nano 2016, 10, 11548–11560. [Google Scholar] [CrossRef]
  29. Kashiwa, H.K.; Mukai, C.D. Lipid-calcium-phosphate spherules in chondrocytes of developing long bones. Clin. Orthop. Relat. Res. 1971, 78, 223–229. [Google Scholar] [CrossRef]
  30. Sethuraman, V.; Janakiraman, K.; Krishnaswami, V.; Natesan, S.; Kandasamy, R. pH responsive delivery of lumefantrine with calcium phosphate nanoparticles loaded lipidic cubosomes for the site specific treatment of lung cancer. Chem. Phys. Lipids 2019, 224, 104763. [Google Scholar] [CrossRef]
  31. Ke, C.H.; Chiu, Y.H.; Huang, K.C.; Lin, C.S. Exposure of Immunogenic Tumor Antigens in Surrendered Immunity and the Significance of Autologous Tumor Cell-Based Vaccination in Precision Medicine. Int. J. Mol. Sci. 2022, 24, 147. [Google Scholar] [CrossRef] [PubMed]
  32. Ramirez-Valdez, R.A.; Baharom, F.; Khalilnezhad, A.; Fussell, S.C.; Hermans, D.J.; Schrager, A.M.; Tobin KK, S.; Lynn, G.M.; Khalilnezhad, S.; Ginhoux, F.; et al. Intravenous heterologous prime-boost vaccination activates innate and adaptive immunity to promote tumor regression. Cell Rep. 2023, 42, 112599. [Google Scholar] [CrossRef]
  33. Jeong, M.; Kim, H.; Yoon, J.; Kim, D.H.; Park, J.H. Coimmunomodulation of tumor and tumor-draining lymph nodes during in situ vaccination promotes antitumor immunity. JCI Insight 2022, 7, e146608. [Google Scholar] [CrossRef] [PubMed]
  34. Mehdizadeh, R.; Shariatpanahi, S.P.; Goliaei, B.; Rüegg, C. Targeting myeloid-derived suppressor cells in combination with tumor cell vaccination predicts anti-tumor immunity and breast cancer dormancy: An in silico experiment. Sci. Rep. 2023, 13, 5875. [Google Scholar] [CrossRef] [PubMed]
  35. Medina-Echeverz, J.; Hinterberger, M.; Testori, M.; Geiger, M.; Giessel, R.; Bathke, B.; Kassub, R.; Gräbnitz, F.; Fiore, G.; Wennier, S.T.; et al. Synergistic cancer immunotherapy combines MVA-CD40L induced innate and adaptive immunity with tumor targeting antibodies. Nat. Commun. 2019, 10, 5041. [Google Scholar] [CrossRef]
  36. Clark, P.A.; Sriramaneni, R.N.; Bates, A.M.; Jin, W.J.; Jagodinsky, J.C.; Hernandez, R.; Le, T.; Jeffery, J.J.; Marsh, I.R.; Grudzinski, J.J.; et al. Low-Dose Radiation Potentiates the Propagation of Anti-Tumor Immunity against Melanoma Tumor in the Brain after In Situ Vaccination at a Tumor outside the Brain. Radiat. Res. 2021, 195, 522–540. [Google Scholar] [CrossRef]
  37. Kim, N.J.; Yoon, J.H.; Tuomi, A.C.; Lee, J.; Kim, D. In-situ tumor vaccination by percutaneous ablative therapy and its synergy with immunotherapeutics: An update on combination therapy. Front. Immunol. 2023, 14, 1118845. [Google Scholar] [CrossRef] [PubMed]
  38. Varypataki, E.M.; Hasler, F.; Waeckerle-Men, Y.; Vogel-Kindgen, S.; Høgset, A.; Kündig, T.M.; Gander, B.; Halin, C.; Johansen, P. Combined Photosensitization and Vaccination Enable CD8 T-Cell Immunity and Tumor Suppression Independent of CD4 T-Cell Help. Front. Immunol. 2019, 10, 1548. [Google Scholar] [CrossRef]
  39. Dong, W.; Du, J.; Shen, H.; Gao, D.; Li, Z.; Wang, G.; Mu, X.; Liu, Q. Administration of embryonic stem cells generates effective antitumor immunity in mice with minor and heavy tumor load. Cancer Immunol. Immunother. CII 2010, 59, 1697–1705. [Google Scholar] [CrossRef]
  40. Koido, S.; Ito, M.; Sagawa, Y.; Okamoto, M.; Hayashi, K.; Nagasaki, E.; Kan, S.; Komita, H.; Kamata, Y.; Homma, S. Vaccination with vascular progenitor cells derived from induced pluripotent stem cells elicits antitumor immunity targeting vascular and tumor cells. Cancer Immunol. Immunother. CII 2014, 63, 459–468. [Google Scholar] [CrossRef]
  41. Accolla, R.S.; Tosi, G. Adequate antigen availability: A key issue for novel approaches to tumor vaccination and tumor immunotherapy. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2013, 8, 28–36. [Google Scholar] [CrossRef]
  42. Tong, W.; Maira, M.; Roychoudhury, R.; Galan, A.; Brahimi, F.; Gilbert, M.; Cunningham, A.M.; Josephy, S.; Pirvulescu, I.; Moffett, S.; et al. Vaccination with Tumor-Ganglioside Glycomimetics Activates a Selective Immunity that Affords Cancer Therapy. Cell Chem. Biol. 2019, 26, 1013–1026.e4. [Google Scholar] [CrossRef] [PubMed]
  43. Park, J.; Hsueh, P.C.; Li, Z.; Ho, P.C. Microenvironment-driven metabolic adaptations guiding CD8+ T cell anti-tumor immunity. Immunity 2023, 56, 32–42. [Google Scholar] [CrossRef]
  44. Castro Dopico, X.; Ols, S.; Loré, K.; Karlsson Hedestam, G.B. Immunity to SARS-CoV-2 induced by infection or vaccination. J. Intern. Med. 2022, 291, 32–50. [Google Scholar] [CrossRef]
  45. Bevers, S.; Kooijmans, S.A.A.; Van de Velde, E.; Evers, M.J.W.; Seghers, S.; Gitz-Francois, J.J.J.M.; van Kronenburg, N.C.H.; Fens, M.H.A.M.; Mastrobattista, E.; Hassler, L.; et al. mRNA-LNP vaccines tuned for systemic immunization induce strong antitumor immunity by engaging splenic immune cells. Mol. Ther. J. Am. Soc. Gene Ther. 2022, 30, 3078–3094. [Google Scholar] [CrossRef]
  46. Schetters, S.T.T.; Li, R.J.E.; Kruijssen, L.J.W.; Engels, S.; Ambrosini, M.; Garcia-Vallejo, J.J.; Kalay, H.; Unger, W.W.J.; van Kooyk, Y. Adaptable antigen matrix platforms for peptide vaccination strategies and T cell-mediated anti-tumor immunity. Biomaterials 2020, 262, 120342. [Google Scholar] [CrossRef]
  47. Bruni, L.; Saura-Lázaro, A.; Montoliu, A.; Brotons, M.; Alemany, L.; Diallo, M.S.; Afsar, O.Z.; LaMontagne, D.S.; Mosina, L.; Contreras, M.; et al. HPV vaccination introduction worldwide and WHO and UNICEF estimates of national HPV immunization coverage 2010–2019. Prev. Med. 2021, 144, 106399. [Google Scholar] [CrossRef]
  48. Li, S.; Zhang, Q.; Bai, H.; Huang, W.; Shu, C.; Ye, C.; Sun, W.; Ma, Y. Self-Assembled Nanofibers Elicit Potent HPV16 E7-Specific Cellular Immunity And Abolish Established TC-1 Graft Tumor. Int. J. Nanomed. 2019, 14, 8209–8219. [Google Scholar] [CrossRef]
  49. Dolina, J.S.; Lee, J.; Brightman, S.E.; McArdle, S.; Hall, S.M.; Thota, R.R.; Zavala, K.S.; Lanka, M.; Ramamoorthy Premlal, A.L.; Greenbaum, J.A.; et al. Linked CD4+/CD8+ T cell neoantigen vaccination overcomes immune checkpoint blockade resistance and enables tumor regression. J. Clin. Investig. 2023, 133, e164258. [Google Scholar] [CrossRef] [PubMed]
  50. Kim, Y.; Lee, S.; Jon, S. Liposomal Delivery of an Immunostimulatory CpG Induces Robust Antitumor Immunity and Long-Term Immune Memory by Reprogramming Tumor-Associated Macrophages. Adv. Healthc. Mater. 2024, 13, e2300549. [Google Scholar] [CrossRef]
  51. Carter, J.A.; Matta, B.; Battaglia, J.; Somerville, C.; Harris, B.D.; LaPan, M.; Atwal, G.S.; Barnes, B.J. Identification of pan-cancer/testis genes and validation of therapeutic targeting in triple-negative breast cancer: Lin28a-based and Siglece-based vaccination induces antitumor immunity and inhibits metastasis. J. Immunother. Cancer 2023, 11, e007935. [Google Scholar] [CrossRef] [PubMed]
  52. Liu, Y.; Li, H.; Zhao, H.; Hao, Y.; Van Herck, S.; Xu, Z.; Wang, G.; Wang, X.; Zhang, X.; Ge, X.; et al. In Situ Tumor Vaccination with Calcium-Linked Degradable Coacervate Nanocomplex Co-Delivering Photosensitizer and TLR7/8 Agonist to Trigger Effective Anti-Tumor Immune Responses. Adv. Healthc. Mater. 2022, 11, e2102781. [Google Scholar] [CrossRef]
  53. Wang, H.; Gan, M.; Wu, B.; Zeng, R.; Wang, Z.; Xu, J.; Li, J.; Zhang, Y.; Cao, J.; Chen, L.; et al. Humoral and cellular immunity of two-dose inactivated COVID-19 vaccination in Chinese children: A prospective cohort study. J. Med. Virol. 2023, 95, e28380. [Google Scholar] [CrossRef]
  54. Zhao, X.; Zhang, J.; Chen, B.; Ding, X.; Zhao, N.; Xu, F.J. Rough Nanovaccines Boost Antitumor Immunity Through the Enhancement of Vaccination Cascade and Immunogenic Cell Death Induction. Small Methods 2023, 7, e2201595. [Google Scholar] [CrossRef]
  55. Luo, X.; Qiu, Y.; Dinesh, P.; Gong, W.; Jiang, L.; Feng, X.; Li, J.; Jiang, Y.; Lei, Y.L.; Chen, Q. The functions of autophagy at the tumour-immune interface. J. Cell. Mol. Med. 2021, 25, 2333–2341. [Google Scholar] [CrossRef] [PubMed]
  56. Perciani, C.T.; Liu, L.Y.; Wood, L.; MacParland, S.A. Enhancing Immunity with Nanomedicine: Employing Nanoparticles to Harness the Immune System. ACS Nano 2021, 15, 7–20. [Google Scholar] [CrossRef]
  57. Abascal, J.; Oh, M.S.; Liclican, E.L.; Dubinett, S.M.; Salehi-Rad, R.; Liu, B. Dendritic Cell Vaccination in Non-Small Cell Lung Cancer: Remodeling the Tumor Immune Microenvironment. Cells 2023, 12, 2404. [Google Scholar] [CrossRef] [PubMed]
  58. McAuliffe, J.; Chan, H.F.; Noblecourt, L.; Ramirez-Valdez, R.A.; Pereira-Almeida, V.; Zhou, Y.; Pollock, E.; Cappuccini, F.; Redchenko, I.; Hill, A.V.; et al. Heterologous prime-boost vaccination targeting MAGE-type antigens promotes tumor T-cell infiltration and improves checkpoint blockade therapy. J. Immunother. Cancer 2021, 9, e003218. [Google Scholar] [CrossRef] [PubMed]
  59. Morera-Díaz, Y.; Canaán-Haden, C.; Sánchez-Ramírez, J.; Bequet-Romero, M.; Gonzalez-Moya, I.; Martínez, R.; Falcón, V.; Palenzuela, D.; Ayala-Ávila, M.; Gavilondo, J.V. Active immunization with a structurally aggregated PD-L1 antigen breaks T and B immune tolerance in non-human primates and exhibits in vivo anti-tumoral effects in immunocompetent mouse tumor models. Cancer Lett. 2023, 561, 216156. [Google Scholar] [CrossRef]
  60. Femel, J.; van Hooren, L.; Herre, M.; Cedervall, J.; Saupe, F.; Huijbers EJ, M.; Verboogen DR, J.; Reichel, M.; Thijssen, V.L.; Griffioen, A.W.; et al. Vaccination against galectin-1 promotes cytotoxic T-cell infiltration in melanoma and reduces tumor burden. Cancer Immunol. Immunother. CII 2022, 71, 2029–2040. [Google Scholar] [CrossRef]
