Back to Journals » International Journal of Nanomedicine » Volume 20

Applications of mRNA Delivery in Cancer Immunotherapy

Authors Pan X, Zhang YWQ, Dai C, Zhang J, Zhang M, Chen X

Received 12 October 2024

Accepted for publication 8 March 2025

Published 17 March 2025 Volume 2025:20 Pages 3339—3361

DOI https://doi.org/10.2147/IJN.S500520

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Jie Huang



Xiaoyu Pan,1,2,* Yang-Wen-Qing Zhang,1,2,* Caixia Dai,1,2,* Junyu Zhang,1,2 Minghe Zhang,1,2 Xi Chen1,2

1Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China; 2Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Hubei, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Xi Chen, Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, People’s Republic of China, Email [email protected] Minghe Zhang, Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, People’s Republic of China, Email [email protected]

Abstract: Cancer treatment is continually advancing, with immunotherapy gaining prominence as a standard modality that has markedly improved the management of various malignancies. Despite these advancements, the efficacy of immunotherapy remains variable, with certain cancers exhibiting limited response and patient outcomes differing considerably. Thus, enhancing the effectiveness of immunotherapy is imperative. A promising avenue is mRNA delivery, employing carriers such as liposomes, peptide nanoparticles, inorganic nanoparticles, and exosomes to introduce mRNA cargos encoding tumor antigens, immune-stimulatory, or immune-modulatory molecules into the tumor immune microenvironment (TIME). This method aims to activate the immune system to target and eradicate tumor cells. In this review, we introduce the characteristics and limitations of these carriers and summarize the application and mechanisms of currently prevalent cargos in mRNA-based tumor treatment. Additionally, given the significant clinical application of immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR)-based cell therapies in solid tumors (including melanoma, non-small-cell lung cancer, head and neck squamous cell carcinoma, triple-negative breast cancer, gastric cancer) and leukemia, which have become first-line treatments, we highlight and discuss recent progress in combining mRNA delivery with ICIs, CAR-T, CAR-NK, and CAR-macrophage therapies. This combination enhances the targeting capabilities and efficacy of ICIs and CAR-cell-based therapies, while also mitigating the long-term off-target toxicities associated with conventional methods. Finally, we analyze the limitations of current mRNA delivery systems, such as nuclease-induced mRNA instability, immunogenicity risks, complex carrier production, and knowledge gaps concerning dosing and safety. Addressing these challenges is crucial for unlocking the potential of mRNA in cancer immunotherapy. Overall, exploring mRNA delivery enriches our comprehension of cancer immunotherapy and holds promise for developing personalized and effective treatment strategies, potentially enhancing the immune responses of cancer patients and extending their survival time.

Keywords: mRNA immunotherapy, LNP-mRNA, CAR, ICI, tumor microenvironment modulation, personalized medicine

Introduction

Cancer treatment encompasses various therapeutic approaches, such as surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy. Currently, the application of concepts such as comprehensive treatment, precision medicine, and immunotherapy has made tumour treatment more personalized and effective.1,2 Tumour immunotherapy is a therapeutic approach that stimulates or controls the immune system to target and destroy cancerous cells. Tumour immunotherapy encompasses various therapeutic approaches, including cancer vaccines, immune checkpoint inhibitors (ICIs) based on monoclonal antibodies, chimeric antigen receptor (CAR) T-cell immunotherapy, etc.3 These therapeutic strategies aim to reactivate and sustain the tumour immune response, thereby restoring the body’s intrinsic antitumour immune mechanisms and improving the management and eradication of cancers.

Despite significant progress in cancer treatment, several challenges and limitations are encountered in tumour immunotherapy. First, the efficacy of immunotherapy varies greatly among different types of cancer and even among patients with the same type of cancer, which is closely related to the genetic and molecular characteristics of the patients.4 Immune escape is one of the main reasons for this.5 Tumor cells exhibit high expression levels of programmed death-ligand 1 (PD-L1), which binds to programmed death-1 (PD-1) receptors on T cells, thereby inhibiting their tumor-cytotoxic functions.6 The population of regulatory T cells (Tregs) also increases within the tumor microenvironment, where PD-1 on these cells interacts with PD-L1 on tumor cells or antigen-presenting cells, further enhancing immunosuppression.7 Additionally, M2 macrophages secrete immunosuppressive cytokines that inhibit T cell activity and promote tumor angiogenesis.8 Within the tumor-associated neutrophils, the N2 subtype releases reactive oxygen species and arginase, leading to damage of immune cells and depletion of arginine.9 Collectively, these immune evasion mechanisms undermine the efficacy of immunotherapy to a certain extent. Second, in traditional immunotherapy, high doses of immune stimulants or antibodies are often used to activate the immune system. This method lacks specificity for particular tumour cells and may cause harm to normal tissues which may lead to severe adverse events.10

Messenger RNA (mRNA) delivery is a strategy that involves the direct administration of mRNA into cells to induce the production of specific proteins, thereby enabling targeted therapeutic intervention for a range of diseases. The mRNA molecules used in delivery can encode any protein required by humans, including tumour-associated antigens (TAAs), immunostimulatory factors, and immune regulatory molecules, thereby achieving the objectives of various therapies. Moreover, mRNA molecules demonstrate high safety, favorable degradability, and the advantage of not integrating into the genome, making them ideal carriers for therapeutic applications.11,12

In recent years, mRNA delivery systems have made significant strides in clinical development. The Pfizer-BioNTech and Moderna COVID-19 vaccines, which utilize this technology, received Emergency Use Authorization (EUA) and full approval from the Food and Drug Administration (FDA).13 These vaccines employ lipid nanoparticles to deliver mRNA encoding the genetic information for the SARS-CoV-2 spike protein into human cells, thereby eliciting an immune response. Furthermore, Moderna’s mRNA-1273 and BioNTech’s BNT162b2 demonstrated outstanding efficacy in COVID-19 clinical trials.14 These instances underscore the potential and reliability of mRNA delivery systems.

The methods of mRNA delivery technology mainly include nanotechnology and cell engineering.15,16 Nanotechnology refers to the utilization of nanomaterials, such as liposomes, polymers, metals, and carbon, to encapsulate or load mRNA molecules, forming mRNA nanoparticles (NPs). These NPs can be administered into the human body via intravenous injection, intramuscular injection, nasal inhalation, or other routes to deliver mRNA into specific locations.17,18 Cell engineering involves the transfection of mRNA molecules into ex vivo cultured human cells (such as dendritic cells (DCs), lymphocytes, and macrophages) through methods such as electroporation, gene guns, and viral vectors, followed by reinfusion of the transfected cells back into the human body.16,19

The application of mRNA delivery in tumour immunotherapy has significant advantages. First, through the personalized design and targeting of specific cancer types with mRNA sequences, a more effective immune response can be induced to improve treatment outcomes.12 Second, mRNA delivery technology can efficiently and accurately deliver mRNAs encoding tumour-related antigens or other immune molecules into cells, triggering a specific immune attack on tumour cells. This precision allows mRNA delivery to identify and eliminate tumour cells effectively without damaging normal cells.11,12 Additionally, compared with DNA vaccines, mRNA does not need to enter the nucleus for transcription, avoiding the potential risks associated with genomic integration, thus enhancing safety. It can also be translated rapidly in the cytoplasm, enabling a faster immune response.20 In contrast to siRNA, mRNA has a broader range of applications. mRNA can encode a variety of antigens and immune-stimulating molecules, and it can customize personalized treatment plans according to different tumor types and the individual conditions of patients. However, siRNA is limited to silencing known disease-causing genes, and its applicability is poor when there are no clear disease-causing genes or when it is necessary to enhance the immune response.21

In the realm of tumor immunotherapy, numerous drug studies leveraging mRNA are being actively pursued, with many already in the clinical trial phase. Through in-depth exploration and meticulous curation on the authoritative platform ClinicalTrials.gov, we have selected several highly representative mRNA-based cancer treatment drugs, aiming to offer valuable references and insights for related research and applications (Table 1).

Table 1 Representative Clinical Trials of mRNA-Based Cancer Therapeutics. All the Data in the Table are Sourced from ClinicalTrials.gov

In this article, we present an overview of mRNA delivery strategies and the most commonly used carriers, including liposomes, peptide NPs, inorganic NPs, and exosomes. We discuss how researchers design mRNA constructs encoding diverse molecules, including tumour-associated antigens (TAAs), immune stimulatory factors, and immunoregulatory molecules, to be delivered into the body for cancer treatment. Furthermore, we explore the synergistic potential of combining mRNA delivery with ICIs to enhance their efficacy in cancer treatment. Additionally, we examine the application of mRNA delivery in CAR cell therapy, focusing on how mRNA delivery enables transient CAR expression to improve the targeting ability and effectiveness of CAR-T, CAR-engineered natural killer (NK) (CAR-NK), and CAR-engineered macrophage (CAR-M) cells while overcoming the long-term off-target toxicity associated with traditional methods. Finally, we analyse the limitations of current mRNA delivery methods and propose future directions to enhance their role in immunotherapy.

mRNA Delivery Carriers

In mRNA delivery systems for cancer therapy, the selection of an appropriate delivery vehicle is crucial to ensure effective cellular uptake and achieve the desired therapeutic outcomes. This chapter provides a detailed exploration of several major delivery vehicles, including liposomes (Figure 1a), peptide NPs (Figure 1b), inorganic NPs (Figure 1c), exosomes (Figure 1d) and other carriers such as polymersomes, dendrimers, and polymer micelles (Figure 1e). Each of these carriers possesses distinct physicochemical properties and biocompatibility, enabling them to serve various therapeutic needs. Liposomes, being among the earliest delivery vehicles, have a structure similar to that of cell membranes, facilitating mRNA membrane fusion and internalization.22,23 Peptide NPs and inorganic NPs are notable for their excellent stability and modifiable surface properties, showing broad application prospects in preclinical studies.24,25 Owing to their natural biocompatibility and targeting capabilities, exosomes have emerged as promising delivery systems.26 Additionally, novel carriers such as polymersomes, dendrimers, and polymer micelles are gaining attention because of their high controllability and targeting potential.27,28 The following sections delve into the characteristics of these different mRNA delivery vehicles.

Figure 1 mRNA delivery carriers. mRNA delivery carriers include liposomes (a), peptide nanoparticles (b), inorganic nanoparticles (c), exosomes (d) and others (e). Created in BioRender. Dai, C. (2025) https://BioRender.com/u23s018.

Liposomes

Liposomes are an early version of lipid NPs (LNPs), and they have become a widely used nanocarrier platform because of their ability to facilitate the transportation of both hydrophobic and hydrophilic molecules, including very small molecules as well as nucleic acids.29 Liposomes are made up of several key components (Figure 2). Ionic lipids are essential because they form stable complexes with negatively charged mRNAs, facilitating their intracellular release. Helper lipids increase the structural stability of liposomes and aid in liposome fusion with cellular membranes. Cholesterol increases the stability and fluidity of liposomes, improving their formation and mRNA delivery efficiency. Finally, polyethylene glycol (PEG)-modified lipids create a “stealth layer” on the surface of liposomes, allowing them to evade immune system recognition and clearance. Compared with other delivery methods, the encapsulation of mRNA in liposomes increases stability and efficacy, improves delivery efficiency, and reduces unnecessary immune responses.29,30

Figure 2 The key components of Liposomes. Liposomes consist of several key components, each with distinct functions. Charged ionizable lipids electrostatically interact with negatively charged mRNA, enabling its encapsulation into liposomes and promoting cell entry. Neutral helper lipids stabilize the bilayer structure, optimize morphology, and prevent aggregation. Cholesterol regulates membrane fluidity and ensures structural integrity during circulation and aids in cell contact. PEG-modified lipids form a hydrophilic surface layer, reducing immune recognition and clearance, prolonging circulation time, and enhancing delivery efficiency. Created in BioRender. Dai, C. (2025) https://BioRender.com/z14b486.

In recent studies, liposomes have been further advanced as mRNA delivery vehicles through the surface modification of liposomes, enabling targeted delivery of mRNA to specific cells. For example, Joel et al modified the surface of liposomes with anti-CD5 antibodies, precisely targeting T cells and successfully reprogramming them in vivo.31 Researchers injected Ai6 mice carrying the Rosa26CAG-LSL-ZsGreen gene with 30 µg of non-targeted/LNP-Cre (abbreviated as NT), IgG/LNP-Cre, or CD5/LNP-Cre. After 24 hours, the expression of ZsGreen protein was observed in 81.1% of CD4+ cells and 75.6% of CD8+ cells. Farasat A et al demonstrated that oligoclonal NPs exhibit greater cytotoxicity towards HER2-positive tumour cells than nontargeted liposomes do. The oligoclonal targeting of liposomes is thus regarded as a promising strategy for treating breast cancer characterized by HER2 overexpression.32 Additionally, Goswami R et al developed a panel of LNPs in which the surface is modified with mannan, revealing elevated levels of IgG1 and IgG2a in mannose-sylated lipid NPs (MLNPs) relative to nonmodified LNPs. This study confirmed the increased efficacy of mannosylated self-amplifying RNA (SAM) LNPs for the delivery of RNA vaccines compared with nonglycosylated delivery systems.33 Current research directions include developing novel lipid materials to improve delivery efficiency and safety, designing targeted delivery systems to increase tissue specificity, and exploring the potential applications of liposomes in treating other diseases. As these technologies continue to advance, liposome-encapsulated mRNA delivery is expected to play a greater role in vaccine development, gene therapy, and other biopharmaceutical fields.