  61. Ishio, T.; Tsukamoto, S.; Yokoyama, E.; Izumiyama, K.; Saito, M.; Muraki, H.; Kobayashi, M.; Mori, A.; Morioka, M.; Kondo, T. Anti-CD20 antibodies and bendamustine attenuate humoral immunity to COVID-19 vaccination in patients with B-cell non-Hodgkin lymphoma. Ann. Hematol. 2023, 102, 1421–1431. [Google Scholar] [CrossRef]
  62. Kershner, L.J.; Choi, K.; Wu, J.; Zhang, X.; Perrino, M.; Salomonis, N.; Shern, J.F.; Ratner, N. Multiple Nf1 Schwann cell populations reprogram the plexiform neurofibroma tumor microenvironment. JCI Insight 2022, 7, e154513. [Google Scholar] [CrossRef]
  63. Top, K.A.; Vaudry, W.; Morris, S.K.; Pham-Huy, A.; Pernica, J.M.; Tapiéro, B.; Gantt, S.; Price, V.E.; Rassekh, S.R.; Sung, L.; et al. Waning Vaccine Immunity and Vaccination Responses in Children Treated for Acute Lymphoblastic Leukemia: A Canadian Immunization Research Network Study. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2020, 71, e439–e448. [Google Scholar] [CrossRef]
  64. Scheer, V.; Goldammer, M.; Flindt, S.; van Zandbergen, G.; Hinz, T. Therapeutische Immunisierungen gegen Tumore und neurodegenerative Erkrankungen [Therapeutic vaccination for tumors and neurodegenerative diseases]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2020, 63, 1373–1379. [Google Scholar] [CrossRef]
  65. Chen, P.M.; Pan, W.Y.; Wu, C.Y.; Yeh, C.Y.; Korupalli, C.; Luo, P.K.; Chou, C.J.; Chia, W.T.; Sung, H.W. Modulation of tumor microenvironment using a TLR-7/8 agonist-loaded nanoparticle system that exerts low-temperature hyperthermia and immunotherapy for in situ cancer vaccination. Biomaterials 2020, 230, 119629. [Google Scholar] [CrossRef]
  66. Li, E.; Butkovich, N.; Tucker, J.A.; Nelson, E.L.; Wang, S.W. Evaluating Anti-tumor Immune Responses of Protein Nanoparticle-Based Cancer Vaccines. Methods Mol. Biol. 2023, 2671, 321–333. [Google Scholar] [CrossRef]
  67. Zhao, B.; Kilian, M.; Bunse, T.; Platten, M.; Bunse, L. Tumor-reactive T helper cells in the context of vaccination against glioma. Cancer Cell 2023, 41, 1829–1834. [Google Scholar] [CrossRef]
  68. Liu, G.; Zhu, M.; Zhao, X.; Nie, G. Nanotechnology-empowered vaccine delivery for enhancing CD8+ T cells-mediated cellular immunity. Adv. Drug Deliv. Rev. 2021, 176, 113889. [Google Scholar] [CrossRef]
  69. Carlson, P.M.; Patel, R.B.; Birstler, J.; Rodriquez, M.; Sun, C.; Erbe, A.K.; Bates, A.M.; Marsh, I.; Grudzinski, J.; Hernandez, R.; et al. Radiation to all macroscopic sites of tumor permits greater systemic antitumor response to in situ vaccination. J. Immunother. Cancer 2023, 11, e005463. [Google Scholar] [CrossRef]
  70. Vajari, M.K.; Sanaei, M.J.; Salari, S.; Rezvani, A.; Ravari, M.S.; Bashash, D. Breast cancer vaccination: Latest advances with an analytical focus on clinical trials. Int. Immunopharmacol. 2023, 123, 110696. [Google Scholar] [CrossRef] [PubMed]
  71. Vieira, J.F.; Peixoto, A.P.; Murta EF, C.; Michelin, M.A. Prophylactic Dendritic Cell Vaccination in Experimental Breast Cancer Controls Immunity and Hepatic Metastases. Anticancer. Res. 2021, 41, 3419–3427. [Google Scholar] [CrossRef] [PubMed]
  72. Ngamcherdtrakul, W.; Reda, M.; Nelson, M.A.; Wang, R.; Zaidan, H.Y.; Bejan, D.S.; Hoang, N.H.; Lane, R.S.; Luoh, S.W.; Leachman, S.A.; et al. In Situ Tumor Vaccination with Nanoparticle Co-Delivering CpG and STAT3 siRNA to Effectively Induce Whole-Body Antitumor Immune Response. Adv. Mater. 2021, 33, e2100628. [Google Scholar] [CrossRef]
  73. Koeken, V.A.C.M.; Qi, C.; Mourits, V.P.; de Bree, L.C.J.; Moorlag, S.J.C.F.M.; Sonawane, V.; Lemmers, H.; Dijkstra, H.; Joosten, L.A.B.; van Laarhoven, A.; et al. Plasma metabolome predicts trained immunity responses after antituberculosis BCG vaccination. PLoS Biol. 2022, 20, e3001765. [Google Scholar] [CrossRef] [PubMed]
  74. Rahdan, S.; Razavi, S.A.; Shojaeian, S.; Shokri, F.; Amiri, M.M.; Zarnani, A.H. Immunization with placenta-specific 1 (plac1) induces potent anti-tumor responses and prolongs survival in a mouse model of melanoma. Adv. Med. Sci. 2022, 67, 338–345. [Google Scholar] [CrossRef] [PubMed]
  75. Ammons, D.T.; Guth, A.; Rozental, A.J.; Kurihara, J.; Marolf, A.J.; Chow, L.; Griffin, J.F., IV; Makii, R.; MacQuiddy, B.; Boss, M.K.; et al. Reprogramming the Canine Glioma Microenvironment with Tumor Vaccination plus Oral Losartan and Propranolol Induces Objective Responses. Cancer Res. Commun. 2022, 2, 1657–1667. [Google Scholar] [CrossRef]
  76. Kostinov, M.P.; Akhmatova, N.K.; Karpocheva, S.V.; Vlasenko, A.E.; Polishchuk, V.B.; Kostinov, A.M. Vaccination Against Diphtheria and Tetanus as a Way to Activate Adaptive Immunity in Children with Solid Tumors. Front. Immunol. 2021, 12, 696816. [Google Scholar] [CrossRef] [PubMed]
  77. Corradini, P.; Agrati, C.; Apolone, G.; Mantovani, A.; Giannarelli, D.; Marasco, V.; Bordoni, V.; Sacchi, A.; Matusali, G.; Salvarani, C.; et al. Humoral and T-Cell Immune Response After 3 Doses of Messenger RNA Severe Acute Respiratory Syndrome Coronavirus 2 Vaccines in Fragile Patients: The Italian VAX4FRAIL Study. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2023, 76, e426–e438. [Google Scholar] [CrossRef]
  78. Perrino, M.R.; Ahmari, N.; Hall, A.; Jackson, M.; Na, Y.; Pundavela, J.; Szabo, S.; Woodruff, T.M.; Dombi, E.; Kim, M.O.; et al. C5aR plus MEK inhibition durably targets the tumor milieu and reveals tumor cell phagocytosis. Life Sci. Alliance 2024, 7, e202302229. [Google Scholar] [CrossRef]
  79. Schrezenmeier, E.; Rincon-Arevalo, H.; Jens, A.; Stefanski, A.L.; Hammett, C.; Osmanodja, B.; Koch, N.; Zukunft, B.; Beck, J.; Oellerich, M.; et al. Temporary antimetabolite treatment hold boosts SARS-CoV-2 vaccination-specific humoral and cellular immunity in kidney transplant recipients. JCI Insight 2022, 7, e157836. [Google Scholar] [CrossRef]
  80. Jiang, C.; Kumar, A.; Yu, Z.; Shipman, T.; Wang, Y.; McKay, R.M.; Xing, C.; Le, L.Q. Basement membrane proteins in extracellular matrix characterize NF1 neurofibroma development and response to MEK inhibitor. J. Clin. Investig. 2023, 133, e168227. [Google Scholar] [CrossRef]
  81. Xu, Y.Y.; Chen, Q.H.; Liu, Y.; Ji, C.; Du, J.; Li, M.Y.; Shen, H.P.; Zhang, X.C.; Che, X.R.; Zhao, G. Research progress of vaccination status, efficacy and safety in children with tumor. Chin. J. Prev. Med. 2024, 58, 87–91. [Google Scholar] [CrossRef]
  82. Cho, H.; Binder, J.; Weeratna, R.; Dermyer, M.; Dai, S.; Boccia, A.; Li, W.; Li, S.; Jooss, K.; Merson, J.; et al. Preclinical development of a vaccine-based immunotherapy regimen (VBIR) that induces potent and durable T cell responses to tumor-associated self-antigens. Cancer Immunol. Immunother. CII 2023, 72, 287–300. [Google Scholar] [CrossRef]
  83. Liu, H.Y.; Altman, A.; Canonigo-Balancio, A.J.; Croft, M. Experimental Melanoma Immunotherapy Model Using Tumor Vaccination with a Hematopoietic Cytokine. J. Vis. Exp. JoVE 2023, 192, e64082. [Google Scholar] [CrossRef]
  84. Flies, A.S.; Flies, E.J.; Fox, S.; Gilbert, A.; Johnson, S.R.; Liu, G.S.; Lyons, A.B.; Patchett, A.L.; Pemberton, D.; Pye, R.J. An oral bait vaccination approach for the Tasmanian devil facial tumor diseases. Expert Rev. Vaccines 2020, 19, 1–10. [Google Scholar] [CrossRef]
  85. Huang, X.; Zhang, G.; Bai, X.; Liang, T. Reviving the role of MET in liver cancer therapy and vaccination: An autophagic perspective. Oncoimmunology 2020, 9, 1818438. [Google Scholar] [CrossRef]
  86. Wan, J.; Ren, L.; Li, X.; He, S.; Fu, Y.; Xu, P.; Meng, F.; Xian, S.; Pu, K.; Wang, H. Photoactivatable nanoagonists chemically programmed for pharmacokinetic tuning and in situ cancer vaccination. Proc. Natl. Acad. Sci. USA 2023, 120, e2210385120. [Google Scholar] [CrossRef]
  87. Hosseinalizadeh, H.; Rahmati, M.; Ebrahimi, A.; O’Connor, R.S. Current Status and Challenges of Vaccination Therapy for Glioblastoma. Mol. Cancer Ther. 2023, 22, 435–446. [Google Scholar] [CrossRef]
  88. van Dam KP, J.; Volkers, A.G.; Wieske, L.; Stalman, E.W.; Kummer LY, L.; van Kempen ZL, E.; Killestein, J.; Tas, S.W.; Boekel, L.; Wolbink, G.J.; et al. Primary SARS-CoV-2 infection in patients with immune-mediated inflammatory diseases: Long-term humoral immune responses and effects on disease activity. BMC Infect. Dis. 2023, 23, 332. [Google Scholar] [CrossRef]
  89. Hall, V.G.; Teh, B.W. COVID-19 Vaccination in Patients with Cancer and Patients Receiving HSCT or CAR-T Therapy: Immune Response, Real-World Effectiveness, and Implications for the Future. J. Infect. Dis. 2023, 228, S55–S69. [Google Scholar] [CrossRef] [PubMed]
  90. Doukas, P.G.; St Pierre, F.; Karmali, R.; Mi, X.; Boyer, J.; Nieves, M.; Ison, M.G.; Winter, J.N.; Gordon, L.I.; Ma, S. Humoral Immunity After COVID-19 Vaccination in Chronic Lymphocytic Leukemia and Other Indolent Lymphomas: A Single-Center Observational Study. Oncol. 2023, 28, e930–e941. [Google Scholar] [CrossRef] [PubMed]
  91. He, Y.; Cheng, C.; Liu, Y.; Chen, F.M.; Chen, Y.; Yang, C.; Zhao, Z.; Dawulieti, J.; Shen, Z.; Zhang, Y.; et al. Intravenous Senescent Erythrocyte Vaccination Modulates Adaptive Immunity and Splenic Complement Production. ACS Nano 2024, 18, 470–482. [Google Scholar] [CrossRef] [PubMed]
  92. Hartmann, A.K.; Bartneck, J.; Pielenhofer, J.; Meiser, S.L.; Arnold-Schild, D.; Klein, M.; Stassen, M.; Schild, H.; Muth, S.; Probst, H.C.; et al. Optimized dithranol-imiquimod-based transcutaneous immunization enables tumor rejection. Front. Immunol. 2023, 14, 1238861. [Google Scholar] [CrossRef]
  93. Del Poeta, G.; Laureana, R.; Bomben, R.; Rossi, F.M.; Pozzo, F.; Zaina, E.; Cattarossi, I.; Varaschin, P.; Nanni, P.; Boschian Boschin, R.; et al. COVID-19 vaccination: Evaluation of humoral and cellular immunity after the booster dose in chronic lymphocytic leukemia patients. Hematol. Oncol. 2023, 41, 559–562. [Google Scholar] [CrossRef] [PubMed]
  94. Andorko, J.I.; Tsai, S.J.; Gammon, J.M.; Carey, S.T.; Zeng, X.; Gosselin, E.A.; Edwards, C.; Shah, S.A.; Hess, K.L.; Jewell, C.M. Spatial delivery of immune cues to lymph nodes to define therapeutic outcomes in cancer vaccination. Biomater. Sci. 2022, 10, 4612–4626. [Google Scholar] [CrossRef]
  95. Zhang, Y.; Sriramaneni, R.N.; Clark, P.A.; Jagodinsky, J.C.; Ye, M.; Jin, W.; Wang, Y.; Bates, A.; Kerr, C.P.; Le, T.; et al. Multifunctional nanoparticle potentiates the in situ vaccination effect of radiation therapy and enhances response to immune checkpoint blockade. Nat. Commun. 2022, 13, 4948. [Google Scholar] [CrossRef]
  96. Bukhari, S.I.; Jehan, F.; Belgaumi, A. Global Immunization Crisis Amid the COVID-19 Pandemic: Implications for Pediatric Oncology. JCO Glob. Oncol. 2024, 10, e2300477. [Google Scholar] [CrossRef] [PubMed]
  97. Alonso-Miguel, D.; Valdivia, G.; Guerrera, D.; Perez-Alenza, M.D.; Pantelyushin, S.; Alonso-Diez, A.; Beiss, V.; Fiering, S.; Steinmetz, N.F.; Suarez-Redondo, M.; et al. Neoadjuvant in situ vaccination with cowpea mosaic virus as a novel therapy against canine inflammatory mammary cancer. J. Immunother. Cancer 2022, 10, e004044. [Google Scholar] [CrossRef]
  98. Lam, B.; Kung, Y.J.; Lin, J.; Tseng, S.H.; Tu, H.F.; Huang, C.; Lee, B.; Velarde, E.; Tsai, Y.C.; Villasmil, R.; et al. In situ vaccination via tissue-targeted cDC1 expansion enhances the immunogenicity of chemoradiation and immunotherapy. J. Clin. Investig. 2024, 134, e171621. [Google Scholar] [CrossRef]
  99. Eini, L.; Naseri, M.; Karimi-Busheri, F.; Bozorgmehr, M.; Ghods, R.; Madjd, Z. Preventive cancer stem cell-based vaccination modulates tumor development in syngeneic colon adenocarcinoma murine model. J. Cancer Res. Clin. Oncol. 2023, 149, 4101–4116. [Google Scholar] [CrossRef]
  100. Peng, S.; Chen, S.; Hu, W.; Mei, J.; Zeng, X.; Su, T.; Wang, W.; Chen, Z.; Xiao, H.; Zhou, Q.; et al. Combination Neoantigen-Based Dendritic Cell Vaccination and Adoptive T-Cell Transfer Induces Antitumor Responses Against Recurrence of Hepatocellular Carcinoma. Cancer Immunol. Res. 2022, 10, 728–744. [Google Scholar] [CrossRef]
  101. Zandvakili, R.; Basirjafar, P.; Masoumi, J.; Zainodini, N.; Taghipour, Z.; Khorramdelazad, H.; Yousefi, S.; Tavakoli, T.; Safdel, S.; Gheitasi, M.; et al. Vaccination with celecoxib-treated dendritic cells improved cellular immune responses in an animal breast cancer model. Adv. Med. Sci. 2023, 68, 157–168. [Google Scholar] [CrossRef]
  102. Shou, J.; Mo, F.; Zhang, S.; Lu, L.; Han, N.; Liu, L.; Qiu, M.; Li, H.; Han, W.; Ma, D.; et al. Combination treatment of radiofrequency ablation and peptide neoantigen vaccination: Promising modality for future cancer immunotherapy. Front. Immunol. 2022, 13, 1000681. [Google Scholar] [CrossRef]
  103. Niavarani, S.R.; St-Cyr, G.; Daniel, L.; Lawson, C.; Giguère, H.; Alkayyal, A.A.; Tai, L.H. Heterologous prime-boost cellular vaccination induces potent antitumor immunity against triple negative breast cancer. Front. Immunol. 2023, 14, 1098344. [Google Scholar] [CrossRef]
  104. Domingos-Pereira, S.; Roh, V.; Hiou-Feige, A.; Galliverti, G.; Simon, C.; Tolstonog, G.V.; Nardelli-Haefliger, D. Vaccination with a nanoparticle E7 vaccine can prevent tumor recurrence following surgery in a human papillomavirus head and neck cancer model. Oncoimmunology 2021, 10, 1912473. [Google Scholar] [CrossRef]
  105. Sunil, V.; Mozhi, A.; Zhan, W.; Teoh, J.H.; Ghode, P.B.; Thakor, N.V.; Wang, C.H. In-situ vaccination using dual responsive organelle targeted nanoreactors. Biomaterials 2022, 290, 121843. [Google Scholar] [CrossRef]
  106. Ellingsen, E.B.; O’Day, S.; Mezheyeuski, A.; Gromadka, A.; Clancy, T.; Kristedja, T.S.; Milhem, M.; Zakharia, Y. Clinical Activity of Combined Telomerase Vaccination and Pembrolizumab in Advanced Melanoma: Results from a Phase I Trial. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2023, 29, 3026–3036. [Google Scholar] [CrossRef]
  107. Repáraz, D.; Ruiz, M.; Silva, L.; Aparicio, B.; Egea, J.; Guruceaga, E.; Ajona, D.; Senent, Y.; Conde, E.; Navarro, F.; et al. Gemcitabine-mediated depletion of immunosuppressive dendritic cells enhances the efficacy of therapeutic vaccination. Front. Immunol. 2022, 13, 991311. [Google Scholar] [CrossRef]
  108. Ghanaat, M.; Kaboosi, H.; Negahdari, B.; Fattahi, E.; Malekshahi, Z.V. Heterologous Prime-boost Vaccination Using Adenovirus and Albumin Nanoparticles as Carriers for Human Papillomavirus 16 E7 Epitope. Curr. Pharm. Biotechnol. 2023, 24, 1195–1203. [Google Scholar] [CrossRef]
  109. Clark, P.A.; Sriramaneni, R.N.; Jin, W.J.; Jagodinsky, J.C.; Bates, A.M.; Jaquish, A.A.; Anderson, B.R.; Le, T.; Lubin, J.A.; Chakravarty, I.; et al. In situ vaccination at a peripheral tumor site augments response against melanoma brain metastases. J. Immunother. Cancer 2020, 8, e000809. [Google Scholar] [CrossRef]
  110. Mair, M.J.; Berger, J.M.; Berghoff, A.S.; Starzer, A.M.; Ortmayr, G.; Puhr, H.C.; Steindl, A.; Perkmann, T.; Haslacher, H.; Strassl, R.; et al. Humoral Immune Response in Hematooncological Patients and Health Care Workers Who Received SARS-CoV-2 Vaccinations. JAMA Oncol. 2022, 8, 106–113. [Google Scholar] [CrossRef]
  111. Son, H.Y.; Jeong, H.K.; Apostolopoulos, V.; Kim, C.W. MUC1 expressing tumor growth was retarded after human mucin 1 (MUC1) plasmid DNA immunization. Int. J. Immunopathol. Pharmacol. 2022, 36, 3946320221112358. [Google Scholar] [CrossRef] [PubMed]
  112. Shin, H.; Na, K. Cancer-Targetable pH-Sensitive Zinc-Based Immunomodulators Combined with Photodynamic Therapy for in Situ Vaccination. ACS Biomater. Sci. Eng. 2020, 6, 3430–3439. [Google Scholar] [CrossRef] [PubMed]
  113. Pol, J.G.; Bridle, B.W.; Lichty, B.D. Detection of Tumor Antigen-Specific T-Cell Responses After Oncolytic Vaccination. Methods Mol. Biol. 2020, 2058, 191–211. [Google Scholar] [CrossRef] [PubMed]
  114. Friedrich, R.E.; Nörnberg LK, N.; Hagel, C. Peripheral Nerve Sheath Tumors in Patients with Neurofibromatosis Type 1: Morphological and Immunohistochemical Study. Anticancer. Res. 2022, 42, 1247–1261. [Google Scholar] [CrossRef] [PubMed]
  115. He, T.; Shi, Y.; Kou, X.; Shen, M.; Liang, X.; Li, X.; Wu, R.; You, Y.; Wu, Q.; Gong, C. Antigenicity and adjuvanticity co-reinforced personalized cell vaccines based on self-adjuvanted hydrogel for post-surgical cancer vaccination. Biomaterials 2023, 301, 122218. [Google Scholar] [CrossRef] [PubMed]
  116. Figueiredo, J.C.; Merin, N.M.; Hamid, O.; Choi, S.Y.; Lemos, T.; Cozen, W.; Nguyen, N.; Finster, L.J.; Foley, J.; Darrah, J.; et al. Longitudinal SARS-CoV-2 mRNA Vaccine-Induced Humoral Immune Responses in Patients with Cancer. Cancer Res. 2021, 81, 6273–6280. [Google Scholar] [CrossRef] [PubMed]
  117. Bakhtadze, S.; Lim, M.; Craiu, D.; Cazacu, C. Vaccination in acute immune-mediated/inflammatory disorders of the central nervous system. Eur. J. Paediatr. Neurol. EJPN Off. J. Eur. Paediatr. Neurol. Soc. 2021, 34, 118–122. [Google Scholar] [CrossRef] [PubMed]
  118. Radbruch, A.; Melchers, F. Warum die Regeneration von immunologischer Toleranz durch Impfen schwierig ist [Why the regeneration of immunological tolerance by vaccination is difficult]. Z. Fur Rheumatol. 2024, 83, 105–111. [Google Scholar] [CrossRef] [PubMed]
  119. Li, Y.; Luo, Y.; Hou, L.; Huang, Z.; Wang, Y.; Zhou, S. Antigen-Capturing Dendritic-Cell-Targeting Nanoparticles for Enhanced Tumor Immunotherapy Based on Photothermal-Therapy-Induced In Situ Vaccination. Adv. Healthc. Mater. 2023, 12, e2202871. [Google Scholar] [CrossRef]
  120. Liu, X.; Su, Q.; Song, H.; Shi, X.; Zhang, Y.; Zhang, C.; Huang, P.; Dong, A.; Kong, D.; Wang, W. PolyTLR7/8a-conjugated, antigen-trapping gold nanorods elicit anticancer immunity against abscopal tumors by photothermal therapy-induced in situ vaccination. Biomaterials 2021, 275, 120921. [Google Scholar] [CrossRef]
  121. Patenaude, R.; Yasmin-Karim, S.; Peng, Y.; Wucherpfennig, K.W.; Ngwa, W.; Kheir, J.N.; Polizzotti, B.D. Injectable Oxygen Microparticles Boost Radiation-Mediated In Situ Vaccination and Systemic Antitumor Immune Responses. Int. J. Radiat. Oncol. Biol. Phys. 2023, 116, 906–915. [Google Scholar] [CrossRef]
  122. Salewski, I.; Gladbach, Y.S.; Kuntoff, S.; Irmscher, N.; Hahn, O.; Junghanss, C.; Maletzki, C. In vivo vaccination with cell line-derived whole tumor lysates: Neoantigen quality, not quantity matters. J. Transl. Med. 2020, 18, 402. [Google Scholar] [CrossRef]
  123. Stegmann, T.; Wiekmeijer, A.S.; Kwappenberg, K.; van Duikeren, S.; Bhoelan, F.; Bemelman, D.; Beenakker, T.J.M.; Krebber, W.J.; Arens, R.; Melief, C.J.M. Enhanced HPV16 E6/E7+ tumor eradication via induction of tumor-specific T cells by therapeutic vaccination with virosomes presenting synthetic long peptides. Cancer Immunol. Immunother. CII 2023, 72, 2851–2864. [Google Scholar] [CrossRef]
  124. Jackson, K.; Samaddar, S.; Markiewicz, M.A.; Bansal, A. Vaccination-Based Immunoprevention of Colorectal Tumors: A Primer for the Clinician. J. Clin. Gastroenterol. 2023, 57, 246–252. [Google Scholar] [CrossRef] [PubMed]
  125. Trabbic, K.R.; Whalen, K.; Abarca-Heideman, K.; Xia, L.; Temme, J.S.; Edmondson, E.F.; Gildersleeve, J.C.; Barchi, J.J., Jr. A Tumor-Selective Monoclonal Antibody from Immunization with a Tumor-Associated Mucin Glycopeptide. Sci. Rep. 2019, 9, 5662. [Google Scholar] [CrossRef]
  126. Preusser, M.; van den Bent, M.J. Autologous tumor lysate-loaded dendritic cell vaccination (DCVax-L) in glioblastoma: Breakthrough or fata morgana? Neuro-Oncol. 2023, 25, 631–634. [Google Scholar] [CrossRef] [PubMed]
  127. Szallasi, Z.; Prosz, A.; Sztupinszki, Z.; Moldvay, J. Are tumor-associated carbohydrates the missing link between the gut microbiome and response to immune checkpoint inhibitor treatment in cancer? Oncoimmunology 2024, 13, 2324493. [Google Scholar] [CrossRef]