Peptide NPs

The formation of peptide NPs is based on the self-assembly process of peptide molecules, which spontaneously assemble into stable nanostructures via noncovalent interactions, including electrostatic interactions, hydrophobic interactions, and hydrogen bonding.24 mRNA can be combined with self-assembled peptide NPs either by direct mixing or by being encapsulated within the particles during the peptide self-assembly process.34 Peptide NPs can be designed to facilitate cellular uptake, thus improving delivery efficiency. Additionally, by incorporating specific cell-targeting signals into peptide sequences, targeted delivery to particular cell types or pathological tissues can be achieved.35 Owing to its unique advantages and potential, the technique of encapsulating mRNAs within peptide NPs has become a research focus in the fields of gene therapy and vaccine development.

Biocompatibility is a crucial factor for any delivery system, and peptide nanoparticles excel in this regard. Their components are usually derived from natural or biocompatible synthetic peptides, which minimizes the possibility of triggering adverse immune responses when introduced into the body. This enables them to integrate more smoothly with biological systems and reduces the risks of rejection or inflammation. Peptides have been widely explored for gene delivery purposes because of their ability to be synthesized as uniform single-molecule entities.36–38 Peptides are inherently biodegradable and offer precise control over various properties, including charge distribution, hydrophobicity, and structural geometry.36,39 Cell-penetrating polypeptides offer the added benefit of facilitating folding-dependent complexation, which allows them to shield nucleic acids through secondary and tertiary interactions, thereby protecting the nucleic acids from enzymatic breakdown. By adjusting these interactions, it is possible to reduce aggregation and achieve size distributions within a narrow range, with 50–300 nm typically considered optimal.40,41 Furthermore, scientists have made it possible to design purely synthetic peptide sequences by investigating the cell-penetrating motifs associated with cellular uptake and their relationship with nucleic acid packaging.42,43

However, despite these advantages, the development of peptide NPs also faces several design challenges. One significant concern is their stability in vivo. Once administered, peptide NPs encounter a complex physiological environment that can potentially disrupt their nanostructure. For example, enzymatic degradation in the bloodstream or within cells may lead to the premature release or breakdown of the encapsulated mRNA. Moreover, changes in pH and ionic strength can also impact the noncovalent interactions that hold the peptide NPs together, affecting their overall stability and functionality.44 Overcoming these challenges requires careful optimization of peptide sequences, chemical modifications, and formulation strategies to ensure that peptide NPs can maintain their integrity and effectively deliver mRNA to the target site.45 With a deeper understanding of peptide design and nanotechnology, along with advances in production and purification techniques, this delivery system is expected to play a progressively significant role in the future of biomedicine.46,47

Inorganic NPs

Inorganic NPs mainly include nonmetal NPs, metal NPs, and oxide NPs, among other types.25 Among the most representative NPs are silica NPs, which have been extensively studied for use in mRNA delivery because of their unique physicochemical properties and biocompatibility.48 As an inorganic material, silica offers several advantages. First, the high surface area of silica enables the loading of a larger amount of mRNA molecules.49 Second, the release rate of mRNA can be controlled by adjusting the pore structure of the silica NPs, enabling controlled release during delivery.50 Third, surface modifications (eg, covalent bonding of targeting ligands) can improve the ability of silica NPs to target specific cell types or pathological tissues. Moreover, silica is degradable and biocompatible.51 Although silica NP-encapsulated mRNA delivery technology has shown tremendous application prospects, further research is needed to determine the in vivo distribution and clearance of silica NPs. Improper distribution may lead to accumulation in nonspecific tissues, whereas inefficient clearance could pose long-term biosafety issues. The future holds the promise of existing challenges being overcome to achieve safer, more effective, and smarter nucleic acid delivery solutions.52,53

The use of metal-core NPs as targeted delivery carriers has gained increasing popularity because of their ability to shield RNA payloads and direct them to their intended destinations.54,55 The biological barriers present in vivo necessitate size requirements for drug delivery carriers; to evade recognition by the human immune system and enter cellular environments, carriers must be very small (<100 nm). Iron oxide and gold-core NPs can be synthesized under highly controlled conditions to produce uniform, ultrasmall drug delivery carriers capable of circumventing critical biological barriers. Additionally, unlike virus-based transfection reagents, the biodistribution of metal-core NPs can be monitored via noninvasive imaging techniques that leverage the interaction between electromagnetic radiation and the inorganic atoms of the NP core.56 Furthermore, metal-core NPs have demonstrated transfection efficiencies comparable to those of traditional RNA delivery carriers while having the advantages of lower immunogenicity and minimal side effects, especially when tumour-targeting ligands are conjugated to their surfaces.57,58

Exosomes

Exosomes are extracellular vesicles containing components of the cells that secrete them, such as DNA and RNA. These exosomes can be absorbed by distant cells, thereby influencing the functions and behaviours of the absorbing cells.26 The advantage of the use of exosomes as mRNA delivery vehicles lies in their natural origin and biological activity. Derived from their ability to be secreted by cells, exosomes possess low immunogenicity and toxicity, thereby reducing the risk of side effects during the treatment process.59 Furthermore, the surface of exosomes is enriched with proteins, lipids, carbohydrates, and other biomolecules, which can serve as recognition signals to guide the precise targeting of exosomes to specific cells or tissues.60 During the process of mRNA delivery, exosomes protect mRNAs from degradation and promote their cellular uptake. The double-layer membrane structure of exosomes provides a stable protective shell for mRNAs, preventing their enzymatic degradation in vivo. Simultaneously, exosomes release mRNAs into target cells through membrane fusion, achieving efficient and safe delivery. Additionally, exosomes exhibit excellent drug-loading capacity.61 Scientists can modify the surface molecules or alter the internal environment of exosomes to load different types and quantities of drugs. This modification can enable exosomes to carry other therapeutic molecules alongside mRNA, facilitating multifunctional combinations.

However, despite these remarkable advantages, the scalability of exosome production remains a significant hurdle for clinical translation. Currently, the isolation and purification methods of exosomes are complex, time-consuming, and often yield relatively low amounts of exosomes. For example, ultracentrifugation, a commonly used isolation technique, requires expensive equipment and skilled operators, and the process is not easily scalable.62 Alternative methods like size-exclusion chromatography and immunoaffinity capture also have their own limitations in terms of throughput and cost.63 Moreover, even if exosomes can be produced in larger quantities, ensuring the consistency and quality of each batch becomes another challenge. Variations in the source cells, culture conditions, and isolation procedures can all lead to differences in exosome properties, which may affect their performance as mRNA delivery vehicles.64,65 Tackling these scalability issues demands the development of innovative and more efficient production, isolation, and quality control strategies to make exosome-based therapies a viable option in the clinic. In conclusion, exosomes possess unique advantages and potential as mRNA delivery vehicles. The natural ability of exosomes to achieve efficient and safe mRNA delivery provides exciting new possibilities in the development of cancer immunotherapy.66,67

Others

In addition to the carriers mentioned above, many other carriers also have high application potential. Polymersomes, dendrimers, and polymer micelles are widely studied polymeric nanocarriers that are primarily used in drug delivery systems. These nanocarriers have garnered significant attention from the scientific community because of their unique properties and potential applications. Polymersomes are vesicular structures formed by the self-assembly of amphiphilic block copolymers, and they resemble liposomes in their structure.27 Polymersomes consist of a bilayer polymer membrane that can encapsulate and protect drugs, thereby increasing drug stability. Compared with traditional liposomes, polymersomes exhibit increased stability and tunable permeability, making them highly useful in drug delivery systems.68,69 Polymersomes can encapsulate both hydrophobic and hydrophilic drugs, and their surfaces can be modified for targeted delivery and controlled release.

Dendrimers are highly branched macromolecules that typically originate from a core molecule and are constructed by sequential chemical reactions that add hierarchical branching structures. The size, shape, and surface functionality of dendrimers can be precisely controlled. Owing to their high loading capacity and ease of modification, dendrimers are ideal drug delivery carriers, enabling targeted delivery through multiple functional groups on their surface.70–72 One of the most remarkable unique advantages of dendrimers lies in their extremely high surface area to volume ratio. This characteristic enables them to carry a large number of functional groups, not only facilitating their modification with various ligands for targeted delivery but also allowing for enhanced drug loading.73 For instance, dendrimers can be conjugated with specific antibodies or peptides that recognize and bind to receptors overexpressed on tumor cells, achieving precise tumor-targeted drug delivery.74 Moreover, their unique structure endows them with the ability to penetrate biological membranes more effectively compared to other carriers.75 This is particularly beneficial in applications such as gene therapy, where efficient intracellular delivery of nucleic acids is crucial. In addition to drug and gene delivery, dendrimers’ functionalization potential extends to other biomedical fields. They can be engineered to serve as imaging agents, by incorporating fluorescent or radioactive substances, enabling real-time monitoring of drug distribution and therapeutic efficacy in vivo.76 Their versatility and multi-functionality make them a promising candidate in modern biomedical research.

Polymer micelles are nanoscale micellar structures formed by the self-assembly of amphiphilic block copolymers. Polymer micelles consist of a hydrophobic core and a hydrophilic shell, allowing them to remain stable in aqueous environments. These nanocarriers provide effective solubilization and protection of drugs, especially hydrophobic drugs. Polymer micelles are commonly used for the delivery of poorly soluble drugs, and their structure can be adjusted to achieve sustained release and targeted delivery.28,77,78 In summary, as advanced drug delivery carriers, polymersomes, dendrimers, and polymer micelles offer new possibilities for increasing drug bioavailability, stability, and efficacy. With the advancement of nanotechnology, these nanocarriers are expected to be more widely applied in mRNA delivery and to play a significant role in clinical cancer therapy in the future.

mRNA Delivery Cargos Related to Cancer Immunotherapy

mRNAs, as versatile therapeutic tools, can increase the ability of the immune system to target cancer by encoding various biomolecules (Figure 3). Once delivered into the body, mRNAs can encode tumour-associated antigens, thereby promoting recognition by the immune system and the attack of cancer cells—a strategy commonly employed in mRNA vaccines.79 Additionally, mRNAs can encode immunostimulatory molecules such as interleukins (ILs) and granulocyte-macrophage colony-stimulating factor (GM-CSF), which effectively activate and improve the functionality of immune cells, increasing their cytotoxicity against cancer cells.48,80 Moreover, mRNAs can also encode immunomodulatory molecules to further optimize the immune response, ensuring the effectiveness and durability of the immune system’s fight against cancer.81 By delivering mRNAs that encode different molecules, we can achieve more precise and potent therapeutic effects in cancer immunotherapy.

Figure 3 mRNA delivery cargos and applications of mRNA delivery therapy. Created in BioRender. Dai, C. (2025) https://BioRender.com/l82a560.

Tumour-Associated Antigens

Tumour-associated antigens (TAAs) are antigen molecules present on both tumour and normal cells that are highly expressed during tumour cell proliferation.82 The use of mRNA vaccines to carry TAAs to activate the immune system has become a feasible approach for cancer treatment.83 An mRNA vaccine is a particular type of vaccine in which mRNA is used to transmit genetic instructions that encode TAAs.84 For example, in one study, mRNA vaccines were used to deliver TAAs to antigen-presenting cells (APCs), which include DCs. After taking up the mRNA vaccines encoding TAAs, DCs process TAAs into antigenic peptides and display them on MHC class I molecules. CD8+ T cells recognize these complexes through TCRs, which is the primary signal for activation. After activation, CD8+ T cells differentiate into CTLs, which can kill cancer cells expressing specific TAAs.85 The co-stimulatory signals (the second signal) provided by molecules such as CD80 and CD86 on DCs work in concert with antigen recognition to enhance the activation of CD8+ T cells.86 The interaction between the two forms the backbone of anti-tumor immunity and enables the targeted attack on cancer cells. Additionally, CD4+ T cells can simultaneously activate antigen-specific B cells, inducing an immunological response in the body’s fluids and achieving a sustained therapeutic reaction due to immune memory.87 Currently, cancer immunotherapy targeting TAAs has entered a popular phase, with TAAs being among the leading research targets in solid tumour treatments.79 Cytokeratin 19 (CK19) is a protein distributed on the cell membrane, and it is also commonly found in lung cancer cells, where it serves as a specific antigen for immune therapy.88,89 In one study, the authors designed a cationic liposome-protamine complex (LPC) for delivering mRNA encoding CK19, thereby triggering an immune response that targets and inhibits the growth of tumours. The results showed that LPC/mRNA efficiently facilitated the internalization of antigens by DCs, induced the maturation of DCs, and augmented the release of cytokines, thus eliciting an inner-cell immunological response. Overall, this study revealed that LPC/mCK19 is a promising mRNA delivery therapeutic.90