  128. Fan, Q.; Ma, Q.; Bai, J.; Xu, J.; Fei, Z.; Dong, Z.; Maruyama, A.; Leong, K.W.; Liu, Z.; Wang, C. An implantable blood clot-based immune niche for enhanced cancer vaccination. Sci. Adv. 2020, 6, eabb4639. [Google Scholar] [CrossRef] [PubMed]
  129. Caldera, F.; Farraye, F.A.; Necela, B.M.; Cogen, D.; Saha, S.; Wald, A.; Daoud, N.D.; Chun, K.; Grimes, I.; Lutz, M.; et al. Higher Cell-Mediated Immune Responses in Patients with Inflammatory Bowel Disease on Anti-TNF Therapy After COVID-19 Vaccination. Inflamm. Bowel Dis. 2023, 29, 1202–1209. [Google Scholar] [CrossRef]
  130. Shahgolzari, M.; Pazhouhandeh, M.; Milani, M.; Fiering, S.; Khosroushahi, A.Y. Alfalfa mosaic virus nanoparticles-based in situ vaccination induces antitumor immune responses in breast cancer model. Nanomedicine 2021, 16, 97–107. [Google Scholar] [CrossRef] [PubMed]
  131. Zhao, Z.; Ukidve, A.; Krishnan, V.; Fehnel, A.; Pan, D.C.; Gao, Y.; Kim, J.; Evans, M.A.; Mandal, A.; Guo, J.; et al. Systemic tumour suppression via the preferential accumulation of erythrocyte-anchored chemokine-encapsulating nanoparticles in lung metastases. Nat. Biomed. Eng. 2021, 5, 441–454. [Google Scholar] [CrossRef]
  132. Nosan, G.; Paro-Panjan, D.; Ihan, A.; Kopitar, A.N.; Čučnik, S.; Avčin, T. Vaccine immune response, autoimmunity and morbidity after neonatal blood exchange transfusion. Vaccine 2019, 37, 4076–4080. [Google Scholar] [CrossRef]
  133. Cerna, K.; Duricova, D.; Hindos, M.; Hindos Hrebackova, J.; Lukas, M.; Machkova, N.; Hruba, V.; Mitrova, K.; Kubickova, K.; Kastylova, K.; et al. Cellular and Humoral Immune Responses to SARS-CoV-2 Vaccination in Inflammatory Bowel Disease Patients. J. Crohn’s Colitis 2022, 16, 1347–1353. [Google Scholar] [CrossRef]
  134. Osborne, N.; Sundseth, R.; Burks, J.; Cao, H.; Liu, X.; Kroemer, A.H.; Sutton, L.; Cato, A.; Smith, J.P. Gastrin vaccine improves response to immune checkpoint antibody in murine pancreatic cancer by altering the tumor microenvironment. Cancer Immunol. Immunother. CII 2019, 68, 1635–1648. [Google Scholar] [CrossRef]
  135. Elizondo, C.R.; Bright, J.D.; Bright, R.K. Vaccination with a shared oncogenic tumor-self antigen elicits a population of CD8+ T cells with a regulatory phenotype. Hum. Vaccines Immunother. 2022, 18, 2108656. [Google Scholar] [CrossRef] [PubMed]
  136. Chung, D.J.; Shah, G.L.; Devlin, S.M.; Ramanathan, L.V.; Doddi, S.; Pessin, M.S.; Hoover, E.; Marcello, L.T.; Young, J.C.; Boutemine, S.R.; et al. Disease- and Therapy-Specific Impact on Humoral Immune Responses to COVID-19 Vaccination in Hematologic Malignancies. Blood Cancer Discov. 2021, 2, 568–576. [Google Scholar] [CrossRef]
  137. Toret, E.; Yel, S.E.; Suman, M.; Duzenli Kar, Y.; Ozdemir, Z.C.; Dinleyici, M.; Bor, O. Immunization status and re-immunization of childhood acute lymphoblastic leukemia survivors. Hum. Vaccines Immunother. 2021, 17, 1132–1135. [Google Scholar] [CrossRef] [PubMed]
  138. Oketch, S.Y.; Ochomo, E.O.; Orwa, J.A.; Mayieka, L.M.; Abdullahi, L.H. Communication strategies to improve human papillomavirus (HPV) immunisation uptake among adolescents in sub-Saharan Africa: A systematic review and meta-analysis. BMJ Open 2023, 13, e067164. [Google Scholar] [CrossRef] [PubMed]
  139. Ellingsen, E.B.; Aamdal, E.; Guren, T.; Lilleby, W.; Brunsvig, P.F.; Mangsbo, S.M.; Aamdal, S.; Hovig, E.; Mensali, N.; Gaudernack, G.; et al. Durable and dynamic hTERT immune responses following vaccination with the long-peptide cancer vaccine UV1: Long-term follow-up of three phase I clinical trials. J. Immunother. Cancer 2022, 10, e004345. [Google Scholar] [CrossRef]
  140. Wagner, A.; Garner-Spitzer, E.; Schötta, A.M.; Orola, M.; Wessely, A.; Zwazl, I.; Ohradanova-Repic, A.; Weseslindtner, L.; Tajti, G.; Gebetsberger, L.; et al. SARS-CoV-2-mRNA Booster Vaccination Reverses Non-Responsiveness and Early Antibody Waning in Immunocompromised Patients—A Phase Four Study Comparing Immune Responses in Patients with Solid Cancers, Multiple Myeloma and Inflammatory Bowel Disease. Front. Immunol. 2022, 13, 889138. [Google Scholar] [CrossRef] [PubMed]
  141. Wieske, L.; Stalman, E.W.; van Dam PJ, K.; Kummer, L.Y.; Steenhuis, M.; van Kempen ZL, E.; Killestein, J.; Volkers, A.G.; Tas, S.W.; Boekel, L.; et al. Persistence of seroconversion at 6 months following primary immunisation in patients with immune-mediated inflammatory diseases. Ann. Rheum. Dis. 2023, 82, 883–885. [Google Scholar] [CrossRef]
  142. Melssen, M.M.; Pollack, K.E.; Meneveau, M.O.; Smolkin, M.E.; Pinczewski, J.; Koeppel, A.F.; Turner, S.D.; Sol-Church, K.; Hickman, A.; Deacon, D.H.; et al. Characterization and comparison of innate and adaptive immune responses at vaccine sites in melanoma vaccine clinical trials. Cancer Immunol. Immunother. CII 2021, 70, 2151–2164. [Google Scholar] [CrossRef]
  143. Ogasawara, M.; Miyashita, M.; Yamagishi, Y.; Ota, S. Wilms’ tumor 1 peptide-loaded dendritic cell vaccination in patients with relapsed or refractory acute leukemia. Ther. Apher. Dial. 2022, 26, 537–547. [Google Scholar] [CrossRef]
  144. Xi, X.; Ye, T.; Wang, S.; Na, X.; Wang, J.; Qing, S.; Gao, X.; Wang, C.; Li, F.; Wei, W.; et al. Self-healing microcapsules synergetically modulate immunization microenvironments for potent cancer vaccination. Sci. Adv. 2020, 6, eaay7735. [Google Scholar] [CrossRef]
  145. Shi, Y.; Zhu, C.; Liu, Y.; Lu, Y.; Li, X.; Qin, B.; Luo, Z.; Luo, L.; Jiang, M.; Zhang, J.; et al. A Vaccination with Boosted Cross Presentation by ER-Targeted Antigen Delivery for Anti-Tumor Immunotherapy. Adv. Healthc. Mater. 2021, 10, e2001934. [Google Scholar] [CrossRef]
  146. Aleman, A.; van Kesteren, M.; Zajdman, A.K.; Srivastava, K.; Cognigni, C.; Mischka, J.; Chen, L.Y.; Upadhyaya, B.; Serebryakova, K.; Nardulli, J.R.; et al. Cellular mechanisms associated with sub-optimal immune responses to SARS-CoV-2 bivalent booster vaccination in patients with Multiple Myeloma. EBioMedicine 2023, 98, 104886. [Google Scholar] [CrossRef] [PubMed]
  147. Pasqualetti, F.; Zanotti, S. Nonrandomised controlled trial in recurrent glioblastoma patients: The promise of autologous tumour lysate-loaded dendritic cell vaccination. Br. J. Cancer 2023, 129, 895–896. [Google Scholar] [CrossRef] [PubMed]
  148. Goradel, N.H.; Negahdari, B.; Mohajel, N.; Malekshahi, Z.V.; Shirazi, M.M.A.; Arashkia, A. Heterologous administration of HPV16 E7 epitope-loaded nanocomplexes inhibits tumor growth in mouse model. Int. Immunopharmacol. 2021, 101, 108298. [Google Scholar] [CrossRef]
  149. Holm-Yildiz, S.; Dysgaard, T.; Krag, T.; Pedersen, B.S.; Hamm, S.R.; Pérez-Alós, L.; Hansen, C.B.; Pries-Heje, M.M.; Heftdal, L.D.; Hasselbalch, R.B.; et al. Humoral immune response to COVID-19 vaccine in patients with myasthenia gravis. J. Neuroimmunol. 2023, 384, 578215. [Google Scholar] [CrossRef]
  150. Bersanelli, M.; Buti, S.; De Giorgi, U.; Di Maio, M.; Giannarelli, D.; Pignata, S.; Banna, G.L. State of the art about influenza vaccination for advanced cancer patients receiving immune checkpoint inhibitors: When common sense is not enough. Crit. Rev. Oncol. Hematol. 2019, 139, 87–90. [Google Scholar] [CrossRef] [PubMed]
  151. Dykman, L.A.; Staroverov, S.A.; Kozlov, S.V.; Fomin, A.S.; Chumakov, D.S.; Gabalov, K.P.; Kozlov, Y.S.; Soldatov, D.A.; Khlebtsov, N.G. Immunization of Mice with Gold Nanoparticles Conjugated to Thermostable Cancer Antigens Prevents the Development of Xenografted Tumors. Int. J. Mol. Sci. 2022, 23, 14313. [Google Scholar] [CrossRef] [PubMed]
  152. Valanparambil, R.M.; Carlisle, J.; Linderman, S.L.; Akthar, A.; Millett, R.L.; Lai, L.; Chang, A.; McCook-Veal, A.A.; Switchenko, J.; Nasti, T.H.; et al. Antibody Response to COVID-19 mRNA Vaccine in Patients with Lung Cancer After Primary Immunization and Booster: Reactivity to the SARS-CoV-2 WT Virus and Omicron Variant. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2022, 40, 3808–3816. [Google Scholar] [CrossRef] [PubMed]
  153. Mair, M.J.; Berger, J.M.; Mitterer, M.; Gansterer, M.; Bathke, A.C.; Trutschnig, W.; Berghoff, A.S.; Perkmann, T.; Haslacher, H.; Lamm, W.W.; et al. Third dose of SARS-CoV-2 vaccination in hemato-oncological patients and health care workers: Immune responses and adverse events—A retrospective cohort study. Eur. J. Cancer 2022, 165, 184–194. [Google Scholar] [CrossRef]
  154. Meneveau, M.O.; Kumar, P.; Lynch, K.T.; Patel, S.P.; Slingluff, C.L. The vaccine-site microenvironment: Impacts of antigen, adjuvant, and same-site vaccination on antigen presentation and immune signaling. J. Immunother. Cancer 2022, 10, e003533. [Google Scholar] [CrossRef]
  155. Wankhede, D.; Grover, S.; Hofman, P. Determinants of humoral immune response to SARS-CoV-2 vaccines in solid cancer patients: A systematic review and meta-analysis. Vaccine 2023, 41, 1791–1798. [Google Scholar] [CrossRef] [PubMed]
  156. Faustini, S.E.; Hall, A.; Brown, S.; Roberts, S.; Hill, H.; Stamataki, Z.; (PITCH) consortium; Jenner, M.W.; Owen, R.G.; Pratt, G.; et al. Immune responses to COVID-19 booster vaccinations in intensively anti-CD38 antibody treated patients with ultra-high-risk multiple myeloma: Results from the Myeloma UK (MUK) nine OPTIMUM trial. Br. J. Haematol. 2023, 201, 845–850. [Google Scholar] [CrossRef]
  157. Meza, L.; Zengin, Z.; Salgia, S.; Malhotra, J.; Karczewska, E.; Dorff, T.; Tripathi, A.; Ely, J.; Kelley, E.; Mead, H.; et al. Twelve-Month Follow-up of the Immune Response After COVID-19 Vaccination in Patients with Genitourinary Cancers: A Prospective Cohort Analysis. The Oncologist 2023, 28, e748–e755. [Google Scholar] [CrossRef] [PubMed]
  158. Deng, M.Y.; Debus, J.; König, L. Verlängerung des Gesamtüberlebens durch die Impfung von autologen tumorlysatbeladenen dendritischen Zellen (DCVax-L) bei Patienten mit neu diagnostiziertem und rezidivierendem Glioblastom [Association of autologous tumor lysate-loaded dendritic cell vaccination with extension of survival among patients with newly diagnosed and recurrent glioblastoma]. Strahlenther. Und Onkol. Organ Der Dtsch. Rontgenges. 2023, 199, 327–329. [Google Scholar] [CrossRef]
  159. Souan, L.; Abdel-Razeq, H.; Al Zughbieh, M.; Al Badr, S.; Sughayer, M.A. Comparative Assessment of the Kinetics of Cellular and Humoral Immune Responses to COVID-19 Vaccination in Cancer Patients. Viruses 2023, 15, 1439. [Google Scholar] [CrossRef]
  160. Yang, J.; Eresen, A.; Shangguan, J.; Ma, Q.; Yaghmai, V.; Zhang, Z. Irreversible electroporation ablation overcomes tumor-associated immunosuppression to improve the efficacy of DC vaccination in a mice model of pancreatic cancer. Oncoimmunology 2021, 10, 1875638. [Google Scholar] [CrossRef] [PubMed]
  161. Pedrazzoli, P.; Lasagna, A.; Cassaniti, I.; Ferrari, A.; Bergami, F.; Silvestris, N.; Sapuppo, E.; Di Maio, M.; Cinieri, S.; Baldanti, F. Vaccination for herpes zoster in patients with solid tumors: A position paper on the behalf of the Associazione Italiana di Oncologia Medica (AIOM). ESMO Open 2022, 7, 100548. [Google Scholar] [CrossRef]
  162. MacKerracher, A.; Sommershof, A.; Groettrup, M. PLGA particle vaccination elicits resident memory CD8 T cells protecting from tumors and infection. Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci. 2022, 175, 106209. [Google Scholar] [CrossRef]
  163. Barrière, J.; Re, D.; Peyrade, F.; Carles, M. Current perspectives for SARS-CoV-2 vaccination efficacy improvement in patients with active treatment against cancer. Eur. J. Cancer 2021, 154, 66–72. [Google Scholar] [CrossRef] [PubMed]
  164. Zhuang, W.H.; Wang, Y.P. Analysis of the immunity effects after enhanced hepatitis B vaccination on patients with lymphoma. Leuk. Lymphoma 2020, 61, 357–363. [Google Scholar] [CrossRef]
  165. Storti, P.; Marchica, V.; Vescovini, R.; Franceschi, V.; Russo, L.; Notarfranchi, L.; Raimondi, V.; Toscani, D.; Burroughs Garcia, J.; Costa, F.; et al. Immune response to SARS-CoV-2 mRNA vaccination and booster dose in patients with multiple myeloma and monoclonal gammopathies: Impact of Omicron variant on the humoral response. Oncoimmunology 2022, 11, 2120275. [Google Scholar] [CrossRef]
  166. Mitchell, D.K.; Burgess, B.; White, E.E.; Smith, A.E.; Sierra Potchanant, E.A.; Mang, H.; Hickey, B.E.; Lu, Q.; Qian, S.; Bessler, W.; et al. Spatial Gene-Expression Profiling Unveils Immuno-oncogenic Programs of NF1-Associated Peripheral Nerve Sheath Tumor Progression. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2024, 30, 1038–1053. [Google Scholar] [CrossRef]
  167. Martin, S.D.; Nziza, N.; Miozzo, P.; Bartsch, Y.; Farkas, E.J.; Kane, A.S.; Boal, L.H.; Friedmann, A.; Alter, G.; Yonker, L.M. Humoral profiling of pediatric patients with cancer reveals robust immunity following anti-SARS-CoV-2 vaccination superior to natural infection. Pediatr. Blood Cancer 2023, 70, e30473. [Google Scholar] [CrossRef]
  168. Rensink, M.J.; van Laarhoven HW, M.; Holleman, F. Cocoon vaccination for influenza in patients with a solid tumor: A retrospective study. Support. Care Cancer Off. J. Multinatl. Assoc. Support. Care Cancer 2021, 29, 3657–3666. [Google Scholar] [CrossRef] [PubMed]
  169. Iavarone, M.; Tosetti, G.; Facchetti, F.; Topa, M.; Er, J.M.; Hang, S.K.; Licari, D.; Lombardi, A.; D’Ambrosio, R.; Degasperi, E.; et al. Spike-specific humoral and cellular immune responses after COVID-19 mRNA vaccination in patients with cirrhosis: A prospective single center study. Dig. Liver Dis. Off. J. Ital. Soc. Gastroenterol. Ital. Assoc. Study Liver 2023, 55, 160–168. [Google Scholar] [CrossRef]
  170. Shakibapour, M.; Kefayat, A.; Reza Mofid, M.; Shojaie, B.; Mohamadi, F.; Maryam Sharafi, S.; Mahmoudzadeh, M.; Yousofi Darani, H. Anti-cancer immunoprotective effects of immunization with hydatid cyst wall antigens in a non-immunogenic and metastatic triple-negative murine mammary carcinoma model. Int. Immunopharmacol. 2021, 99, 107955. [Google Scholar] [CrossRef] [PubMed]
  171. Oltmanns, F.; Vieira Antão, A.; Irrgang, P.; Viherlehto, V.; Jörg, L.; Schmidt, A.; Wagner, J.T.; Rückert, M.; Flohr, A.S.; Geppert, C.I.; et al. Mucosal tumor vaccination delivering endogenous tumor antigens protects against pulmonary breast cancer metastases. J. Immunother. Cancer 2024, 12, e008652. [Google Scholar] [CrossRef]
  172. Patchett, A.L.; Tovar, C.; Blackburn, N.B.; Woods, G.M.; Lyons, A.B. Mesenchymal plasticity of devil facial tumour cells during in vivo vaccine and immunotherapy trials. Immunol. Cell Biol. 2021, 99, 711–723. [Google Scholar] [CrossRef]
  173. Masoumi, J.; Jafarzadeh, A.; Tavakoli, T.; Basirjafar, P.; Zandvakili, R.; Javan, M.R.; Taghipour, Z.; Moazzeni, S.M. Inhibition of apelin/APJ axis enhances the potential of dendritic cell-based vaccination to modulate TH1 and TH2 cell-related immune responses in an animal model of metastatic breast cancer. Adv. Med. Sci. 2022, 67, 170–178. [Google Scholar] [CrossRef]
  174. Kim, J.; Jeong, J.; Lee, C.M.; Lee, D.W.; Kang, C.K.; Choe, P.G.; Kim, N.J.; Oh, M.D.; Lee, C.H.; Park, W.B.; et al. Prospective longitudinal analysis of antibody response after standard and booster doses of SARS-COV2 vaccination in patients with early breast cancer. Front. Immunol. 2022, 13, 1028102. [Google Scholar] [CrossRef] [PubMed]
  175. Lyski, Z.L.; Kim, M.S.; Xthona Lee, D.; Raué, H.P.; Raghunathan, V.; Griffin, J.; Ryan, D.; Brunton, A.E.; Curlin, M.E.; Slifka, M.K.; et al. Cellular and humoral immune response to mRNA COVID-19 vaccination in subjects with chronic lymphocytic leukemia. Blood Adv. 2022, 6, 1207–1211. [Google Scholar] [CrossRef]
  176. Jung, E.; Mao, C.; Bhatia, M.; Koellhoffer, E.C.; Fiering, S.N.; Steinmetz, N.F. Inactivated Cowpea Mosaic Virus for In Situ Vaccination: Differential Efficacy of Formalin vs UV-Inactivated Formulations. Mol. Pharm. 2023, 20, 500–507. [Google Scholar] [CrossRef]
  177. Cecil, D.L.; Liao, J.B.; Dang, Y.; Coveler, A.L.; Kask, A.; Yang, Y.; Childs, J.S.; Higgins, D.M.; Disis, M.L. Immunization with a Plasmid DNA Vaccine Encoding the N-Terminus of Insulin-like Growth Factor Binding Protein-2 in Advanced Ovarian Cancer Leads to High-level Type I Immune Responses. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 6405–6412. [Google Scholar] [CrossRef]
  178. Taylor, A.C.; Hopkins, L.W.; Moore, G. Increasing human papillomavirus immunization in the primary care setting. Nurse Pract. 2021, 46, 37–42. [Google Scholar] [CrossRef]
  179. Stumpf, J.; Anders, L.; Siepmann, T.; Schwöbel, J.; Karger, C.; Lindner, T.; Faulhaber-Walter, R.; Langer, T.; Escher, K.; Anding-Rost, K.; et al. 9-Month observational Dia-Vacc study of vaccine type influence on SARS-CoV-2 immunity in dialysis and kidney transplant patients. Vaccine 2024, 42, 120–128. [Google Scholar] [CrossRef] [PubMed]
  180. Purshouse, K.; Thomson, J.P.; Vallet, M.; Alexander, L.; Bonisteel, I.; Brennan, M.; Cameron, D.A.; Figueroa, J.D.; Furrie, E.; Haig, P.; et al. The Scottish COVID Cancer Immunity Prevalence Study: A Longitudinal Study of SARS-CoV-2 Immune Response in Patients Receiving Anti-Cancer Treatment. Oncol. 2023, 28, e145–e155. [Google Scholar] [CrossRef] [PubMed]
  181. Bacova, B.; Kohutova, Z.; Zubata, I.; Gaherova, L.; Kucera, P.; Heizer, T.; Mikesova, M.; Karel, T.; Novak, J. Cellular and humoral immune response to SARS-CoV-2 mRNA vaccines in patients treated with either Ibrutinib or Rituximab. Clin. Exp. Med. 2023, 23, 371–379. [Google Scholar] [CrossRef] [PubMed]
  182. Ukidve, A.; Zhao, Z.; Fehnel, A.; Krishnan, V.; Pan, D.C.; Gao, Y.; Mandal, A.; Muzykantov, V.; Mitragotri, S. Erythrocyte-driven immunization via biomimicry of their natural antigen-presenting function. Proc. Natl. Acad. Sci. USA 2020, 117, 17727–17736. [Google Scholar] [CrossRef] [PubMed]
  183. Müller, K.E.; Dohos, D.; Sipos, Z.; Kiss, S.; Dembrovszky, F.; Kovács, N.; Solymár, M.; Erőss, B.; Hegyi, P.; Sarlós, P. Immune response to influenza and pneumococcal vaccines in adults with inflammatory bowel disease: A systematic review and meta-analysis of 1429 patients. Vaccine 2022, 40, 2076–2086. [Google Scholar] [CrossRef] [PubMed]
  184. Debie, Y.; Van Audenaerde, J.R.M.; Vandamme, T.; Croes, L.; Teuwen, L.A.; Verbruggen, L.; Vanhoutte, G.; Marcq, E.; Verheggen, L.; Le Blon, D.; et al. Humoral and Cellular Immune Responses against SARS-CoV-2 after Third Dose BNT162b2 following Double-Dose Vaccination with BNT162b2 versus ChAdOx1 in Patients with Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2023, 29, 635–646. [Google Scholar] [CrossRef] [PubMed]
  185. Salmon, C.; Conus, F.; Parent M, É.; Benedetti, A.; Rousseau, M.C. Association between Bacillus Calmette-Guerin (BCG) vaccination and lymphoma risk: A systematic review and meta-analysis. Cancer Epidemiol. 2020, 65, 101696. [Google Scholar] [CrossRef] [PubMed]
  186. Gharibi, Z.; Rahdar, M.; Pirestani, M.; Tavalla, M.; Tabandeh, M.R. The Immunization of Protoscolices P29 DNA Vaccine on Experimental Cystic Echinococosis in Balb/c Mice. Acta Parasitol. 2021, 66, 1114–1121. [Google Scholar] [CrossRef] [PubMed]
  187. Ishida, E.; Lee, J.; Campbell, J.S.; Chakravarty, P.D.; Katori, Y.; Ogawa, T.; Johnson, L.; Mukhopadhyay, A.; Faquin, W.C.; Lin, D.T.; et al. Intratumoral delivery of an HPV vaccine elicits a broad anti-tumor immune response that translates into a potent anti-tumor effect in a preclinical murine HPV model. Cancer Immunol. Immunother. CII 2019, 68, 1273–1286. [Google Scholar] [CrossRef]
  188. Aleman, A.; Van Oekelen, O.; Upadhyaya, B.; Beach, K.; Kogan Zajdman, A.; Alshammary, H.; Serebryakova, K.; Agte, S.; Kappes, K.; Gleason, C.R.; et al. Augmentation of humoral and cellular immune responses after third-dose SARS-CoV-2 vaccination and viral neutralization in myeloma patients. Cancer Cell 2022, 40, 441–443. [Google Scholar] [CrossRef]