In addition to presenting TAAs to APCs, mRNA vaccines require additional activation signals for APCs to be effective. The efficacy of mRNA vaccine administration relies on the uptake of mRNAs by APCs through phagocytosis and the ability of mRNAs to stimulate the maturation of APCs. In one study, Sandra van Lint et al used codelivery of TAA mRNA with TriMix (a mixture of mRNAs encoding a CD40 ligand, constitutively active Toll-like receptor 4, and CD70), and the results showed that this method induced an environment that attracted and stimulated T cells, including the process of enlisting CD4+ and CD8+ T cells, as well as cytotoxic T lymphocytes (CTLs), that specifically target different TAAs.91

In a study by Chen et al, the authors investigated the use of LNPs for targeting lymph nodes in the delivery of mRNA cancer vaccines. The authors demonstrated that this approach effectively directs the mRNA vaccine to lymph nodes, leading to robust activation of CD8+ T cells. LNP-mediated delivery significantly improves the immune response by inducing a strong CD8+ T-cell response against cancer cells. This targeting strategy not only increases vaccine efficacy but also optimizes immune activation in lymphoid tissues, which is crucial for generating a potent antitumour immune response. The findings of this study underscore the potential of LNP-based mRNA vaccines in advancing cancer immunotherapy by effectively leveraging lymph node targeting to boost immune responses.92

The method of administering mRNA tumour vaccines is a critical factor in ensuring their effectiveness and safety. Depending on the specific type of vaccine and the treatment goals, various administration methods can be chosen, such as subcutaneous injection, intravenous injection, and intramuscular injection.93–95 Administration through the transfection of DCs is also a promising approach that can leverage the crucial role of DCs in the immune system to achieve targeted delivery and efficient expression of the vaccine in the body.96 For example, a Phase 1 trial was conducted to evaluate the effectiveness of monocyte-derived DCs that were exposed to tumour RNA in children and young adults with brain cancer. Additionally, nasal administration has been proposed as a potentially effective vaccination strategy.97

Immune Stimulatory Factors

Immune stimulatory factors, such as ILs, interferons (IFNs), tumour necrosis factors (TNFs), and CSFs, are a group of molecules capable of activating and improving the body’s immune response. By interacting with cells or molecules within the immune system, immune stimulatory factors trigger the activation and multiplication of immune cells, thereby improving the body’s immunological response.98 In the exploration of new strategies for cancer immunotherapy, the use of mRNAs encoding immune stimulatory factors as therapeutic agents has emerged as a focal area of interest. The introduction of mRNAs encoding specific immune stimulatory factors into the body enables the intracellular expression and release of the corresponding cytokines. These immune stimulatory factors can bind to receptors on immune cells, promoting the activation and proliferation of these cells and thus improving the body’s ability to recognize and eliminate tumour cells.

Interleukins

The IL family comprises a collection of cytokines that are synthesized by multiple cells, and these ILs bind to specific receptors, transmit information, and activate immune cells. Among them, IL-2, IL-12, IL27, IL-15, and IL23, as well as others, have been widely studied and applied to immunotherapy for cancer.

IL-2 was one of the earliest ILs discovered. Through its stimulatory and activating effects on effector lymphocytes, IL-2 plays an essential role in immune homeostasis.99 By delivering IL-2 mRNA, the immune system can be activated to target and destroy cancerous cells. A previous study described the progress of the delivery of tumour-activated IL-2 mRNA by LNPs. The researchers prepared ionizable lipid U-101-derived NPs (U-101-LNPs), and through in vitro transcription, they designed and synthesized IL-2F mRNA encoding a fusion protein composed of the linker IL-2 and CD25 (IL-2Rα). The results showed that the U-101-LNP/IL-2F mRNA formulation exhibited effective antitumour activity and safety, demonstrating its potential applicability in clinical tumour immunotherapy.100 In another study, intratumoral injection of porous silica NP (PSNP) IL-2 mRNA into mouse tumour models stimulated immunogenic cancer cell death in tumour-bearing mice, reflecting the potential application of IL-2 mRNA delivery in tumour immunotherapy.48

IL-12 is a powerful immunostimulatory factor that can activate NK cells and T cells, inducing a strong antitumour effect.101 In a MYC oncogene-driven hepatocellular carcinoma model, IL-12 mRNA LNPs exhibited significant antitumour effects. Treatment with IL-12-LNPs significantly reduced the liver tumour burden, delayed tumour progression, and increased survival rates in MYC-induced hepatocellular carcinoma (HCC) transgenic mice. By activating and increasing the activity of immune cells, such as by increasing the infiltration of immune cells, such as helper CD3+CD4+ T cells, into the tumour, IL-12-LNPs effectively suppressed tumour growth and metastasis.102 IL27 is also an important research direction in cancer studies.103 Researchers have used LNPs as a delivery vehicle to directly administer mRNAs encoding IL-12 and IL-27 into tumours, leading to the death of tumour cells and the suppression of tumour expansion. Furthermore, the combined use of IL-12 and IL-27 mRNAs further improved the therapeutic effect.104

Di Trani CA et al investigated the antitumour effects of intratumoral injection of IL-12-encoding mRNAs in Ringer’s lactate solution. The researchers verified mRNA expression in tumour tissues by injecting luciferase (LUC)-encoding mRNAs. The in vivo results demonstrated that different doses of IL-12 mRNA (ranging from 10 μg to 0.5 μg) significantly increased IL-12p70 levels in tumour tissue compared with those in control tissue. In the B16-OVA tumour model, local injection of IL-12 mRNA at various doses had pronounced antitumour effects, with tumours in the treated sites completely disappearing in at least 50% of the mice and notable suppression of distant untreated tumours. All three doses (10 μg, 6 μg, and 0.5 μg) of IL-12 mRNA significantly increased overall survival, with the three-injection regimen being more effective than the single- or double-injection regimens. Overall, even the lowest dose of 0.5 μg of mRNA achieved effective antitumour outcomes.105

IL-15 has various functions, including determining T-cell responses and activating NK cells.106 By delivering IL-15 mRNA, the antitumour activity of T cells can be improved. In one study, researchers prepared a cationic liposome–protamine complex system (CLPP) that efficiently delivered mRNA encoding IL-15 to C26 cells. The CLPP/mIL-15 complex, whether administered locally or systemically, effectively suppressed the growth of tumours in various C26 mouse colon cancer models.107 A group of researchers have developed a nanoplatform called DPPA/IL-15 NPs. This nanoplatform was designed to specifically target PD-L1 in tumours and deliver IL-15 mRNA. The release of IL-15 mRNA resulted in a powerful immune response throughout the body and effectively suppressed tumour growth.108

Christian Hotz et al investigated the therapeutic potential of intratumoral injection of a cytokine–mRNA mixture, including IL-12sc, GM-CSF, IFN-α, and IL-15 sushi, for antitumour activity. The researchers reported that repeated injections of this cytokine–mRNA mixture led to substantial control of tumour growth and complete regression in the majority of B16F10 and CT26 tumour-bearing mice. A single injection was also effective in controlling tumour growth in the B16F10 model mice. The mRNA-encoded cytokines were expressed intratumorally and increased serum cytokine levels. In vivo experiments confirmed that the four-component mixture had superior antitumour effects and overall survival compared with single cytokine mRNA treatments, with complete tumour regression observed in 6 of 10 mice treated with the full cytokine mixture. Additionally, successful treatment led to resistance to tumour rechallenge, suggesting the development of immunologic memory. Antibody-mediated depletion studies showed that CD8+ T cells were the most critical for antitumour effects, followed by CD4+ T cells and NK cells.109

In addition, in a previous study, Hewitt SL, Bai A, and others provided a detailed introduction on how the cytokines IL-23, IL-36γ, and OX40L interact with immune cells and affect them to promote antitumour immune responses when encoded by mRNAs and delivered to the tumour microenvironment (TME).110 Currently, an increasing number of ILs are being used in antitumour immune research, which not only demonstrates the potential of IL mRNA delivery-based cancer therapy but also provides a direction for future anticancer treatments.

However, it is crucial to note the potential side effects associated with interleukin - based immunotherapies. One significant concern is the occurrence of inflammatory cytokine storms. When interleukins are delivered or over - expressed during immunotherapy, they can lead to an excessive activation of immune cells. These over - activated immune cells then initiate a cascade of cytokine release.111 Normally, the body has regulatory mechanisms in place to control cytokine levels. But with the potent stimulatory effect of certain interleukins in immunotherapies, this balance can be disrupted. The massive release of cytokines overwhelms the body’s normal regulatory systems, potentially leading to life - threatening conditions such as systemic inflammation, fever, organ damage, and other severe symptoms.112 To alleviate these adverse effects, dose modulation represents a direct and efficacious strategy. By meticulously adjusting the dosage of delivered interleukin - related mRNA, an optimal equilibrium can be achieved between attaining potent antitumour efficacy and minimizing the potential risk of cytokine storms. Another viable approach entails the combination of interleukins with other pharmacologic agents. For instance, certain drugs capable of modulating the immune response, such as corticosteroids or specific monoclonal antibodies, can be administered concomitantly.113 This combinatorial regimen serves to attenuate the excessive immune activation instigated by interleukins.

GM-CSF and IFN-γ

GM-CSF is a crucial cytokine that is involved primarily in stimulating and regulating the generation, differentiation, and function of granulocytes and macrophages.114 Currently, GM-CSF mRNA can be injected into the body together with mRNA vaccines as a component to increase the immunogenicity of tumour vaccines. In one study, researchers used SV40 large T antigen (TAg) as a model tumour/autoantigen, and the experimental results in transgenic mouse prostate cancer (TRAMP) demonstrated that GM-CSF increased the ability of mRNA vaccines to treat established spontaneous tumours.80 GM-CSF mRNA can also be used in combination with cytokine-encoded mRNAs to increase the efficacy of tumour immunotherapy. In a previous study, Liu et al used cytokine-encoded mRNA-loaded LNPs for anticancer therapy, and the results showed that GM-CSF mRNA exhibited a synergistic effect with cytokine-encoded mRNA. The combined use of GM-CSF mRNA with IL12 and IL27 mRNAs improved the anticancer therapeutic effect.104 When it comes to the delivery of GM-CSF mRNA, the choice of delivery vehicle plays a significant role. LNPs can protect GM-CSF mRNA from degradation by encapsulating it, thus enhancing its stability during in vivo transport. Their size and surface properties can also influence the bioavailability of GM-CSF. Smaller LNPs with appropriate surface modifications may facilitate better cellular uptake, leading to more efficient translation of GM-CSF mRNA and increased local and systemic bioavailability of the cytokine.115

IFN-γ plays a crucial role in the activation of cellular immunity and the subsequent stimulation of antitumour immune responses.116 Researchers have demonstrated, using IFN-γ as an example, that the cell-enhancing RNA system (CERS) mediates mRNA therapy to induce an approximately 40% increase in translation efficiency and captures approximately 40% of unstable IFN-γ protein, resulting in a doubled duration of IFN-γ activity. Regarding the delivery aspect, CERS not only improves the translation efficiency but also impacts the stability and bioavailability of IFN-γ. The unique structure of CERS provides a protected microenvironment for IFN-γ mRNA, preventing its premature degradation. Moreover, the CERS and cells promotes the internalization of IFN-γ mRNA, enhancing its bioavailability. When applied to immunotherapy for osteosarcoma, CERS-mediated IFN-γ mRNA delivery effectively reprogrammed tumour-associated macrophages (TAMs) into tumoricidal macrophages and significantly inhibited tumour growth through the induction of antitumour immunity. Notably, CERS-mediated IFN-γ mRNA delivery combined with PD-L1 monoclonal antibody (αPD-L1) therapy remarkably led to the disappearance of highly malignant pulmonary metastases.117

There are also some potential immunostimulatory molecules that can be used for mRNA delivery to promote immunotherapy. Chemokines such as CXC chemokine ligand (CXCL9/10) and tumor necrosis factor-α (TNFα) have emerged as highly promising research directions in the future. Firstly, CXCL9/10, belonging to the chemokine family, can recruit and activate cytotoxic T lymphocytes and natural killer cells to the tumor site, enhancing the anti-tumor immune response.118 When delivered via mRNA, their sustained local expression in the tumor is expected to improve the infiltration and killing effect of immune cells. Secondly, TNFα is capable of activating macrophages and enhancing their cytotoxicity to directly attack cancer cells.119 It can also induce apoptosis of tumor cells. In combination with mRNA delivery technology, it may enhance its local effect at the tumor site while reducing systemic side effects. Current studies have provided preliminary evidence of their potential, and further investigations are warranted to fully explore their therapeutic applications in cancer treatment.

Immunomodulatory Molecules

Under normal circumstances, the primary function of immune cells is to recognize and attack pathogens and abnormal cells,120 thereby inhibiting the growth and spread of tumours. However, in certain special circumstances, immune cells may also promote tumour development. The delivery of mRNAs encoding immunoregulatory molecules to suppress the protumour effects of immune cells offers a new strategy in the fight against cancer.