  189. Hou, X.; Shi, Y.; Kang, X.; Rousu, Z.; Li, D.; Wang, M.; Ainiwaer, A.; Zheng, X.; Wang, M.; Jiensihan, B.; et al. Echinococcus granulosus: The establishment of the metacestode in the liver is associated with control of the CD4+ T-cell-mediated immune response in patients with cystic echinococcosis and a mouse model. Front. Cell. Infect. Microbiol. 2022, 12, 983119. [Google Scholar] [CrossRef] [PubMed]
  190. Campal-Espinosa, A.C.; Junco-Barranco, J.A.; Fuentes-Aguilar, F.; Calzada-Aguilera, L.; Rivacoba-Betancourt, A.; Rodríguez-Bueno, R.H.; Bover-Campal, A.C.; Bover-Fuentes, E.E.; González, L.; de Quesada, L.; et al. Influence of Humoral Response Against GnRH, Generated by Immunization with a Therapeutic Vaccine Candidate on the Evolution of Patients with Castration-Sensitive Prostate Adenocarcinoma. Technol. Cancer Res. Treat. 2023, 22, 15330338231207318. [Google Scholar] [CrossRef]
  191. Behrendt, D.; Burger, D.; Gremmes, S.; Szunyog, K.; Röthemeier, S.; Sieme, H. Active immunisation against GnRH as treatment for unilateral granulosa theca cell tumour in mares. Equine Vet. J. 2021, 53, 740–745. [Google Scholar] [CrossRef]
  192. Lehrnbecher, T.; Sack, U.; Speckmann, C.; Groll, A.H.; Boldt, A.; Siebald, B.; Hettmer, S.; Demmerath, E.M.; Reemtsma, J.; Schenk, B.; et al. Longitudinal Immune Response to 3 Doses of Messenger RNA Vaccine Against Coronavirus Disease 2019 (COVID-19) in Pediatric Patients Receiving Chemotherapy for Cancer. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2023, 76, e510–e513. [Google Scholar] [CrossRef]
  193. Cobanoglu, O.; Delval, L.; Ferrari, D.; Deruyter, L.; Heumel, S.; Wolowczuk, I.; Hussein, A.; Menevse, A.N.; Bernard, D.; Beckhove, P.; et al. Depletion of preexisting B-cell lymphoma 2-expressing senescent cells before vaccination impacts antigen-specific antitumor immune responses in old mice. Aging Cell 2023, 22, e14007. [Google Scholar] [CrossRef]
  194. Kallionpää, R.A.; Peltonen, S.; Le, K.M.; Martikkala, E.; Jääskeläinen, M.; Fazeli, E.; Riihilä, P.; Haapaniemi, P.; Rokka, A.; Salmi, M.; et al. Characterization of Immune Cell Populations of Cutaneous Neurofibromas in Neurofibromatosis 1. Lab. Investi. A J. Tech. Methods Pathol. 2024, 104, 100285. [Google Scholar] [CrossRef]
  195. Borgogna, C.; Bruna, R.; Griffante, G.; Martuscelli, L.; De Andrea, M.; Ferrante, D.; Patriarca, A.; Mahmoud, A.M.; Ucciero, M.A.; Gaidano, V.; et al. Induction of robust humoral immunity against SARS-CoV-2 after vaccine administration in previously infected haematological cancer patients. Br. J. Haematol. 2022, 199, 463–467. [Google Scholar] [CrossRef]
  196. Kang, C.K.; Kim, H.R.; Song, K.H.; Keam, B.; Choi, S.J.; Choe, P.G.; Kim, E.S.; Kim, N.J.; Kim, Y.J.; Park, W.B.; et al. Cell-Mediated Immunogenicity of Influenza Vaccination in Patients with Cancer Receiving Immune Checkpoint Inhibitors. J. Infect. Dis. 2020, 222, 1902–1909. [Google Scholar] [CrossRef]
  197. Viana, J.H.M.; Pereira, N.E.S.; Faria, O.A.C.; Dias, L.R.O.; Oliveira, E.R.; Fernandes, C.A.C.; Siqueira, L.G.B. Active immunization against GnRH as an alternative therapeutic approach for the management of Bos indicus oocyte donors diagnosed with chronic cystic ovarian disease. Theriogenology 2021, 172, 133–141. [Google Scholar] [CrossRef] [PubMed]
  198. Martins-Branco, D.; Nader-Marta, G.; Tecic Vuger, A.; Debien, V.; Ameye, L.; Brandão, M.; Punie, K.; Loizidou, A.; Willard-Gallo, K.; Spilleboudt, C.; et al. Immune response to anti-SARS-CoV-2 prime-vaccination in patients with cancer: A systematic review and meta-analysis. J. Cancer Res. Clin. Oncol. 2023, 149, 3075–3080. [Google Scholar] [CrossRef] [PubMed]
  199. Vanni, A.; Salvati, L.; Mazzoni, A.; Lamacchia, G.; Capone, M.; Francalanci, S.; Kiros, S.T.; Cosmi, L.; Puccini, B.; Ciceri, M.; et al. Bendamustine impairs humoral but not cellular immunity to SARS-CoV-2 vaccination in rituximab-treated B-cell lymphoma-affected patients. Front. Immunol. 2023, 14, 1322594. [Google Scholar] [CrossRef]
  200. Titova, E.; Kan, V.W.; Lozy, T.; Ip, A.; Shier, K.; Prakash, V.P.; Starolis, M.; Ansari, S.; Goldgirsh, K.; Kim, S.; et al. Humoral and cellular immune responses against SARS-CoV-2 post-vaccination in immunocompetent and immunocompromised cancer populations. Microbiol. Spectr. 2024, 12, e0205023. [Google Scholar] [CrossRef]
  201. Weitgasser, L.; Mahrhofer, M.; Schoeller, T. Potential immune response to breast implants after immunization with COVID-19 vaccines. Breast 2021, 59, 76–78. [Google Scholar] [CrossRef]
  202. Aurisicchio, L.; Fridman, A.; Mauro, D.; Sheloditna, R.; Chiappori, A.; Bagchi, A.; Ciliberto, G. Safety, tolerability and immunogenicity of V934/V935 hTERT vaccination in cancer patients with selected solid tumors: A phase I study. J. Transl. Med. 2020, 18, 39. [Google Scholar] [CrossRef] [PubMed]
  203. Xu, P.; Ma, J.; Zhou, Y.; Gu, Y.; Cheng, X.; Wang, Y.; Wang, Y.; Gao, M. Radiotherapy-Triggered In Situ Tumor Vaccination Boosts Checkpoint Blockaded Immune Response via Antigen-Capturing Nanoadjuvants. ACS Nano 2024, 18, 1022–1040. [Google Scholar] [CrossRef]
  204. Peeters, M.; Verbruggen, L.; Teuwen, L.; Vanhoutte, G.; Vande Kerckhove, S.; Peeters, B.; Raats, S.; Van der Massen, I.; De Keersmaecker, S.; Debie, Y.; et al. Reduced humoral immune response after BNT162b2 coronavirus disease 2019 messenger RNA vaccination in cancer patients under antineoplastic treatment. ESMO Open 2021, 6, 100274. [Google Scholar] [CrossRef]
  205. Lövgren, T.; Wolodarski, M.; Wickström, S.; Edbäck, U.; Wallin, M.; Martell, E.; Markland, K.; Blomberg, P.; Nyström, M.; Lundqvist, A.; et al. Complete and long-lasting clinical responses in immune checkpoint inhibitor-resistant, metastasized melanoma treated with adoptive T cell transfer combined with DC vaccination. Oncoimmunology 2020, 9, 1792058. [Google Scholar] [CrossRef]
  206. Enssle, J.C.; Campe, J.; Büchel, S.; Moter, A.; See, F.; Grießbaum, K.; Rieger, M.A.; Wolf, S.; Ballo, O.; Steffen, B.; et al. Enhanced but variant-dependent serological and cellular immune responses to third-dose BNT162b2 vaccination in patients with multiple myeloma. Cancer Cell 2022, 40, 587–589. [Google Scholar] [CrossRef] [PubMed]
  207. Cole, G.; Ali, A.A.; McErlean, E.; Mulholland, E.J.; Short, A.; McCrudden, C.M.; McCaffrey, J.; Robson, T.; Kett, V.L.; Coulter, J.A.; et al. DNA vaccination via RALA nanoparticles in a microneedle delivery system induces a potent immune response against the endogenous prostate cancer stem cell antigen. Acta Biomater. 2019, 96, 480–490. [Google Scholar] [CrossRef]
  208. Alimam, S.; Ann Timms, J.; Harrison, C.N.; Dillon, R.; Mare, T.; DeLavallade, H.; Radia, D.; Woodley, C.; Francis, Y.; Sanchez, K.; et al. Altered immune response to the annual influenza A vaccine in patients with myeloproliferative neoplasms. Br. J. Haematol. 2021, 193, 150–154. [Google Scholar] [CrossRef] [PubMed]
  209. Zou, Z.; Guo, L.; Mautner, V.; Smeets, R.; Kiuwe, L.; Friedrich, R.E. Propranolol Specifically Suppresses the Viability of Tumorous Schwann Cells Derived from Plexiform Neurofibromas In Vitro. Vivo 2020, 34, 1031–1036. [Google Scholar] [CrossRef]
  210. Mohan, M.; Nagavally, S.; Shah, N.N.N.; Michaelis, L.; Chhabra, S.; Souza, A.D.; Abedin, S.; Runaas, L.; Guru Murthy, G.S.; Longo, W.; et al. Shorter Interval between Treatment and COVID Immunization Is Associated with Poor Seroconversion in Patients with Hematological Malignancies. Clin. Lymphoma Myeloma Leuk. 2022, 22, e495–e497. [Google Scholar] [CrossRef]
  211. Meena, J.; Kumar, R.; Singh, M.; Ahmed, A.; Panda, A.K. Modulation of immune response and enhanced clearance of Salmonella typhi by delivery of Vi polysaccharide conjugate using PLA nanoparticles. Eur. J. Pharm. Biopharm. Off. J. Arbeitsgemeinschaft Fur Pharm. Verfahrenstechnik E.V 2020, 152, 270–281. [Google Scholar] [CrossRef] [PubMed]
  212. Suzuki, T.; Kusumoto, S.; Kamezaki, Y.; Hashimoto, H.; Nishitarumizu, N.; Nakanishi, Y.; Kato, Y.; Kawai, A.; Matsunaga, N.; Ebina, T.; et al. A comprehensive evaluation of humoral immune response to second and third SARS-CoV-2 mRNA vaccination in patients with malignant lymphoma. Int. J. Hematol. 2023, 117, 900–909. [Google Scholar] [CrossRef]
  213. Cerda, C.; Martínez-Valdebenito, C.; Barriga, F.; Contreras, M.; Vidal, M.; Moreno, R.; Claverie, X.; Contreras, P.; Huenuman, L.; García, T.; et al. Respuesta inmune humoral inducida por la vacuna influenza en niños con diagnóstico de leucemia linfoblástica aguda [Humoral immune response induced by influenza vaccine in children with acute lymphoblastic leukemia]. Rev. Chil. De Infectol. Organo Of. De La Soc. Chil. De Infectol. 2020, 37, 138–146. [Google Scholar] [CrossRef] [PubMed]
  214. He, Y.; Chen, D.; Fu, Y.; Huo, X.; Zhao, F.; Yao, L.; Zhou, X.; Qi, P.; Yin, H.; Cao, L.; et al. Immunization with Tp0954, an adhesin of Treponema pallidum, provides protective efficacy in the rabbit model of experimental syphilis. Front. Immunol. 2023, 14, 1130593. [Google Scholar] [CrossRef] [PubMed]
  215. Dahiya, S.; Luetkens, T.; Lutfi, F.; Avila, S.; Iraguha, T.; Margiotta, P.; Hankey, K.G.; Lesho, P.; Law, J.Y.; Lee, S.T.; et al. Impaired immune response to COVID-19 vaccination in patients with B-cell malignancies after CD19 CAR T-cell therapy. Blood Adv. 2022, 6, 686–689. [Google Scholar] [CrossRef]
  216. Barber, V.S.; Peckham, N.; Duley, L.; Francis, A.; Abhishek, A.; Moss, P.; Cook, J.A.; Parry, H.M. Protocol for a multicentre randomised controlled trial examining the effects of temporarily pausing Bruton tyrosine kinase inhibitor therapy to coincide with SARS-CoV-2 vaccination and its impact on immune responses in patients with chronic lymphocytic leukaemia. BMJ Open 2023, 13, e077946. [Google Scholar] [CrossRef] [PubMed]