Tumour-associated macrophages (TAMs) constitute one of the primary types of immune cells infiltrating tumours, constituting 30–50% of the stromal cells in the TME.121 TAMs are generally classified into two functionally distinct subtypes: classically activated M1 macrophages and alternatively activated M2 macrophages. M1 macrophages typically exert antitumour effects by directly mediating cytotoxicity and antibody-dependent cellular cytotoxicity (ADCC) to kill tumour cells. In contrast, M2 macrophages can promote tumorigenesis and metastasis, suppress T-cell-mediated antitumour immune responses, increase tumour angiogenesis, and thereby contribute to tumour progression.122 TAMs display an immunosuppressive M2-like phenotype in advanced malignancies. TAMs play pivotal roles in promoting tumour development, invasion, migration, angiogenesis, and immune suppression.123 Therefore, TAM-targeted therapies are particularly important in anticancer strategies. By modulating or eliminating the immunosuppressive functions of TAMs, the ability of immune cells to attack tumour cells can be restored, thereby inhibiting tumour growth and spread. The application of TAMs as targets for cancer treatment holds promise for overcoming the limitations of traditional tumour therapy methods and achieving significant therapeutic effects.124

Reprogramming these TAMs to the M1 phenotype may inhibit their protumorigenic activities and unleash antitumour immunity. To explore this therapeutic strategy, researchers are utilizing nanomedicine carriers to deliver specific molecules or signals into TAMs, reprogramming their function towards an immune-activating state. These molecules can activate the antitumour functions of TAMs, promoting their attack on tumour cells and improving the overall immune response.81 In previous studies, F. Zhang, N.N. Parayath, and others utilized nanocarriers as delivery systems for mRNAs. In ovarian cancer, melanoma, and glioblastoma models, infusion of NPs containing IRF5-encoding mRNA and its activating kinase IKKβ could reverse the immunosuppression and tumour-supportive state of TAMs and reprogramme them into a phenotype that could induce antitumour immunity and promote tumour regression. These genetically programmed macrophages could perform antitumour functions. In addition, this strategy increased the infiltration and activation of immune cells in the TME, improving the overall immunotherapy effect.125 Michelle Seif et al introduced a novel method in which recombinant Saccharomyces cerevisiae is used as a gene delivery vehicle for primary human macrophages. The opsonized yeast was efficiently internalized by M2 macrophages, facilitating effective mRNA delivery and initiating the expression of proinflammatory cytokines. Notably, the expression of the protein encoded by the delivered mRNA was greater in M2 macrophages than in M1 macrophages. Moreover, delivering mRNA encoding the proinflammatory regulators MYD88 and TNF to M2 macrophages led to a sustained increase in proinflammatory and cytotoxic cytokines, indicating successful reprogramming of M2 macrophages towards the antitumour M1 phenotype. This study highlights that yeast-based gene delivery is a promising approach for treating certain conditions, such as cancer, that could benefit from M1-polarized macrophages.126

In the reprogramming of tumor-associated macrophages (TAMs), numerous challenges remain in targeting TAMs in solid tumors. The complex anatomical structure of solid tumors presents a physical barrier due to their dense stroma and chaotic vasculature, impeding the efficient delivery of mRNA-loaded carriers to TAMs. For instance, lipid nanoparticles (LNPs) struggle to penetrate deep into the tumor core.127 The plasticity and heterogeneity of TAMs pose difficulties as well. Different solid tumors can induce diverse TAM phenotypes.128,129 Soluble factors such as interleukin-10 (IL-10) and vascular endothelial growth factor (VEGF) secreted by tumors can inhibit TAM activation and nullify reprogramming signals.130,131 Overcoming these obstacles is crucial for effective mRNA-based cancer treatment.

Regulatory T (Treg) cells play crucial roles in maintaining immune tolerance and preventing autoimmune diseases. However, in the TME, Treg cells typically assist tumours in evading immune surveillance by suppressing effective immune responses.132 Compared with reprogramming TAM strategies, strategies for reprogramming Treg cells into T effector cells via NP drugs have been reported in only a limited number of studies.133 Thus, further research into mRNA delivery therapies that target Treg cells to improve anticancer immune responses is warranted. Key receptors involved in the immunosuppressive functions of Treg cells include cytotoxic T-lymphocyte associated antigen 4 (CTLA-4), a critical inhibitory receptor on Treg cells that suppresses effector T-cell activation by binding to CD80/CD86, thereby competitively inhibiting CD28-mediated activation signals.134 CTLA-4 binding leads to the secretion of suppressive cytokines (eg, IL-10 and TGF-β) and reduces the activation and proliferation of effector T cells. Nrp1, a transmembrane glycoprotein specifically expressed on Treg cells, interacts with vascular endothelial growth factor (VEGF) and neurogenic molecules to influence Treg cell stability and function. Studies have demonstrated that mixed NPs targeting Treg cells (tLyp1-hNPs), decorated with tLyp1 peptide and carrying an anti-CTLA-4 ICI, effectively inhibit Treg suppressive functions and improve CD8+ T-cell antitumour effects.135 IL-2R (particularly its α chain CD25) is highly expressed on Treg cells and is a key marker of their function and stability. IL-2 promotes Treg cell survival and proliferation by binding to IL-2R.136 Additionally, receptors such as glucocorticoid-induced TNFR-related protein (GITR) and lymphocyte activation gene-3 (LAG-3) also contribute to maintaining Treg cell function.137 We propose designing mRNA constructs encoding proteins that target these receptors to potentially alter their protumour effects. Additionally, myeloid-derived suppressor cells (MDSCs) are another type of protumour cell that can serve as a target for cancer therapy.138

mRNA Delivery Combined with ICIs

Immune checkpoints are regulatory pathways in the immune system that are essential for preserving self-tolerance and regulating the length and intensity of physiological immunological responses.139 Tumour cells produce chemicals that stimulate these immunological checkpoints. Upon activation, antigens can no longer be presented to T cells, blocking the antigen presentation process in tumour immunity and thus suppressing the immunological activity of T cells. This enables tumour cells to evade detection and persist.140,141 The most common immune checkpoint molecules include PD-1, CTLA-4, Lymphocyte Activation Gene-3 (LAG3), and T-cell immunoglobulin mucin receptor 3 (TIM3).142144 Currently, ICIs are established as first-line treatment options for a variety of solid tumors, including melanoma,145 non-small-cell lung cancer,146 head and neck squamous cell carcinoma,147 triple-negative breast cancer,148 and advanced gastric cancer.149 Nonetheless, a significant proportion of patients do not experience tumor progression cessation. This is primarily due to chronic inflammation or immunosuppressive signals within the tumor immune microenvironment, which promote tumor immune escape and lead to resistance to ICI therapy.

Perez-Nunez et al reported that an increase in the expression level of a protein called ligand-dependent corepressor (LCOR) can disrupt tumour immune escape. The overexpression of LCOR makes cancer cells “visible” to the immune system, thereby generating an antitumour effect. Consequently, the team utilized extracellular vesicles to encapsulate LCOR mRNA and combined this with anti-PD-L1 treatment. The results showed that the combination of extracellular vesicle) LCOR mRNA therapy and anti-PD-L1 therapy overcame drug resistance and eradicated breast cancer metastasis in preclinical models.150

In mouse models, mutations or deletions of certain tumour suppressor genes, such as phosphatase and tensin homologue (PTEN), can lead to inadequate response or resistance to immune checkpoint blockade (ICB) therapy.151,152 PTEN is a critical tumour suppressor gene with significant roles in tumour suppression and the regulation of immune responses.153 Lin et al developed a new polymeric NP platform comprising mPEG-PLGA and the cationic lipid-like material G0-C14. In vivo experiments demonstrated that PTEN mRNA NPs have the ability to counteract the immunosuppressive TME by facilitating the infiltration of CD8+ T cells into tumour tissues, increasing the production of proinflammatory cytokines (such as IL-12, TNF-α, and IFN-γ), and decreasing the presence of T cells and MDSCs. Compared with treatment with anti-PD-1 alone, combining PTEN mRNA NPs with PD-1 antibodies achieved significant antitumour efficacy in mouse models of PTEN-mutant melanoma and PTEN-deleted prostate cancer.154

In the study by Li et al, the authors investigated the effects of mRNA lipid NP-mediated pyroptosis on “cold” tumours and its combination with ICIs. ‘Cold’ tumours are characterized by a lack of pre-existing immune infiltration, which means there are few immune cells actively engaging with the tumour cells. This paucity of immune cell activity is often due to immunosuppressive factors secreted by the tumour itself or the surrounding microenvironment. These factors can inhibit the recruitment and activation of immune cells, leading to a state where the tumour remains undetected or unchallenged by the immune system. Research has shown that mRNAs delivered by LNPs encode proteins that induce pyroptosis in tumour cells. This form of cell death not only effectively activates tumour-specific immune responses but also promotes immune activation of the TME. This transformation makes originally “cold” tumours, which are less responsive to ICIs, more “hot” and significantly increases their sensitivity to ICIs. The combination of this pyroptosis-inducing strategy with ICIs improves the overall antitumour response, thereby improving therapeutic outcomes. This study provided a novel solution to the common issue of resistance in immunotherapy, and its findings highlighted the significant potential of combining mRNA delivery therapies with ICIs to increase the efficacy of cancer immunotherapy.155

In light of the potential involvement of p53 dysfunction in immunosuppression, scientists have conducted studies to examine the effects of reintroducing p53 expression to the TME and the efficacy of ICB. Researchers have developed and optimized an mRNA NP platform targeting CXCR4 to efficiently stimulate p53 expression in HCC models. In both orthotopic and ectopic p53-null HCC mouse models, the combination of CXCR4-targeted p53 mRNA NPs and anti-PD-1 therapy achieved better antitumour effects than did anti-PD-1 treatment alone. These findings suggest that the use of p53 mRNA nanomedicines in conjunction with ICB can effectively counteract immunosuppression in HCC and facilitate the adoption of this approach in cancer therapy.156 In another study, researchers created NPs to deliver mRNA vaccines encoding the TA MUC1 to DCs in lymph nodes to stimulate and increase the population of T lymphocytes that specifically target tumours. The findings of the study indicate that, compared with the use of monoclonal antibodies alone, the concurrent use of vaccines and anti-CTLA-4 monoclonal antibodies in immunotherapy can greatly augment the immune response against tumours.157

mRNA Delivery in CAR-Based Cancer Immunotherapy

Chimeric Antigen Receptor (CAR) therapy is a personalized treatment approach involving ex vivo genetic modification of autologous immune cells to selectively target cancer cells.158 While CAR-T-cell therapy has demonstrated significant efficacy in haematological malignancies, its clinical application to solid tumours is hindered by challenges such as poor tumour penetration, tumour heterogeneity, and immunosuppressive microenvironments.159,160 mRNA-based CAR therapy offers the advantage of transient CAR expression without genomic integration, which may reduce toxicity and side effects.161 Additionally, to address the issue of tumour heterogeneity, mRNA NP delivery systems can transport mRNAs encoding Tumor-Specific Antigens (TSAs) or TAAs into the bloodstream via intravenous injection. Upon uptake by APCs, these mRNA NPs facilitate the expression of TSAs/TAAs on the APC membrane. The interaction between these APCs and the corresponding CAR-T cells can effectively promote CAR-T-cell proliferation, enabling them to recognize and destroy targeted tumour cells.162,163 Therefore, the application of mRNA delivery in CAR-T-cell therapy holds significant promise for advancing cancer treatment (Figure 4).

Figure 4 mRNA delivery in chimeric antigen receptor (CAR)-based cancer immunotherapy. Created in BioRender. Dai, C. (2025) https://BioRender.com/p45z401.

There have been significant advancements in the application of mRNA delivery in CAR-T-cell therapy. For example, Matthias Stephan et al utilized anti-CD8 antibody-modified NP-like carriers to achieve transient expression of CAR mRNA in circulating T cells in vivo.164 Repeated administration of these NP formulations induced tumour regression. In a separate study, DLin-MC3-DMA LNPs functionalized with anti-CD5 antibodies were developed as a new platform for delivering anti-fibroblast activation protein (FAP) CAR mRNA, aimed at engineering T cells to target activated fibroblasts.31 A single intravenous injection of LNPs containing anti-FAP CAR mRNA was able to transiently generate anti-FAP T cells in vivo, ultimately restoring cardiac function and reducing fibrosis in established hypertensive mouse models of cardiac injury. The findings of these studies highlight the potential of mRNA delivery systems in CAR-T-cell therapy for cancer. To address the issue of off-target toxicity caused by the expression of target antigens in normal tissues during CAR T-cell engineering, researchers have established a clinical platform using in vitro transcribed mRNAs encoding CARs (including CD3-ζ and 4–1BB costimulatory domains) to engineer T cells with transient CAR expression. Case reports from two ongoing trials demonstrate that adoptive transfer of mRNA CAR-T cells targeting mesothelin (CAR-T cells) is feasible and safe.165

Additionally, several mRNA delivery systems encoding TAAs have advanced to clinical research stages. For example, CV9201, an mRNA drug for non-small cell lung cancer (NSCLC) that encodes five TAAs and is delivered via RNActive technology, demonstrated good tolerability and T-cell responses in 63% of patients in Phase I/II trials. Additionally, in a phase Ib trial, combining CV9201 with radiotherapy resulted in 84% of patients showing antigen-specific immune responses.166,167 Similarly, BNT111, designed for melanoma and encoding four TAAs (tyrosinase, MAGE-A3, NYESO-1, and TPTE), also uses the LPX system. In a Phase I trial, 75% of patients developed an immune response to one or more antigens, and combination therapy with anti-PD-1 antibodies resulted in significant clinical responses. Ongoing Phase II trials are evaluating the combined use of BNT111 and cemiplimab, with preliminary results indicating an increased objective response rate in melanoma patients.168 BNT211, an mRNA drug encoding the TAA CLDN6, also utilizes the LPX delivery system. Studies have shown that LPX-loaded CLDN6-mRNA effectively induced CAR-T-cell expansion and significantly suppressed ovarian cancer growth in animal models, highlighting its increased therapeutic efficacy.169 Clinical trials have demonstrated that 57% of patients receiving CLDN6 CAR-T cells achieved partial remission, whereas 14% experienced stable disease, underscoring the potential of this therapy as a treatment option.170

Currently, mRNA-CAR-T therapy has its own current situations and advantages. The non-viral vector technique is getting noticed as it can address stability issues by regulating CAR expression with mRNA. Different non-viral delivery systems have unique features, like the flexibility of electroporation and the customizability and low toxicity of nano-carriers. New treatments encoding CAR-related proteins with mRNA also show promise. However, challenges remain. Electroporation may harm cells, and nano-carriers have stability, storage, and biocompatibility issues. Future development can be driven by integrating nanoparticle delivery with gene editing, identifying tumor targets, optimizing delivery carriers, and enabling in vivo CAR-T cell generation.171

Owing to their significant cytotoxic potential through their involvement in both innate and adaptive immunity, NK cells are considered promising candidates for CAR therapy alongside T cells.172 NK-92 is a homogeneous and stable human cell line commonly used as a model for NK cells.173,174 Given these factors, the NK-92 cell line represents an attractive platform for ready-made CAR therapy, as it also avoids graft-versus-host disease. Indeed, several clinical studies have demonstrated the efficacy of engineered NK-92 cells against various cancers, including haematological malignancies and solid tumours.175,176 In one study, researchers used an innovative proprietary delivery platform to produce protein transduction domain (PTD)-IVT-mRNA for expressing CARs on NK-92 cells. CAR-T1E-engineered NK-92 cells were created by incorporating the T1E single-chain variable fragment (scFv) sequence to recognize the ErbB receptor, with either CD28 or 4–1BB as costimulatory signalling domains, and their efficacy was evaluated in two different ErbB (+) cancer cell lines. The results showed that the PTD-IVT-mRNA for the CAR was safely transduced and expressed in NK-92 cells. CAR-T1E-engineered NK-92 cells, which are used as effector cells in coculture assays against HSC-3 (oral squamous cell carcinoma) and MCF-7 (breast metastatic adenocarcinoma) human cell lines, triggered high levels of cell death (25–33%).177 Studies have shown that the expression of the NKG2D RNA CAR significantly increased the cytotoxic activity of NK cells against various solid tumour cell lines in vitro and provided notable therapeutic benefits for mice with solid tumours. Following the confirmation of the efficacy of NKG2D ligand-targeted mRNA CAR-NK cells in vitro and in animal studies, Lin Xiao et al initiated a pilot clinical trial (NCT03415100), demonstrating that the intraperitoneal infusion and intratumoral injection of CAR-NK cells is a safe and effective local treatment for metastatic colorectal cancer.178

Recently, it has been demonstrated that delivering mRNA encoding an anti-GPC3 CAR and an engineered receptor that competitively binds to the phagocytic signal protein CD24 via PPZ-A10 LNPs to target liver macrophages is an effective strategy for increasing the phagocytic activity of CAR-M cells in vivo. In a mouse model of orthotopic HCC, systemic injection of LNPs containing the dual-mRNA formulation resulted in a reduced tumour burden and increased survival time.179 These promising results underscore the potential of mRNA delivery systems for optimizing CAR-M-cell therapy by enabling precise, adaptable, and safe targeting of TAMs. This approach not only increases the efficacy of CAR-M cells but also represents a significant advancement in the development of personalized cancer treatments, potentially transforming therapeutic strategies for solid tumours and metastatic diseases.

Conclusion and Future Directions

Tumour immunotherapy is a treatment method that attacks tumour cells by activating or regulating the human immune system. The application prospects of mRNA delivery in cancer immunotherapy are extensive. From the initial delivery of naked RNA to the use of liposome carriers, peptide carriers, inorganic NP carriers, and exosomes, mRNA delivery systems have become more stable, more targeted, and exhibit improved release effects. mRNAs have the ability to encode TAAs, immunostimulatory molecules (such as IL-2, IL-12, IL-27, IL-15, IL-23, GM-CSF, IFNγ, etc)., molecules that facilitate the transformation of immune cells with tumor-promoting effects, and molecules that enhance the efficacy of ICIs, as well as those used in CAR-based cell therapy to enhance antitumor immune effects. The combination of mRNA delivery with ICIs can effectively enhance the immune response, thereby overcoming the limitations of single-agent therapies and improving overall efficacy (Figure 5). In CAR-based cell therapy, mRNA delivery technology allows for transient CAR expression, effectively addressing the issue of long-term off-target toxicity associated with traditional CAR-based cell therapies. This strategy not only enhances the targeting ability and efficacy of CAR-T, CAR-NK, and CAR-M cells, but also provides safer and more adaptable treatment options.

Figure 5 Mechanism of mRNA delivery in tumour immunotherapy. Taking lipid nanoparticle (LNP)-encapsulated mRNAs as an example, these encapsulated mRNAs are introduced into the human body through intravenous injection or other administration methods. They travel via the bloodstream to reach target cells, such as the antigen-presenting cells illustrated in the diagram. Once inside these cells, the LNPs release their mRNA cargo, which then encodes corresponding proteins. These proteins include tumour-associated antigens that activate T cells by binding to signal molecules between antigen-presenting cells and T cells, triggering subsequent antitumour responses. mRNA can also encode immune stimulatory molecules such as interleukins and granulocyte-macrophage colony stimulating factor (GM-CSF), promoting the activation and aggregation of immune cells. Additionally, mRNA can encode signalling molecules that promote the transformation of M2 macrophages into M1 macrophages, thereby improving the anticancer effect. mRNAs that target tumour suppressor genes such as P53 can activate their expression. Furthermore, mRNA delivery therapy can be combined with immune checkpoint blockade therapies or CAR-based cell therapies for cancer treatment, increasing therapeutic efficacy. Created in BioRender. Dai, C. (2025) https://BioRender.com/v27w914.

However, there are still several issues and challenges associated with tumor immunotherapy based on mRNA delivery. Firstly, the design and functionality of mRNA delivery systems depend on various factors, such as the size, shape, charge, and surface modifications of the carriers. If these parameters cannot be precisely controlled, the carriers may enter off-target cells.180 To mitigate this risk, researchers are continuously optimizing the design and functionality of delivery systems to enhance their safety and efficacy in vivo. Secondly, there are currently no comprehensive regulations for assessing the safety of mRNA delivery therapeutics. A standardized system for evaluating the efficacy and safety of these therapeutics needs to be established to ensure comparability among different research findings. Thirdly, the stringent storage requirements for mRNA delivery therapeutics and the current inability to achieve large-scale production contribute to high costs for patients.181 Improving production processes and reducing costs are necessary to make mRNA delivery therapeutics more accessible for widespread use.

In the future, several promising research directions and suggestions can be considered.182 Firstly, developing novel vectors is of paramount importance. For example, exploring biomimetic vectors that mimic the natural cell communication mechanisms could potentially enhance the delivery efficiency and biocompatibility. Synthetic polymers with unique nanostructures might also be designed to precisely control the release of mRNA and target specific cell types.183 Secondly, optimizing mRNA sequences is another crucial aspect. By employing advanced bioinformatics tools, researchers can predict and modify mRNA sequences to improve their stability, translation efficiency, and immunogenicity.184 This could involve minimizing secondary structure formation to facilitate ribosome binding and translation initiation. Furthermore, efforts should be directed towards engineering self-amplifying mRNAs that can produce multiple copies of the encoded protein, thereby amplifying the therapeutic effect. Finally, interdisciplinary collaborations between materials science, bioengineering, and immunology will be essential to address the complex challenges in mRNA-based tumour immunotherapy and drive innovation in this field.185 It will be necessary to focus on and address these issues to achieve wider application and better development of mRNA-based tumour immunotherapy.

Acknowledgments

The Fundamental Research Funds for the Central Universities (2042024YXB009 to X.C.), the Special Foundation for Knowledge Innovation of Wuhan Science and Technology Innovation Bureau (2023020201020510 to X.C.), and the Fundamental Research Funds for the Central Universities (2042024YXB009 to X.C.).

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Disclosure

The authors declare that they have no competing interests in this work.

References

1. Wang M, Herbst RS, Boshoff C. Toward personalized treatment approaches for non-small-cell lung cancer. Nat Med. 2021;27(8):1345–1356. doi:10.1038/s41591-021-01450-2

2. Morrison AH, Byrne KT, Vonderheide RH. Immunotherapy and Prevention of Pancreatic Cancer. Trends Cancer. 2018;4(6):418–428. doi:10.1016/j.trecan.2018.04.001

3. Baxevanis CN, Perez SA, Papamichail M. Critical Reviews in Clinical Laboratory Sciences. Cancer Immunotherap. 2009;46(4):167–189.

4. Zhu Y, Zhu X, Tang C, et al. Progress and challenges of immunotherapy in triple-negative breast cancer. Biochim Biophys Acta. 2021;1876(2):188593. doi:10.1016/j.bbcan.2021.188593

5. Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 2021;11(4). doi:10.1038/s41408-021-00459-7

6. Cha J-H, Chan L-C, Li C-W, et al. Mechanisms Controlling PD-L1 Expression in Cancer. Molecular Cell. 2019;76(3):359–370. doi:10.1016/j.molcel.2019.09.030

7. Kim MJ, Kim K, Park HJ, et al. Deletion of PD-1 destabilizes the lineage identity and metabolic fitness of tumor-infiltrating regulatory T cells. Nat Immunol. 2022;24(1):148–161. doi:10.1038/s41590-022-01373-1

8. Mantovani A, Sozzani S, Locati M, et al. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002;23(11):549–555. doi:10.1016/S1471-4906(02)02302-5

9. Giese MA, Hind LE, Huttenlocher A. Neutrophil plasticity in the tumor microenvironment. Blood. 2019;133(20):2159–2167. doi:10.1182/blood-2018-11-844548

10. Martins F, Sofiya L, Sykiotis GP, et al. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat Rev Clin Oncol. 2019;16(9):563–580. doi:10.1038/s41571-019-0218-0

11. Lorentzen CL, Haanen JB, Met Ö, et al. Clinical advances and ongoing trials of mRNA vaccines for cancer treatment. Lancet Oncol. 2022;23(10):e450–e458. doi:10.1016/S1470-2045(22)00372-2

12. Qin S, Tang X, Chen Y, et al. mRNA-based therapeutics: powerful and versatile tools to combat diseases. Signal Transduct Target Ther. 2022;7(1):166. doi:10.1038/s41392-022-01007-w

13. Self WH, Tenforde MW, Rhoads JP, et al. Comparative Effectiveness of Moderna, Pfizer-BioNTech, and Janssen (Johnson & Johnson) Vaccines in Preventing COVID-19 hospitalizations Among Adults Without Immunocompromising Conditions — United States, March–August 2021. MMWR. 2021;70(38):1337–1343. doi:10.15585/mmwr.mm7038e1

14. Munro APS, Feng S, Janani L, et al. Safety, immunogenicity, and reactogenicity of BNT162b2 and mRNA-1273 COVID-19 vaccines given as fourth-dose boosters following two doses of ChAdOx1 nCoV-19 or BNT162b2 and a third dose of BNT162b2 (COV-BOOST): a multicentre, blinded, Phase 2, randomised trial. The Lancet. Infectious Diseases. 2022;22(8):1131–1141. doi:10.1016/S1473-3099(22)00271-7

15. Li X, Qi J, Wang J, et al. Nanoparticle technology for mRNA: delivery strategy, clinical application and developmental landscape. Theranostics. 2024;14(2):738–760. doi:10.7150/thno.84291

16. Billingsley MM, Singh N, Ravikumar P, et al. Ionizable Lipid Nanoparticle-Mediated mRNA Delivery for Human CAR T Cell Engineering. Nano Lett. 2020;20(3):1578–1589. doi:10.1021/acs.nanolett.9b04246

17. Dilliard SA, Cheng Q, Siegwart DJ. On the mechanism of tissue-specific mRNA delivery by selective organ targeting nanoparticles. Proc Natl Acad Sci U S A. 2021;118(52). doi:10.1073/pnas.2109256118

18. Wang X, Liu S, Sun Y, et al. Preparation of selective organ-targeting (SORT) lipid nanoparticles (LNPs) using multiple technical methods for tissue-specific mRNA delivery. Nat Protoc. 2023;18(1):265–291. doi:10.1038/s41596-022-00755-x

19. Billingsley MM, Hamilton AG, Mai D, et al. Orthogonal Design of Experiments for Optimization of Lipid Nanoparticles for mRNA Engineering of CAR T Cells. Nano Lett. 2022;22(1):533–542. doi:10.1021/acs.nanolett.1c02503

20. Ho W, Gao M, Li F, et al. Next‐Generation Vaccines: nanoparticle‐Mediated DNA and mRNA Delivery. Adv Healthcare Mater. 2021;10(8). doi:10.1002/adhm.202001812.

21. Friedrich M, Aigner A. Therapeutic siRNA: state-of-the-Art and Future Perspectives. BioDrugs. 2022;36(5):549–571. doi:10.1007/s40259-022-00549-3

22. Zong Y, Lin Y, Wei T, et al. Lipid Nanoparticle (LNP) Enables mRNA Delivery for Cancer Therapy. Adv Mater. 2023;35(51):e2303261. doi:10.1002/adma.202303261

23. Wang C, Zhao C, Wang W, et al. Biomimetic noncationic lipid nanoparticles for mRNA delivery. Proc Natl Acad Sci U S A. 2023;120(51):e2311276120. doi:10.1073/pnas.2311276120

24. Doll TAPF, Dey R, Burkhard P. Design and optimization of peptide nanoparticles. J Nanobiotechnol. 2015;13(1). doi:10.1186/s12951-015-0119-z

25. Huang-Chiao H, Barua S, Sharma G, Dey SK, Rege K. Inorganic nanoparticles for cancer imaging and therapy. J Control Rel. 2011;155(3):344–357. doi:10.1016/j.jconrel.2011.06.004

26. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):640. doi:10.1126/science.aau6977

27. Xu Q, Wang Y, Zheng Y, et al. Polymersomes in Drug Delivery horizontal line From Experiment to Computational Modeling. Biomacromolecules. 2024;25(4):2114–2135. doi:10.1021/acs.biomac.3c00903

28. Lin M, Dai Y, Xia F, et al. Advances in non-covalent crosslinked polymer micelles for biomedical applications. Mater Sci Eng C Mater Biol Appl. 2021;119:111626. doi:10.1016/j.msec.2020.111626

29. Tenchov R, Bird R, Curtze AE, et al. Lipid Nanoparticles horizontal line From Liposomes to mRNA Vaccine Delivery, a Landscape of Research Diversity and Advancement. ACS Nano. 2021;15(11):16982–17015. doi:10.1021/acsnano.1c04996

30. Kon E, Ad-El N, Hazan-Halevy I, et al. Targeting cancer with mRNA-lipid nanoparticles: key considerations and future prospects. Nat Rev Clin Oncol. 2023;20(11):739–754. doi:10.1038/s41571-023-00811-9

31. Rurik JG, Tombácz I, Yadegari A, et al. CAR T cells produced in vivo to treat cardiac injury. Science. 2022;375(6576):91–96. doi:10.1126/science.abm0594

32. Alireza F, Rahbarizadeh F, Ahmadvand D, Ranjbar S, Khoshtinat Nikkhoi S. Effective suppression of tumour cells by oligoclonal HER2-targeted delivery of liposomal doxorubicin. J Liposome Res. 2018;29(1):53–65. doi:10.1080/08982104.2018.1430829

33. Goswami R, O’Hagan DT, Adamo R, et al. Conjugation of Mannans to Enhance the Potency of Liposome Nanoparticles for the Delivery of RNA Vaccines. Pharmaceutics. 2021;13(2):240. doi:10.3390/pharmaceutics13020240

34. Yokoo H, Oba M, Uchida S. Cell-Penetrating Peptides: emerging Tools for mRNA Delivery. Pharmaceutics. 2022;14(1):78.

35. D’haese S, Laeremans T, Roover S, et al. Efficient Induction of Antigen-Specific CD8+ T-Cell Responses by Cationic Peptide-Based mRNA Nanoparticles. Pharmaceutics. 2022;14(7):1387. doi:10.3390/pharmaceutics14071387

36. Martin ME, Rice KG. Peptide-guided gene delivery. AAPS J. 2007;9(1):E18–29. doi:10.1208/aapsj0901003

37. Lehto T, Ezzat K, Wood MJA, et al. Peptides for nucleic acid delivery. Adv Drug Deliv Rev. 2016;106(Pt A):172–182. doi:10.1016/j.addr.2016.06.008

38. Thapa RK, Sullivan MO. Gene delivery by peptide-assisted transport. Curr Opin Biomed Eng. 2018;7:71–82. doi:10.1016/j.cobme.2018.10.002

39. Ni R, Feng R, Chau Y. Synthetic approaches for nucleic acid delivery: choosing the right carriers. Life. 2019;9(3):59. doi:10.3390/life9030059

40. Margus H, Arukuusk P, Langel Ü, et al. Characteristics of cell-penetrating peptide/nucleic acid nanoparticles. Mol Pharm. 2016;13(1):172–179. doi:10.1021/acs.molpharmaceut.5b00598

41. Konate K, Lindberg MF, Vaissiere A, et al. Optimisation of vectorisation property: a comparative study for a secondary amphipathic peptide. Int J Pharm. 2016;509(1–2):71–84. doi:10.1016/j.ijpharm.2016.05.030

42. Deshayes S, Morris MC, Divita G, et al. Cell-penetrating peptides: tools for intracellular delivery of therapeutics. 2005;62(16):1839–1849. doi:10.1007/s00018-005-5109-0

43. Lamarre B, Ravi J, Ryadnov MG. GeT peptides: a single domain approach to gene delivery. Chem Commun. 2011;47(32):9045–9047. doi:10.1039/c1cc13043a

44. Carnal F, Clavier A, Stoll S. Polypeptide-Nanoparticle Interactions and Corona Formation Investigated by Monte Carlo Simulations. Polymers. 2016;8(6):203.

45. Cox A, Lim SA, Chung EJ. Strategies to deliver RNA by nanoparticles for therapeutic potential. mol Aspect Med. 2022;83:100991. doi:10.1016/j.mam.2021.100991

46. Zhou Y, Li Q, Wu Y, et al. Molecularly Stimuli-Responsive Self-Assembled Peptide Nanoparticles for Targeted Imaging and Therapy. ACS Nano. 2023;17(9):8004–8025. doi:10.1021/acsnano.3c01452

47. Wickline SA, Hou KK, Pan H. Peptide-Based Nanoparticles for Systemic Extrahepatic Delivery of Therapeutic Nucleotides. Int J mol Sci. 2023;24(11):9455. doi:10.3390/ijms24119455

48. Shin H, Kang S, Won C, et al. Enhanced Local Delivery of Engineered IL-2 mRNA by Porous Silica Nanoparticles to Promote Effective Antitumor Immunity. ACS Nano. 2023;17(17):17554–17567. doi:10.1021/acsnano.3c06733

49. Janjua TI, Cao Y, Kleitz F, et al. Silica nanoparticles: a review of their safety and current strategies to overcome biological barriers. Adv Drug Deliv Rev. 2023;203:115115. doi:10.1016/j.addr.2023.115115

50. Noureddine A, Maestas-Olguin A, Tang L, et al. Future of Mesoporous Silica Nanoparticles in Nanomedicine: protocol for Reproducible Synthesis, Characterization, Lipid Coating, and Loading of Therapeutics (Chemotherapeutic, Proteins, siRNA and mRNA). ACS Nano. 2023;17(17):16308–16325. doi:10.1021/acsnano.3c07621

51. Yang Y, Zhang M, Song H, et al. Silica-Based Nanoparticles for Biomedical Applications: from Nanocarriers to Biomodulators. Acc Chem Res. 2020;53(8):1545–1556. doi:10.1021/acs.accounts.0c00280

52. Sun B, Wu W, Narasipura EA, et al. Engineering nanoparticle toolkits for mRNA delivery. Adv Drug Deliv Rev. 2023;200:115042. doi:10.1016/j.addr.2023.115042

53. Narayan R, Nayak UY, Raichur AM, et al. Mesoporous Silica Nanoparticles: a Comprehensive Review on Synthesis and Recent Advances. Pharmaceutics. 2018;10(3):118. doi:10.3390/pharmaceutics10030118

54. Gusta MF, Edel MJ, Salazar VA, et al. Exploiting endocytosis for transfection of mRNA for cytoplasmatic delivery using cationic gold nanoparticles. Front Immunol. 2023;14:1128582. doi:10.3389/fimmu.2023.1128582

55. Huang W, Liu Z, Zhou G, et al. Magnetic gold nanoparticle-mediated small interference RNA silencing Bag-1 gene for colon cancer therapy. Oncol Rep. 2016;35(2):978–984. doi:10.3892/or.2015.4453

56. Stephen ZR, Kievit FM, Veiseh O, et al. Redox-Responsive Magnetic Nanoparticle for Targeted Convection-Enhanced Delivery of O 6 -Benzylguanine to Brain Tumors. ACS Nano. 2014;8(10):10383–10395. doi:10.1021/nn503735w

57. Kong L, Qiu J, Sun W, et al. Multifunctional PEI-entrapped gold nanoparticles enable efficient delivery of therapeutic siRNA into glioblastoma cells. Biomater Sci. 2017;5(2):258–266. doi:10.1039/C6BM00708B

58. Revia RA, Stephen ZR, Zhang M. Theranostic Nanoparticles for RNA-Based Cancer Treatment. Acc Chem Res. 2019;52(6):1496–1506. doi:10.1021/acs.accounts.9b00101

59. Tsai SJ, Atai NA, Cacciottolo M, et al. Exosome-mediated mRNA delivery in vivo is safe and can be used to induce SARS-CoV-2 immunity. J Biol Chem. 2021;297(5):101266. doi:10.1016/j.jbc.2021.101266

60. Morishita M, Takahashi Y, Matsumoto A, et al. Exosome-based tumor antigens–adjuvant co-delivery utilizing genetically engineered tumor cell-derived exosomes with immunostimulatory CpG DNA. Biomaterials. 2016;111:55–65. doi:10.1016/j.biomaterials.2016.09.031

61. Patil SM, Sawant SS, Kunda NK. Exosomes as drug delivery systems: a brief overview and progress update. Eur J Pharm Biopharm. 2020;154:259–269. doi:10.1016/j.ejpb.2020.07.026

62. Jiaci C, Li P, Zhang T. Review on Strategies and Technologies for Exosome Isolation and Purification. Front Bioeng Biotechnol. 2022;9. doi:10.3389/fbioe.2021.811971

63. Sidhom K, Obi PO, Saleem A. A Review of Exosomal Isolation Methods: is Size Exclusion Chromatography the Best Option? Int J mol Sci. 2020;21(18):6466. doi:10.3390/ijms21186466

64. Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30(1):255–289. doi:10.1146/annurev-cellbio-101512-122326

65. Zhang Y, Bi J, Huang J, et al. Exosome: a Review of Its Classification, Isolation Techniques, Storage, Diagnostic and Targeted Therapy Applications. Int j Nanomed. 2020;15:6917–6934. doi:10.2147/IJN.S264498

66. Wang JH, Forterre AV, Zhao J, et al. Anti-HER2 scFv-directed extracellular vesicle-mediated mRNA-based gene delivery inhibits growth of HER2-positive human breast tumor xenografts by prodrug activation. mol Cancer Ther. 2018;17(5):1133–1142. doi:10.1158/1535-7163.MCT-17-0827

67. Forterre AV, Wang J-H, Delcayre A, et al. Extracellular Vesicle–Mediated In Vitro Transcribed mRNA Delivery for Treatment of HER2+ Breast Cancer Xenografts in Mice by Prodrug CB1954 without General Toxicity. mol Cancer Ther. 2020;19(3):858–867. doi:10.1158/1535-7163.MCT-19-0928

68. Miller AJ, Pearce AK, Foster JC, et al. Probing and Tuning the Permeability of Polymersomes. ACS Cent Sci. 2021;7(1):30–38. doi:10.1021/acscentsci.0c01196

69. Negut I, Bita B. Polymersomes as Innovative, Stimuli-Responsive Platforms for Cancer Therapy. Pharmaceutics. 2024;16(4):463. doi:10.3390/pharmaceutics16040463

70. Sherje AP, Jadhav M, Dravyakar BR, et al. Dendrimers: a versatile nanocarrier for drug delivery and targeting. Int J Pharm. 2018;548(1):707–720. doi:10.1016/j.ijpharm.2018.07.030

71. Palmerston Mendes L, Pan J, Torchilin VP. Dendrimers as Nanocarriers for Nucleic Acid and Drug Delivery in Cancer Therapy. Molecules. 2017;22(9):1401. doi:10.3390/molecules22091401

72. Chauhan AS. Dendrimers for Drug Delivery. Molecules. 2018;23(4):938. doi:10.3390/molecules23040938

73. Wang J, Li B, Qiu L, et al. Dendrimer-based drug delivery systems: history, challenges, and latest developments. J Biol Eng. 2022;16(1). doi:10.1186/s13036-022-00298-5.

74. Shukla R, Thomas TP, Desai AM, et al. HER2 specific delivery of methotrexate by dendrimer conjugated anti-HER2 mAb. Nanotechnology. 2008;19(29):295102. doi:10.1088/0957-4484/19/29/295102

75. Cheng YY, Xu Z, Ma M, et al. Dendrimers as drug carriers: applications in different routes of drug administration. J Pharma Sci. 2008;97(1):123–143. doi:10.1002/jps.21079

76. Longmire M, Choyke P, Kobayashi H. Dendrimer-based contrast agents for molecular imaging. Curr Top Med Chem. 2008;8(14):1180–1186. doi:10.2174/156802608785849021

77. Movassaghian S, Merkel OM, Torchilin VP. Applications of polymer micelles for imaging and drug delivery. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2015;7(5):691–707. doi:10.1002/wnan.1332

78. Gothwal A, Khan I, Gupta U. Polymeric Micelles: recent Advancements in the Delivery of Anticancer Drugs. Pharm Res. 2016;33(1):18–39. doi:10.1007/s11095-015-1784-1

79. Wang QT, Nie Y, Sun S-N, et al. Tumor-associated antigen-based personalized dendritic cell vaccine in solid tumor patients. Cancer Immunol Immunother. 2020;69(7):1375–1387. doi:10.1007/s00262-020-02496-w

80. Hess PR, Boczkowski D, Nair SK, et al. Vaccination with mRNAs encoding tumor-associated antigens and granulocyte-macrophage colony-stimulating factor efficiently primes CTL responses, but is insufficient to overcome tolerance to a model tumor/self antigen. Cancer Immunol Immunother. 2006;55(6):672–683. doi:10.1007/s00262-005-0064-z

81. Li X, Guo X, Ling J, et al. Nanomedicine-based cancer immunotherapies developed by reprogramming tumor-associated macrophages. Nanoscale. 2021;13(9):4705–4727. doi:10.1039/D0NR08050K

82. Andersen MH. Tumor microenvironment antigens. Semin Immunopathol. 2022;45(2):253–264. doi:10.1007/s00281-022-00966-0

83. Rittig SM, Haentschel M, Weimer KJ, et al. Long-term survival correlates with immunological responses in renal cell carcinoma patients treated with mRNA-based immunotherapy. Oncoimmunology. 2016;5(5):e1108511. doi:10.1080/2162402X.2015.1108511

84. Vishweshwaraiah YL, Dokholyan NV. mRNA vaccines for cancer immunotherapy. Front Immunol. 2022;13. doi:10.3389/fimmu.2022.1029069

85. Zimmermannova O, Ferreira AG, Ascic E, et al. Restoring tumor immunogenicity with dendritic cell reprogramming. Sci Immunol. 2023;8(85). doi:10.1126/sciimmunol.add4817.

86. Wang TT, Han M, Han Y, et al. Antigen Self-Presented Personalized Nanovaccines Boost the Immunotherapy of Highly Invasive and Metastatic Tumors. Acs Nano. 2024;18(8):6333–6347. doi:10.1021/acsnano.3c11189

87. Miao L, Zhang Y, Huang L. mRNA vaccine for cancer immunotherapy. mol Cancer. 2021;20(1):41. doi:10.1186/s12943-021-01335-5

88. Sun QF, Zhao XN, Peng CL, et al. Immunotherapy for Lewis lung carcinoma utilizing dendritic cells infected with CK19 gene recombinant adenoviral vectors. Oncol Rep. 2015;34(5):2289–2295. doi:10.3892/or.2015.4231

89. Mehrpouya M, Pourhashem Z, Yardehnavi N, et al. Evaluation of cytokeratin 19 as a prognostic tumoral and metastatic marker with focus on improved detection methods. J Cell Physiol. 2019;234(12):21425–21435. doi:10.1002/jcp.28768

90. Mai Y, Guo J, Zhao Y, et al. Intranasal delivery of cationic liposome-protamine complex mRNA vaccine elicits effective anti-tumor immunity. Cell Immunol. 2020;354:104143. doi:10.1016/j.cellimm.2020.104143

91. Van Lint S, Goyvaerts C, Maenhout S, et al. Preclinical evaluation of TriMix and antigen mRNA-based antitumor therapy. Cancer Res. 2012;72(7):1661–1671. doi:10.1158/0008-5472.CAN-11-2957

92. Chen J, Ye Z, Huang C, et al. Lipid nanoparticle-mediated lymph node–targeting delivery of mRNA cancer vaccine elicits robust CD8 + T cell response. Proc Natl Acad Sci U S A. 2022;119(34):e2207841119. doi:10.1073/pnas.2207841119

93. Sun WQ, Ji P, Zhou T, et al. Ultrasound Responsive Nanovaccine Armed with Engineered Cancer Cell Membrane and RNA to Prevent Foreseeable Metastasis. Adv Sci. 2023;10(19). doi:10.1002/advs.202301107.

94. Kranz LM, Diken M, Haas H, et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature. 2016;534(7607):396. doi:10.1038/nature18300

95. Lei JQ, et al. Development of Mannosylated Lipid Nanoparticles for mRNA Cancer Vaccine with High Antigen Presentation Efficiency and Immunomodulatory Capability. Angew Chem Int Ed. 2024;136(13):e202318515.

96. Mu LJ, Kyte JA, Kvalheim G, et al. Immunotherapy with allotumour mRNA-transfected dendritic cells in androgen-resistant prostate cancer patients. Br J Cancer. 2005;93(7):749–756. doi:10.1038/sj.bjc.6602761

97. Phua KK, Staats HF, Leong KW, et al. Intranasal mRNA nanoparticle vaccination induces prophylactic and therapeutic anti-tumor immunity. Sci Rep. 2014;4(1):5128. doi:10.1038/srep05128

98. Garland KM, Sheehy TL, Wilson JT. Chemical and Biomolecular Strategies for STING Pathway Activation in Cancer Immunotherapy. Chem Rev. 2022;122(6):5977–6039. doi:10.1021/acs.chemrev.1c00750

99. Arenas-Ramirez N, Woytschak J, Boyman O. Interleukin-2: biology, Design and Application. Trends Immunol. 2015;36(12):763–777. doi:10.1016/j.it.2015.10.003

100. Jiang Y, Zhang Y, Liu C, et al. Tumor-activated IL-2 mRNA delivered by lipid nanoparticles for cancer immunotherapy. J Control Rel. 2024;368:663–675. doi:10.1016/j.jconrel.2024.03.016

101. Berraondo P, Etxeberria I, Ponz-Sarvise M, et al. Revisiting Interleukin-12 as a Cancer Immunotherapy Agent. Clin Cancer Res. 2018;24(12):2716–2718. doi:10.1158/1078-0432.CCR-18-0381

102. Lai I, Swaminathan S, Baylot V, et al. Lipid nanoparticles that deliver IL-12 messenger RNA suppress tumorigenesis in MYC oncogene-driven hepatocellular carcinoma. J Immunother Cancer. 2018;6(1):125. doi:10.1186/s40425-018-0431-x

103. Dong CY, Dang D, Zhao X, et al. Integrative Characterization of the Role of IL27 In Melanoma Using Bioinformatics Analysis. Front Immunol. 2021;12. doi:10.3389/fimmu.2021.713001

104. Liu J-Q, Zhang C, Zhang X, et al. Intratumoral delivery of IL-12 and IL-27 mRNA using lipid nanoparticles for cancer immunotherapy. J Control Release. 2022;345:306–313. doi:10.1016/j.jconrel.2022.03.021

105. Di Trani CA, Cirella A, Arrizabalaga L, et al. Intratumoral injection of IL-12-encoding mRNA targeted to CSF1R and PD-L1 exerts potent anti-tumor effects without substantial systemic exposure. mol Ther Nucleic Acids. 2023;33:599–616. doi:10.1016/j.omtn.2023.07.020

106. Patidar M, Yadav N, Dalai SK. Interleukin 15: a key cytokine for immunotherapy. Cytokine Growth Factor Rev. 2016;31:49–59. doi:10.1016/j.cytogfr.2016.06.001

107. Lei SB, Zhang X, Men K, et al. Efficient Colorectal Cancer Gene Therapy with IL-15 mRNA Nanoformulation. Mol Pharmaceut. 2020;17(9):3378–3391. doi:10.1021/acs.molpharmaceut.0c00451

108. Wang XX, Li F, Zhang J, et al. A combination of PD-L1-targeted IL-15 mRNA nanotherapy and ultrasound-targeted microbubble destruction for tumor immunotherapy. J Control Rel. 2024;367:45–60. doi:10.1016/j.jconrel.2024.01.039

109. Hotz C, Wagenaar TR, Gieseke F, et al. Local delivery of mRNA-encoded cytokines promotes antitumor immunity and tumor eradication across multiple preclinical tumor models. Sci Transl Med. 2021;13(610):eabc7804. doi:10.1126/scitranslmed.abc7804

110. Hewitt SL, Bai A, Bailey D, et al. Durable anticancer immunity from intratumoral administration of IL-23, IL-36γ, and OX40L mRNAs. Sci Transl Med. 2019;11(477). doi:10.1126/scitranslmed.aat9143.

111. Panelli MC, White R, Foster M, et al. Forecasting the cytokine storm following systemic interleukin (IL)-2 administration. J Transl Med. 2004;2(1). doi:10.1186/1479-5876-2-17.

112. Shimabukuro-Vornhagen A, Gödel P, Subklewe M, et al. Cytokine release syndrome. J Immuno Therapy Cancer. 2018;6(1). doi:10.1186/s40425-018-0343-9.

113. Gaestel M, Nebreda AR, Yaffe MB. Cytokine Storm. N Engl J Med. 2021;384(16):1.

114. Chen Y, Li F, Hua M, et al. Role of GM-CSF in lung balance and disease. Front Immunol. 2023;14:1158859.

115. Kamiya M, Matsumoto M, Yamashita K, et al. Stability Study of mRNA-Lipid Nanoparticles Exposed to Various Conditions Based on the Evaluation between Physicochemical Properties and Their Relation with Protein Expression Ability. Pharmaceutics. 2022;14(11):2357. doi:10.3390/pharmaceutics14112357

116. Jorgovanovic D, Song M, Wang L, et al. Roles of IFN-γ in tumor progression and regression: a review. Biomark Res. 2020;8(1):49. doi:10.1186/s40364-020-00228-x

117. Shao Z, Chen L, Zhang Z, et al. KERS-Inspired Nanostructured Mineral Coatings Boost IFN-γ mRNA Therapeutic Index for Antitumor Immunotherapy. Adv Mater. 2023;35(51):e2304296. doi:10.1002/adma.202304296

118. Lim RJ, Salehi-Rad R, Tran LM, et al. CXCL9/10-engineered dendritic cells promote T cell activation and enhance immune checkpoint blockade for lung cancer. Cell Rep Med. 2024;5(4):101479. doi:10.1016/j.xcrm.2024.101479

119. Aggarwal BB, Gupta SC, Kim JH. Historical perspectives on tumor necrosis factor and its superfamily: 25 years later, a golden journey. Blood. 2012;119(3):651–665. doi:10.1182/blood-2011-04-325225

120. Parkin J, Cohen B. An overview of the immune system. Lancet. 2001;357(9270):1777–1789. doi:10.1016/S0140-6736(00)04904-7

121. Pathria P, Louis TL, Varner JA. Targeting Tumor-Associated Macrophages in Cancer. Trends Immunol. 2019;40(4):310–327. doi:10.1016/j.it.2019.02.003

122. Pan Y, Yu Y, Wang X, et al. Tumor-Associated Macrophages in Tumor Immunity. Front Immunol. 2020;11:583084. doi:10.3389/fimmu.2020.583084

123. Li D, Ji H, Niu X, et al. Tumor-associated macrophages secrete CC-chemokine ligand 2 and induce tamoxifen resistance by activating PI3K/Akt/mTOR in breast cancer. Cancer Sci. 2020;111(1):47–58. doi:10.1111/cas.14230

124. Li M, He L, Zhu J, et al. Targeting tumor-associated macrophages for cancer treatment. Cell Biosci. 2022;12(1):85. doi:10.1186/s13578-022-00823-5

125. Zhang F, Parayath NN, Ene CI, et al. Genetic programming of macrophages to perform anti-tumor functions using targeted mRNA nanocarriers. Nat Commun. 2019;10(1):3974. doi:10.1038/s41467-019-11911-5

126. Seif M, Hoppstädter J, Breinig F, et al. Yeast-mediated mRNA delivery polarizes immuno-suppressive macrophages towards an immuno-stimulatory phenotype. Eur J Pharm Biopharm. 2017;117:1–13. doi:10.1016/j.ejpb.2017.03.008

127. Chenab KK, Malektaj H, Nadinlooie AAR, Mohammadi S, Zamani-Meymian MR. Intertumoral and intratumoral barriers as approaches for drug delivery and theranostics to solid tumors using stimuli-responsive materials. Mikrochim Acta. 2024;191(9):541.

128. Dai XM, Lu L, Deng S, et al. USP7 targeting modulates anti-tumor immune response by reprogramming Tumor-associated Macrophages in Lung Cancer. Theranostics. 2020;10(20):9332–9347. doi:10.7150/thno.47137

129. Wang H, Tian T, Zhang JH. Tumor-Associated Macrophages (TAMs) in Colorectal Cancer (CRC): from Mechanism to Therapy and Prognosis. Int J mol Sci. 2021;22(16):8470.

130. Wang L-X, Talebian F, Liu J-Q, et al. IL-10 contributes to the suppressive function of tumour-associated myeloid cells and enhances myeloid cell accumulation in tumours. Scandinavian J Immunol. 2012;75(3):273–281. doi:10.1111/j.1365-3083.2011.02662.x

131. Okikawa S, Morine Y, Saito Y, et al. Inhibition of the VEGF signaling pathway attenuates tumor‑associated macrophage activity in liver cancer. Oncol Rep. 2022;47(4). doi:10.3892/or.2022.8282.

132. Qin D, Zhang Y, Shu P, et al. Targeting tumor-infiltrating tregs for improved antitumor responses. Front Immunol. 2024;15:1325946. doi:10.3389/fimmu.2024.1325946

133. Haist M, Mailander V, Bros M. Nanodrugs Targeting T Cells in Tumor Therapy. Front Immunol. 2022;13:912594. doi:10.3389/fimmu.2022.912594

134. Hosseini A, Gharibi T, Marofi F, et al. CTLA-4: from mechanism to autoimmune therapy. Int Immunopharmacol. 2020;80:106221. doi:10.1016/j.intimp.2020.106221

135. Ou W, Thapa RK, Jiang L, et al. Regulatory T cell-targeted hybrid nanoparticles combined with immuno-checkpoint blockage for cancer immunotherapy. J Control Release. 2018;281:84–96. doi:10.1016/j.jconrel.2018.05.018

136. Peng Y, Tao Y, Zhang Y, et al. CD25: a potential tumor therapeutic target. Int J Cancer. 2023;152(7):1290–1303. doi:10.1002/ijc.34281

137. Park HJ, Kusnadi A, Lee E-J, et al. Tumor-infiltrating regulatory T cells delineated by upregulation of PD-1 and inhibitory receptors. Cell Immunol. 2012;278(1–2):76–83. doi:10.1016/j.cellimm.2012.07.001

138. Law AMK, Valdes-Mora F, Gallego-Ortega D. Myeloid-Derived Suppressor Cells as a Therapeutic Target for Cancer. Cells. 2020;9(3):561. doi:10.3390/cells9030561

139. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–264. doi:10.1038/nrc3239

140. Pathania AS, Prathipati P, Murakonda SP, et al. Immune checkpoint molecules in neuroblastoma: a clinical perspective. Semin Cancer Biol. 2022;86(Pt 2):247–258. doi:10.1016/j.semcancer.2022.06.013

141. Mei Y, Wang X, Zhang J, et al. Siglec-9 acts as an immune-checkpoint molecule on macrophages in glioblastoma, restricting T-cell priming and immunotherapy response. Nat Cancer. 2023;4(9):1273–1291. doi:10.1038/s43018-023-00598-9

142. Naimi A, Mohammed RN, Raji A, et al. Tumor immunotherapies by immune checkpoint inhibitors (ICIs); the pros and cons. Cell Commun Signal. 2022;20(1):44. doi:10.1186/s12964-022-00854-y

143. Ishida Y, Agata Y, Shibahara K, et al. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11(11):3887–3895. doi:10.1002/j.1460-2075.1992.tb05481.x

144. Zak KM, Grudnik P, Magiera K, et al. Structural Biology of the Immune Checkpoint Receptor PD-1 and Its Ligands PD-L1/PD-L2. Structure. 2017;25(8):1163–1174. doi:10.1016/j.str.2017.06.011

145. Ossato A, Damuzzo V, Baldo P, et al. Immune checkpoint inhibitors as first line in advanced melanoma: evaluating progression-free survival based on reconstructed individual patient data. Cancer Med. 2023;12(3):2155–2165. doi:10.1002/cam4.5067

146. Wang JY, Wu L. First-line immunotherapy for advanced non-small cell lung cancer: current progress and future prospects. Cancer Biol Med. 2024;21(2):117–124.

147. Haddad RI, Harrington K, Tahara M, et al. Nivolumab Plus Ipilimumab Versus EXTREME Regimen as First-Line Treatment for Recurrent/Metastatic Squamous Cell Carcinoma of the Head and Neck: the Final Results of CheckMate 651. J Clin Oncol. 2023;41(12):2166–2180. doi:10.1200/JCO.22.00332

148. Liang X, Chen X, Li H, et al. Immune checkpoint inhibitors in first-line therapies of metastatic or early triple-negative breast cancer: a systematic review and network meta-analysis. Front Endocrinol. 2023;14. doi:10.3389/fendo.2023.1137464

149. Zhang L, Huang L, Liu Z, et al. Immune Checkpoint Inhibitor Plus Chemotherapy as First-Line Treatment for Advanced Gastric or Gastroesophageal Junction Cancer: a Systematic Review and Meta-Analysis. Technol Cancer Res Treat. 2024;23. doi:10.1177/15330338241273286

150. Perez-Nunez I, Rozalén C, Palomeque JÁ, et al. LCOR mediates interferon-independent tumor immunogenicity and responsiveness to immune-checkpoint blockade in triple-negative breast cancer. Nat Cancer. 2022;3(3):355–370. doi:10.1038/s43018-022-00339-4

151. Peng W, Chen JQ, Liu C, et al. Loss of PTEN Promotes Resistance to T Cell–Mediated Immunotherapy. Cancer Discov. 2016;6(2):202–216. doi:10.1158/2159-8290.CD-15-0283

152. Kirchhammer N, Trefny MP, Auf der Maur P, et al. Combination cancer immunotherapies: emerging treatment strategies adapted to the tumor microenvironment. Sci Transl Med. 2022;14(670):eabo3605. doi:10.1126/scitranslmed.abo3605

153. Chen L, Guo D. The functions of tumor suppressor PTEN in innate and adaptive immunity. Cell mol Immunol. 2017;14(7):581–589. doi:10.1038/cmi.2017.30

154. Lin Y-X, Wang Y, Ding J, et al. Reactivation of the tumor suppressor PTEN by mRNA nanoparticles enhances antitumor immunity in preclinical models. Sci Transl Med. 2021;13(599). doi:10.1126/scitranslmed.aba9772.

155. Li F, Zhang XQ, Ho W, et al. mRNA lipid nanoparticle-mediated pyroptosis sensitizes immunologically cold tumors to checkpoint immunotherapy. Nat Commun. 2023;14(1):4223.

156. Xiao YL, Chen J, Zhou H, et al. Combining p53 mRNA nanotherapy with immune checkpoint blockade reprograms the immune microenvironment for effective cancer therapy. Nat Commun. 2022;13(1). doi:10.1038/s41467-022-28279-8.

157. Liu LN, Wang Y, Miao L, et al. Combination Immunotherapy of MUC1 mRNA Nano-vaccine and CTLA-4 Blockade Effectively Inhibits Growth of Triple Negative Breast Cancer. Mol Ther. 2018;26(1):45–55. doi:10.1016/j.ymthe.2017.10.020

158. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A. 1989;86(24):10024–10028. doi:10.1073/pnas.86.24.10024

159. Gargett T, Yu W, Dotti G, et al. GD2-specific CAR T Cells Undergo Potent Activation and Deletion Following Antigen Encounter but can be Protected From Activation-induced Cell Death by PD-1 Blockade. Mol Ther. 2016;24(6):1135–1149. doi:10.1038/mt.2016.63

160. Hoskin D, Mader J, Furlong S, et al. Inhibition of T cell and natural killer cell function by adenosine and its contribution to immune evasion by tumor cells (Review). Int J Oncol. 2008. doi:10.3892/ijo.32.3.527

161. Riley RS, June CH, Langer R, et al. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov. 2019;18(3):175–196. doi:10.1038/s41573-018-0006-z

162. Porter DL, Hwang W-T, Frey NV, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7(303):303ra139. doi:10.1126/scitranslmed.aac5415

163. Zhang Y, Hou X, Du S, et al. Close the cancer–immunity cycle by integrating lipid nanoparticle–mRNA formulations and dendritic cell therapy. Nat Nanotechnol. 2023;18(11):1364–1374. doi:10.1038/s41565-023-01453-9

164. Parayath NN, Stephan SB, Koehne AL, et al. In vitro-transcribed antigen receptor mRNA nanocarriers for transient expression in circulating T cells in vivo. Nat Commun. 2020;11(1):6080. doi:10.1038/s41467-020-19486-2

165. Beatty GL, Haas AR, Maus MV, et al. Mesothelin-Specific Chimeric Antigen Receptor mRNA-Engineered T Cells Induce Antitumor Activity in Solid Malignancies. Cancer Immunol Res. 2014;2(2):112–120. doi:10.1158/2326-6066.CIR-13-0170

166. Sebastian M, Schröder A, Scheel B, et al. A phase I/IIa study of the mRNA-based cancer immunotherapy CV9201 in patients with stage IIIB/IV non-small cell lung cancer. Cancer Immunol Immunother. 2019;68(5):799–812. doi:10.1007/s00262-019-02315-x

167. Papachristofilou A, Hipp MM, Klinkhardt U, et al. Phase Ib evaluation of a self-adjuvanted protamine formulated mRNA-based active cancer immunotherapy, BI1361849 (CV9202), combined with local radiation treatment in patients with stage IV non-small cell lung cancer. J Immunother Cancer. 2019;7(1):38. doi:10.1186/s40425-019-0520-5

168. Sahin U, Oehm P, Derhovanessian E, et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature. 2020;585(7823):107–112. doi:10.1038/s41586-020-2537-9

169. Reinhard K, Rengstl B, Oehm P, et al. An RNA vaccine drives expansion and efficacy of claudin-CAR-T cells against solid tumors. Science. 2020;367(6476):446–453. doi:10.1126/science.aay5967

170. Haanen JBAG, Mackensen A, Koenecke C, et al. Abstract CT002: BNT211: a Phase I trial to evaluate safety and efficacy of CLDN6 CAR-T cells and CARVac-mediated in vivo expansion in patients with CLDN6-positive advanced solid tumors. Cancer Res. 2022;82(12_Supplement):CT002–CT002. doi:10.1158/1538-7445.AM2022-CT002

171. Xiao K, Lai Y, Yuan W, et al. mRNA‐based chimeric antigen receptor T cell therapy: basic principles, recent advances and future directions. Interdisciplinary Med. 2023;2(1):e20230036.

172. Biederstadt A, Rezvani K. Engineering the next generation of CAR-NK immunotherapies. Int J Hematol. 2021;114(5):554–571. doi:10.1007/s12185-021-03209-4

173. Klingemann H, Boissel L, Toneguzzo F. Natural Killer Cells for Immunotherapy – advantages of the NK-92 Cell Line over Blood NK Cells. Front Immunol. 2016;7:91. doi:10.3389/fimmu.2016.00091

174. Zhang C, Oberoi P, Oelsner S, et al. Chimeric Antigen Receptor-Engineered NK-92 Cells: an Off-the-Shelf Cellular Therapeutic for Targeted Elimination of Cancer Cells and Induction of Protective Antitumor Immunity. Front Immunol. 2017;8:533. doi:10.3389/fimmu.2017.00533

175. Khawar MB, Sun H. CAR-NK Cells: from Natural Basis to Design for Kill. Front Immunol. 2021;12:707542. doi:10.3389/fimmu.2021.707542

176. Sabbah M, Jondreville L, Lacan C, et al. CAR-NK Cells: a Chimeric Hope or a Promising Therapy? Cancers. 2022;14(15):3839. doi:10.3390/cancers14153839

177. Georgiou-Siafis SK, Miliotou AN, Ntenti C, et al. An Innovative PTD-IVT-mRNA Delivery Platform for CAR Immunotherapy of ErbB(+) Solid Tumor Neoplastic Cells. Biomedicines. 2022;10(11):2885. doi:10.3390/biomedicines10112885

178. Xiao L, Cen D, Gan H, et al. Adoptive Transfer of NKG2D CAR mRNA-Engineered Natural Killer Cells in Colorectal Cancer Patients. Mol Ther. 2019;27(6):1114–1125. doi:10.1016/j.ymthe.2019.03.011

179. Yang Z, Liu Y, Zhao K, et al. Dual mRNA co-delivery for in situ generation of phagocytosis-enhanced CAR macrophages augments hepatocellular carcinoma immunotherapy. J Control Release. 2023;360:718–733. doi:10.1016/j.jconrel.2023.07.021

180. Wang T, Upponi JR, Torchilin VP. Design of multifunctional non-viral gene vectors to overcome physiological barriers: dilemmas and strategies. Int J Pharm. 2012;427(1):3–20. doi:10.1016/j.ijpharm.2011.07.013

181. Zhang D, Wang G, Yu X, et al. Enhancing CRISPR/Cas gene editing through modulating cellular mechanical properties for cancer therapy. Nature Nanotechnol. 2022;17(7):777. doi:10.1038/s41565-022-01122-3

182. Zhou F, Huang L, Li S, et al. From structural design to delivery: mRNA therapeutics for cancer immunotherapy. Exploration. 2023;4(2). doi:10.1002/EXP.20210146.

183. Zhou L, Yi W, Zhang Z, et al. STING agonist-boosted mRNA immunization via intelligent design of nanovaccines for enhancing cancer immunotherapy. Natl Sci Rev. 2023;10(10):nwad214. doi:10.1093/nsr/nwad214

184. Shariati A, Khani P, Nasri F, et al. mRNA cancer vaccines from bench to bedside: a new era in cancer immunotherapy. Biomarker Res. 2024;12(1). doi:10.1186/s40364-024-00692-9.

185. Qin QS, Yan H, Gao W, et al. Engineered mRNAs With Stable Structures Minimize Double-stranded RNA Formation and Increase Protein Expression. J mol Biol. 2024;436(22):168822. doi:10.1016/j.jmb.2024.168822

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.