  217. Kanjanapan, Y.; Blinman, P.; Underhill, C.; Karikios, D.; Segelov, E.; Yip, D. Medical Oncology Group of Australia position statement: COVID-19 vaccination in patients with solid tumours. Intern. Med. J. 2021, 51, 955–959. [Google Scholar] [CrossRef] [PubMed]
  218. Fang, S.; Agostinis, P.; Salven, P.; Garg, A.D. Decoding cancer cell death-driven immune cell recruitment: An in vivo method for site-of-vaccination analyses. Methods Enzymol. 2020, 636, 185–207. [Google Scholar] [CrossRef] [PubMed]
  219. Stumpf, J.; Klimova, A.; Mauer, R.; Steglich, A.; Gembardt, F.; Martin, H.; Glombig, G.; Frank, K.; Tonn, T.; Hugo, C. Equivalent humoral and cellular immune response but different side effect rates following SARS-CoV-2 vaccination in peritoneal and haemodialysis patients using messenger RNA vaccines. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. Eur. Ren. Assoc. 2022, 37, 796–798. [Google Scholar] [CrossRef]
  220. Lozano-Rodríguez, R.; Terrón-Arcos, V.; Montalbán-Hernández, K.; Casalvilla-Dueñas, J.C.; Bergón-Gutierrez, M.; Pascual-Iglesias, A.; Quiroga, J.V.; Aguirre, L.A.; Pérez de Diego, R.; Vela-Olmo, C.; et al. Prior SARS-CoV-2 infection balances immune responses triggered by four EMA-approved COVID-19 vaccines: An observational study. Clin. Transl. Med. 2022, 12, e869. [Google Scholar] [CrossRef]
  221. Boerenkamp, L.S.; Pothast, C.R.; Dijkland, R.C.; van Dijk, K.; van Gorkom, G.N.Y.; van Loo, I.H.M.; Wieten, L.; Halkes, C.J.M.; Heemskerk, M.H.M.; Van Elssen, C.H. Increased CD8 T-cell immunity after COVID-19 vaccination in lymphoid malignancy patients lacking adequate humoral response: An immune compensation mechanism? Am. J. Hematol. 2022, 97, E457–E461. [Google Scholar] [CrossRef]
  222. Choi, D.K.; Strzepka, J.T.; Hunt, S.R.; Tannenbaum, V.L.; Jang, I.E. Vaccination in pediatric cancer survivors: Vaccination rates, immune status, and knowledge regarding compliance. Pediatr. Blood Cancer 2020, 67, e28565. [Google Scholar] [CrossRef]
  223. Xing, Y.; Yang, J.; Yao, P.; Xie, L.; Liu, M.; Cai, Y. Comparison of the immune response and protection against the experimental Toxoplasma gondii infection elicited by immunization with the recombinant proteins BAG1, ROP8, and BAG1-ROP8. Parasite Immunol. 2024, 46, e13023. [Google Scholar] [CrossRef]
  224. Sesques, P.; Bachy, E.; Ferrant, E.; Safar, V.; Gossez, M.; Morfin-Sherpa, F.; Venet, F.; Ader, F. Immune response to three doses of mRNA SARS-CoV-2 vaccines in CD19-targeted chimeric antigen receptor T cell immunotherapy recipients. Cancer Cell 2022, 40, 236–237. [Google Scholar] [CrossRef]
  225. Seban, R.D.; Champion, L.; Yeh, R.; Schwartz, L.H.; Dercle, L. Assessing immune response upon systemic RNA vaccination on [18F]-FDG PET/CT for COVID-19 vaccine and then for immuno-oncology? Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 3351–3352. [Google Scholar] [CrossRef]
  226. Moulik, N.R.; Mandal, P.; Chandra, J.; Bansal, S.; Jog, P.; Sanjay, S.; Shah, N.; Arora, R.S. Immunization of Children with Cancer in India Treated with Chemotherapy—Consensus Guideline from the Pediatric Hematology-Oncology Chapter and the Advisory Committee on Vaccination and Immunization Practices of the Indian Academy of Pediatrics. Indian Pediatr. 2019, 56, 1041–1048. [Google Scholar] [CrossRef]
  227. Motwani, K.K.; Hashash, J.G.; Farraye, F.A.; Kappelman, M.D.; Weaver, K.N.; Zhang, X.; Long, M.D.; Cross, R.K. Impact of Holding Immunosuppressive Therapy in Patients with Inflammatory Bowel Disease Around mRNA COVID-19 Vaccine Administration on Humoral Immune Response and Development of COVID-19 Infection. J. Crohn’s Colitis 2023, 17, 1681–1688. [Google Scholar] [CrossRef]
  228. Safavi, A.; Kefayat, A.; Ghahremani, F.; Mahdevar, E.; Moshtaghian, J. Immunization using male germ cells and gametes as rich sources of cancer/testis antigens for inhibition of 4T1 breast tumors’ growth and metastasis in BALB/c mice. Int. Immunopharmacol. 2019, 74, 105719. [Google Scholar] [CrossRef]
  229. Lundstrom, K. Immune Responses of Alphavirus Vaccination in Patients with HPV-Induced Cancers. Mol. Ther. J. Am. Soc. Gene Ther. 2021, 29, 415–416. [Google Scholar] [CrossRef]
  230. Oosting, S.F.; van der Veldt, A.A.M.; Fehrmann, R.S.N.; Bhattacharya, A.; van Binnendijk, R.S.; GeurtsvanKessel, C.H.; Dingemans, A.C.; Smit, E.F.; Hiltermann, T.J.N.; den Hartog, G.; et al. Factors associated with long-term antibody response after COVID-19 vaccination in patients treated with systemic treatment for solid tumors. ESMO Open 2023, 8, 101599. [Google Scholar] [CrossRef]
  231. Óskarsson, Ý.; Thors, V.; Vias, R.D.; Lúðvíksson, B.R.; Brynjólfsson, S.F.; Gianchecchi, E.; Razzano, I.; Montomoli, E.; Gísli Jónsson, Ó.; Haraldsson, Á. Adequate immune responses to vaccines after chemotherapy for leukaemia diagnosed in childhood. Acta Paediatr. 2024, 113, 606–614. [Google Scholar] [CrossRef]
  232. Kleebayoon, A.; Wiwanitkit, V. Comment on: Humoral profiling of pediatric patients with cancer reveals robust immunity following anti-SARS-CoV-2 vaccination superior to natural infection. Pediatr. Blood Cancer 2023, 70, e30509. [Google Scholar] [CrossRef]
  233. Woodfield, M.C.; Carpenter, P.A.; Pergam, S.A. Shots, Not Moonshots-The Importance of Broad Population Immunization to Patients Who Undergo Cancer Treatment. JAMA Oncol. 2020, 6, 23–24. [Google Scholar] [CrossRef]
  234. Veinalde, R. Evaluation of Oncolytic Virus-Induced Therapeutic Tumor Vaccination Effects in Murine Tumor Models. Methods Mol. Biol. 2020, 2058, 213–227. [Google Scholar] [CrossRef]
  235. Ryu, H.H.; Chang, K.; Kim, N.; Lee, H.S.; Hwang, S.W.; Park, S.H.; Yang, D.H.; Byeon, J.S.; Myung, S.J.; Yang, S.K.; et al. Insufficient vaccination and inadequate immunization rates among Korean patients with inflammatory bowel diseases. Medicine 2021, 100, e27714. [Google Scholar] [CrossRef] [PubMed]
  236. Wang, W.; Li, X.; Qin, X.; Miao, Y.; Zhang, Y.; Li, S.; Yao, R.; Yang, Y.; Yu, L.; Zhu, H.; et al. Germline Neurofibromin 1 mutation enhances the anti-tumour immune response and decreases juvenile myelomonocytic leukaemia tumourigenicity. Br. J. Haematol. 2023, 202, 328–343. [Google Scholar] [CrossRef] [PubMed]
  237. Ginefra, P.; Lorusso, G.; Vannini, N. Innate Immune Cells and Their Contribution to T-Cell-Based Immunotherapy. Int. J. Mol. Sci. 2020, 21, 4441. [Google Scholar] [CrossRef]
  238. Alicke, B.; Totpal, K.; Schartner, J.M.; Berkley, A.M.; Lehar, S.M.; Capietto, A.H.; Cubas, R.A.; Gould, S.E. Immunization associated with primary tumor growth leads to rejection of commonly used syngeneic tumors upon tumor rechallenge. J. Immunother. Cancer 2020, 8, e000532. [Google Scholar] [CrossRef]
  239. Song, X.; Jiang, Y.; Zhang, W.; Elfawal, G.; Wang, K.; Jiang, D.; Hong, H.; Wu, J.; He, C.; Mo, X.; et al. Transcutaneous tumor vaccination combined with anti-programmed death-1 monoclonal antibody treatment produces a synergistic antitumor effect. Acta Biomater. 2022, 140, 247–260. [Google Scholar] [CrossRef]
  240. Muhammad, Q.; Jang, Y.; Kang, S.H.; Moon, J.; Kim, W.J.; Park, H. Modulation of immune responses with nanoparticles and reduction of their immunotoxicity. Biomater. Sci. 2020, 8, 1490–1501. [Google Scholar] [CrossRef]
  241. Fujii, S.I.; Shimizu, K. Immune Networks and Therapeutic Targeting of iNKT Cells in Cancer. Trends Immunol. 2019, 40, 984–997. [Google Scholar] [CrossRef] [PubMed]
  242. Ollila, T.A.; Masel, R.H.; Reagan, J.L.; Lu, S.; Rogers, R.D.; Paiva, K.J.; Taher, R.; Burguera-Couce, E.; Zayac, A.S.; Yakirevich, I.; et al. Seroconversion and outcomes after initial and booster COVID-19 vaccination in adults with hematologic malignancies. Cancer 2022, 128, 3319–3329. [Google Scholar] [CrossRef] [PubMed]
  243. Mao, C.; Beiss, V.; Ho, G.W.; Fields, J.; Steinmetz, N.F.; Fiering, S. In situ vaccination with cowpea mosaic virus elicits systemic antitumor immunity and potentiates immune checkpoint blockade. J. Immunother. Cancer 2022, 10, e005834. [Google Scholar] [CrossRef]
  244. You, W.; Ouyang, J.; Cai, Z.; Chen, Y.; Wu, X. Comprehensive Analyses of Immune Subtypes of Stomach Adenocarcinoma for mRNA Vaccination. Front. Immunol. 2022, 13, 827506. [Google Scholar] [CrossRef]
  245. Elizondo, C.R.; Bright, J.D.; Byrne, J.A.; Bright, R.K. Analysis of the CD8+ IL-10+ T cell response elicited by vaccination with the oncogenic tumor-self protein D52. Hum. Vaccines Immunother. 2020, 16, 1413–1423. [Google Scholar] [CrossRef]
  246. Sangeeta, K.; Yenugu, S. Ablation of the sperm-associated antigen 11A (SPAG11A) protein by active immunization promotes epididymal oncogenesis in the rat. Cell Tissue Res. 2022, 389, 115–128. [Google Scholar] [CrossRef]
  247. Rakshit, S.; Adiga, V.; Ahmed, A.; Parthiban, C.; Chetan Kumar, N.; Dwarkanath, P.; Shivalingaiah, S.; Rao, S.; D’Souza, G.; Dias, M.; et al. Evidence for the heterologous benefits of prior BCG vaccination on COVISHIELD™ vaccine-induced immune responses in SARS-CoV-2 seronegative young Indian adults. Front. Immunol. 2022, 13, 985938. [Google Scholar] [CrossRef]
  248. Xu, H.; Zhao, F.; Wu, D.; Zhang, Y.; Bao, X.; Shi, F.; Cai, Y.; Dou, J. Eliciting effective tumor immunity against ovarian cancer by cancer stem cell vaccination. Biomed. Pharmacother. Biomed. Pharmacother. 2023, 161, 114547. [Google Scholar] [CrossRef]
  249. Takeshita, K.; Ishiwada, N.; Takeuchi, N.; Ohkusu, M.; Ohata, M.; Hino, M.; Hishiki, H.; Takeda, Y.; Sakaida, E.; Takahashi, Y.; et al. Immunogenicity and safety of routine 13-valent pneumococcal conjugate vaccination outside recommended age range in patients with hematological malignancies and solid tumors. Vaccine 2022, 40, 1238–1245. [Google Scholar] [CrossRef]
  250. Fitzpatrick, T.; Alsager, K.; Sadarangani, M.; Pham-Huy, A.; Murguía-Favela, L.; Morris, S.K.; Seow, C.H.; Piché-Renaud, P.P.; Jadavji, T.; Vanderkooi, O.G.; et al. Immunological effects and safety of live rotavirus vaccination after antenatal exposure to immunomodulatory biologic agents: A prospective cohort study from the Canadian Immunization Research Network. Lancet Child Adolesc. Health 2023, 7, 648–656. [Google Scholar] [CrossRef]
  251. Mezzapelle, R.; De Marchis, F.; Passera, C.; Leo, M.; Brambilla, F.; Colombo, F.; Casalgrandi, M.; Preti, A.; Zambrano, S.; Castellani, P.; et al. CXCR4 engagement triggers CD47 internalization and antitumor immunization in a mouse model of mesothelioma. EMBO Mol. Med. 2021, 13, e12344. [Google Scholar] [CrossRef]
  252. Jindra, C.; Hainisch, E.K.; Rümmele, A.; Wolschek, M.; Muster, T.; Brandt, S. Influenza virus vector iNS1 expressing bovine papillomavirus 1 (BPV1) antigens efficiently induces tumour regression in equine sarcoid patients. PLoS ONE 2021, 16, e0260155. [Google Scholar] [CrossRef] [PubMed]
  253. Huang, M.; Xiong, D.; Pan, J.; Zhang, Q.; Wang, Y.; Myers, C.R.; Johnson, B.D.; Hardy, M.; Kalyanaraman, B.; You, M. Prevention of Tumor Growth and Dissemination by In Situ Vaccination with Mitochondria-Targeted Atovaquone. Adv. Sci. 2022, 9, e2101267. [Google Scholar] [CrossRef] [PubMed]
  254. Abdolkarimi, B.; Amanati, A.; Molavi Vardanjani, H.; Jamshidi, S.; Tabaeian, S.A.P. Antibody waning after immunosuppressive chemotherapy and immunomodulators, re-immunization considerations in pediatric patients with malignancy and chronic immune thrombocytopenic purpura. BMC Infect. Dis. 2022, 22, 657. [Google Scholar] [CrossRef]
  255. Ota, S.; Miyashita, M.; Yamagishi, Y.; Ogasawara, M. Baseline immunity predicts prognosis of pancreatic cancer patients treated with WT1 and/or MUC1 peptide-loaded dendritic cell vaccination and a standard chemotherapy. Hum. Vaccines Immunother. 2021, 17, 5563–5572. [Google Scholar] [CrossRef]
Figure 1. Three-dimensional bioprinting technology and the comprehensive nanocarrier application platform enable precise antitumor immunotherapy by combining biomaterials and nanotechnology. The platform uses bioprinting technology to accurately manufacture complex three-dimensional structures and load modified lipid calcium and phosphorus nanoparticles onto the vaccine, thereby enhancing the efficiency and specificity of antigen delivery, significantly regulating the tumor microenvironment, and enhancing the antitumor response of the immune system.
Figure 1. Three-dimensional bioprinting technology and the comprehensive nanocarrier application platform enable precise antitumor immunotherapy by combining biomaterials and nanotechnology. The platform uses bioprinting technology to accurately manufacture complex three-dimensional structures and load modified lipid calcium and phosphorus nanoparticles onto the vaccine, thereby enhancing the efficiency and specificity of antigen delivery, significantly regulating the tumor microenvironment, and enhancing the antitumor response of the immune system.
Jfb 15 00229 g001
Figure 2. The nanoparticle carrier successfully penetrated the meninges and targeted and killed tumor cells in a mouse model of brain metastases. Through the modified lipid calcium and phosphorus nanoparticles, the carrier can efficiently deliver antitumor drugs, enhance the drug concentration at the tumor site, effectively destroy the tumor microenvironment, and promote the antitumor response of the immune system.
Figure 2. The nanoparticle carrier successfully penetrated the meninges and targeted and killed tumor cells in a mouse model of brain metastases. Through the modified lipid calcium and phosphorus nanoparticles, the carrier can efficiently deliver antitumor drugs, enhance the drug concentration at the tumor site, effectively destroy the tumor microenvironment, and promote the antitumor response of the immune system.
Jfb 15 00229 g002
Figure 3. Photoacoustic imaging (PAI) of mannan-decorated lipid calcium–phosphorus nanoparticle vaccine in a mouse model. PAI technology uses its high resolution and deep imaging capabilities to clearly show the distribution and targeting effects of this nanoparticle in the body. Studies have shown that this vaccine can precisely accumulate in tumor tissue, significantly enhancing the immune system’s resistance to tumors through enhanced antigen delivery and regulation of the tumor microenvironment.
Figure 3. Photoacoustic imaging (PAI) of mannan-decorated lipid calcium–phosphorus nanoparticle vaccine in a mouse model. PAI technology uses its high resolution and deep imaging capabilities to clearly show the distribution and targeting effects of this nanoparticle in the body. Studies have shown that this vaccine can precisely accumulate in tumor tissue, significantly enhancing the immune system’s resistance to tumors through enhanced antigen delivery and regulation of the tumor microenvironment.
Jfb 15 00229 g003
Figure 4. Schematic illustration of lipid calcium–phosphorus nanoparticles used to measure oxidative stress via photoacoustic imaging (PA). This schematic shows the mechanism of action of nanoparticles in vivo: mannan-decorated nanoparticles target tumor cells to monitor oxidative stress levels at tumor sites in real-time using photoacoustic imaging.
Figure 4. Schematic illustration of lipid calcium–phosphorus nanoparticles used to measure oxidative stress via photoacoustic imaging (PA). This schematic shows the mechanism of action of nanoparticles in vivo: mannan-decorated nanoparticles target tumor cells to monitor oxidative stress levels at tumor sites in real-time using photoacoustic imaging.
Jfb 15 00229 g004
Figure 5. Schematic illustration of mannan-decorated lipid calcium–phosphorus nanoparticles (mannose-LNP-CaP) targeting carcinogenic long noncoding RNAs (lncrnas) for cancer therapy. This figure shows that mannan-decorated nanoparticles can specifically recognize and bind to cancer-causing lncrnas in tumor cells, inhibit their expression and function, and thus block the proliferation and metastasis of tumor cells.
Figure 5. Schematic illustration of mannan-decorated lipid calcium–phosphorus nanoparticles (mannose-LNP-CaP) targeting carcinogenic long noncoding RNAs (lncrnas) for cancer therapy. This figure shows that mannan-decorated nanoparticles can specifically recognize and bind to cancer-causing lncrnas in tumor cells, inhibit their expression and function, and thus block the proliferation and metastasis of tumor cells.
Jfb 15 00229 g005
Figure 6. Summary diagram of clinical trial phase related to nanoparticle vaccine. This chart clearly shows the various stages of nanoparticle vaccine development from the early stage to clinical application, including preliminary safety evaluation (Phase I), dose optimization and immune response analysis (Phase II), large-scale multi-center efficacy and safety validation (Phase III), and continuous post-marketing monitoring and efficacy evaluation (Phase IV).
Figure 6. Summary diagram of clinical trial phase related to nanoparticle vaccine. This chart clearly shows the various stages of nanoparticle vaccine development from the early stage to clinical application, including preliminary safety evaluation (Phase I), dose optimization and immune response analysis (Phase II), large-scale multi-center efficacy and safety validation (Phase III), and continuous post-marketing monitoring and efficacy evaluation (Phase IV).
Jfb 15 00229 g006
Figure 7. Drug resistance factors in cancer and treatment of mannose-modified lipid calcium and phosphorus nanoparticles (mannose-LNP-CAP). This figure shows that cancer drug resistance includes key factors such as the complexity of the tumor microenvironment, gene mutations, and drug efflux pumps. Mannose-LNP-CaP therapy overcomes drug resistance by targeting these resistance mechanisms, especially by precisely regulating the tumor microenvironment, enhancing antigen delivery, and activating immune cells, and significantly improves the efficacy of antitumor immune responses.
Figure 7. Drug resistance factors in cancer and treatment of mannose-modified lipid calcium and phosphorus nanoparticles (mannose-LNP-CAP). This figure shows that cancer drug resistance includes key factors such as the complexity of the tumor microenvironment, gene mutations, and drug efflux pumps. Mannose-LNP-CaP therapy overcomes drug resistance by targeting these resistance mechanisms, especially by precisely regulating the tumor microenvironment, enhancing antigen delivery, and activating immune cells, and significantly improves the efficacy of antitumor immune responses.
Jfb 15 00229 g007
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wu, L.; Yang, L.; Qian, X.; Hu, W.; Wang, S.; Yan, J. Mannan-Decorated Lipid Calcium Phosphate Nanoparticle Vaccine Increased the Antitumor Immune Response by Modulating the Tumor Microenvironment. J. Funct. Biomater. 2024, 15, 229. https://doi.org/10.3390/jfb15080229

AMA Style

Wu L, Yang L, Qian X, Hu W, Wang S, Yan J. Mannan-Decorated Lipid Calcium Phosphate Nanoparticle Vaccine Increased the Antitumor Immune Response by Modulating the Tumor Microenvironment. Journal of Functional Biomaterials. 2024; 15(8):229. https://doi.org/10.3390/jfb15080229

Chicago/Turabian Style

Wu, Liusheng, Lei Yang, Xinye Qian, Wang Hu, Shuang Wang, and Jun Yan. 2024. "Mannan-Decorated Lipid Calcium Phosphate Nanoparticle Vaccine Increased the Antitumor Immune Response by Modulating the Tumor Microenvironment" Journal of Functional Biomaterials 15, no. 8: 229. https://doi.org/10.3390/jfb15080229

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop