4,303
Views
0
CrossRef citations to date
0
Altmetric
Immunotherapy - Cancer

Recent progress in mRNA cancer vaccines

, &
Article: 2307187 | Received 28 Sep 2023, Accepted 16 Jan 2024, Published online: 28 Jan 2024

ABSTRACT

The research and development of messenger RNA (mRNA) cancer vaccines have gradually overcome numerous challenges through the application of personalized cancer antigens, structural optimization of mRNA, and the development of alternative RNA-based vectors and efficient targeted delivery vectors. Clinical trials are currently underway for various cancer vaccines that encode tumor-associated antigens (TAAs), tumor-specific antigens (TSAs), or immunomodulators. In this paper, we summarize the optimization of mRNA and the emergence of RNA-based expression vectors in cancer vaccines. We begin by reviewing the advancement and utilization of state-of-the-art targeted lipid nanoparticles (LNPs), followed by presenting the primary classifications and clinical applications of mRNA cancer vaccines. Collectively, mRNA vaccines are emerging as a central focus in cancer immunotherapy, offering the potential to address multiple challenges in cancer treatment, either as standalone therapies or in combination with current cancer treatments.

This article is part of the following collections:
Cancer Vaccines

Introduction

The field of oncology has recently witnessed a significant advancement with the introduction of mRNA-based tumor vaccines, signaling a new epoch in cancer immunotherapy.Citation1 These vaccines utilize mRNA’s versatility to elicit potent immune responses against cancer by encoding tumor antigens. This approach prompts the immune system to recognize and eliminate cancer cells displaying these antigens.Citation2

One of the key advantages of mRNA vaccines is their exceptional safety profile. Unlike DNA-based therapies, mRNA vaccines do not integrate into the host genome, thereby negating the risk of insertional mutagenesis. Furthermore, mRNA’s ability to concurrently encode multiple antigens boosts vaccine efficacy and paves the way for the development of personalized vaccines. The transient nature of mRNA expression ensures that antigen stimulation is temporary, effectively reducing the likelihood of chronic inflammations or autoimmune responses.Citation3 Additionally, the rapid design and synthesis of mRNA vaccines enable swift responses to evolving oncological challenges, including customizing vaccines to the unique tumor characteristics of individual patients, marking a significant stride toward personalized cancer therapy.Citation4

Although mRNA cancer vaccines are showing considerable promise, their broader application is hindered by several challenges. Challenges among these are the inherent instability of mRNA, the need for efficient and targeted delivery systems, and the possibility of immune tolerance, as well as evasion by tumor cells. These challenges present significant obstacles that are the focus of current research efforts.Citation5 Consequently, the optimization of mRNA sequences for maximal protein expression, ensuring mRNA stability, and enhancing delivery methods to achieve optimal cellular uptake and antigen presentation are pivotal areas of ongoing research.

The successful development of mRNA vaccines for Corona Virus Disease 2019 (COVID-19) has overcome technical bottlenecks via a comprehensive optimization of the mRNA construct. This achievement was made possible through the redesign and enhancement of each aspect of the mRNA, thereby improving its stability and expression efficiency.Citation6–9 Furthermore, the advancement of diverse mRNA delivery systems has facilitated the efficient and targeted delivery of mRNA molecules.Citation10 mRNA cancer vaccine benefits from all of these advancements as well, and as clinical trials progress, mRNA cancer vaccines are emerging as a promising tool in the fight against cancer.

This review delves into the development and application of mRNA vaccines in the field of oncology. We provide a comprehensive summary of the development history of mRNA cancer vaccines, highlighting the structural optimization of antigen expression vectors to enhance mRNA stability and translation efficiency. Additionally, we discuss the application and future prospects of other RNA-based antigen expression vectors in cancer vaccine development. An overview of the LNP delivery system used for mRNA vaccines is presented, with an emphasis on targeting strategies. We also elucidate various types of mRNA cancer vaccines and their current state of advancement. It is anticipated that mRNA vaccines will surmount various challenges in cancer treatment and emerge as a major modality in cancer immunotherapy.

The concept of mRNA cancer vaccine as an innovative therapeutic approach

The popularity of mRNA vaccines has significantly surged with the application of two COVID-19 vaccines, marking a pivotal moment in their development and public recognition.Citation6,Citation7,Citation9 However, the investigation and development of mRNA vaccines long predates the COVID-19 pandemic. The journey began following the successful synthesis of mRNA in vitro and its expression in vivo ().Citation11 In the early 1990s, efforts to develop mRNA vaccines for tumor prevention commenced. A landmark study by Martinon F. et al. demonstrated the induction of anti-influenza cytotoxic T lymphocytes (CTLs) in mice using liposome-encapsulated mRNA encoding the influenza virus nucleoprotein (NP), showcasing the first instance of using mRNA to stimulate in vivo antigen-specific immune responses.Citation12 Boczkowski et al. furthered this research by transfecting mRNA encoding tumor antigens into dendritic cells, leading to the production of CTLs both in vitro and in vivo. This study was pivotal in demonstrating mRNA’s potential to induce anti-tumor immune responses.Citation13 However, early mRNA molecules faced limitations due to instability and low expression efficiency. Unmodified mRNA was initially noted for its high immunogenicity, activating innate immune responses through toll-like receptors (TLR3, TLR7, TLR8, and TLR9).Citation14–17 The subsequent incorporation of modified nucleosides (such as m5C, m6A, m5U, s2U, and pseudouridine) into mRNA formulations markedly reduced their immunogenicity, with the complete replacement of uridine with pseudouridine improving mRNA stability and translation efficiency.Citation18,Citation19 The scientists who made these breakthrough findings, Drs. Katalin Karikó and Drew Weissman, were awarded the 2023 Nobel Prize in Physiology or Medicine for their discoveries concerning nucleoside base modifications that enabled the development of effective mRNA vaccines against COVID-19.

Table 1. Recent milestone events in the development of mRNA vaccines.

The investigation into liposomal delivery of siRNA provided valuable insights for the advancement of LNP systems.Citation20 LNPs have since been validated as effective carriers for in vivo mRNA delivery in various preventive and therapeutic vaccination strategies.Citation21 The COVID-19 pandemic has accentuated the immense potential of mRNA technology,Citation6,Citation7,Citation9 offering valuable insights for the development of mRNA-based cancer vaccines. Furthermore, the advent of personalized neoantigen vaccines has yielded substantial clinical benefits for patients suffering from a variety of malignancies.Citation22 The progression of mRNA-4157 in combination with pembrolizumab to Phase III clinical trials for melanoma patients represents a significant milestone in the field of personalized mRNA cancer vaccines. Current research on mRNA cancer vaccines primarily focuses on eliminating the immunogenicity and instability of mRNA, developing efficient and targeted mRNA delivery systems, and advancing the development of personalized mRNA cancer vaccines. The subsequent sections of this paper will provide a more comprehensive exploration of these critical aspects.

RNA-based vectors for antigen expression

Design strategies for in vitro transcribed mRNA

The structure of in vitro transcribed (IVT) mRNA, mirroring eukaryotic mRNA, includes a 5’ cap, a protein-coding open reading frame (ORF), 5’ and 3’ untranslated regions (UTRs), and a 3’ poly(A) tail. The structural complexity of each mRNA segment significantly impacts translation efficiency, properties, and immunogenicity,Citation23,Citation24 making optimization crucial for the efficacy of mRNA technologies.

For IVT mRNA, the 5’-cap structure (m7GpppN) is vital for stability and translation efficiency.Citation25 Two main methods for cap formation are Vaccinia capping enzyme (VCE)-mediated capping and co-transcriptional capping. VCE capping offers specificity but at higher cost and complexity, while co-transcriptional capping is simpler but less effective.Citation26 Recent advancements include the use of VCE to introduce GTP analogs at the 5’ end, creating modified mRNAs.Citation27 Moreover, Integrating T7 RNA polymerase with VCE simplifies cap-modified mRNA synthesis, reducing the need for expensive analogs.Citation28 Continuous cap analog optimization has significantly improved mRNA stability and transcription, enhancing its research and application potential ().Citation29–31 Furthermore, a recent study shows a shift from cap1 to cap2 in mRNA caps with age, decreasing RIG-I receptor activation and increasing mRNA stability.Citation32 Incorporating cap2 may reduce immunogenicity and extend mRNA lifespan.

Table 2. Different generation of mRNA cap analogs.

The 5’ untranslated region (UTR) of mRNA, crucial for gene expression regulation, can be optimized for enhanced translation efficiency.Citation33 For instance, adjustment like altering the AUG codon position or incorporating unstructured sequences can improve translation efficiency.Citation34,Citation35 A shorter 5’ UTR, around 17 nucleotides, also aids in reducing variability and translation issues.Citation36

Similar to the 5‘UTR, the 3’ UTR also harbors several regulatory elements and plays a pivotal role in regulating mRNA stability, subcellular localization, and translation efficiency.Citation37 Key strategies include avoiding miRNA binding sites, eliminating adenine/uridine-rich elements, and minimizing its length.Citation38–40 Despite challenges in designing optimal UTRs, natural UTRs, such as the 5’ UTR of human α-globin mRNA used in the BNT162b2 COVID-19 vaccine, are often preferred for their proven gene expression efficiency.Citation41 However, UTRs impact different cell types variably, requiring tailored optimization for specific target cells. The rapid advancements in high-throughput screening (HTS) technology and genetic algorithms have facilitated the identification of potential optimization sequences from a diverse range of 5‘UTR and 3‘UTR libraries derived from cellular or viral genomes.Citation42,Citation43

Optimization of the coding sequence (CDS) in mRNA is crucial for effective gene expression. Common strategies involve replacing rare codons with more prevalent synonymous ones in host cells.Citation44 In 2005, Karikó et al. addressed the challenges of high mRNA immunogenicity and low protein expression via incorporating the modified nucleosides, as mentioned earlier.Citation18,Citation19 Additionally, factors like GC content and mRNA secondary structure significantly influence mRNA translation.Citation45,Citation46 However, these optimization strategies, while effective, have limitations in fully harnessing the potential of mRNA sequences for optimal performance. Within the field of precision medicine, the development of mRNA design algorithms is a promising approach to overcome these challenges. These algorithms focus on identifying mRNA sequences with stable secondary structures, enhanced translation efficiency, and optimized codons, expected to significantly contribute to advancing mRNA-based technologies.Citation47,Citation48 We have listed a few here with noted features and advantages/disadvantages ().

Table 3. mRNA design algorithms.

The Poly(A) tail, characterized by a long repetitive polyadenylate sequence, plays a crucial role in defending mRNA against enzymatic deacetylation and degradation, thereby augmenting its stability and translational efficacy.Citation31 The incorporation of short sequences or thiophosphate between polyadenylate sequences has been shown to further enhance stability. For example, the Poly(A) tail sequence of BNT162b2 comprises a structure of 30A + 10GCAUAUGACU +70A.Citation49,Citation50 Additionally, a recent study demonstrated that a Poly(A) tail containing 20% cytidine can protect mRNA from deacetylase activity and significantly improve the translation efficiency and half-life of mRNA.Citation51,Citation52

Optimization strategies for mRNA structure can be synergistically employed to enhance protein production. The advancement of mRNA vaccines underscores the need for a universally standardized mRNA design and screening protocol. Comprehensive screening and precise customization of target-specific sequences hold the promise of diverse enhancements in future mRNA designs. This approach enables personalized adaptations to specific cell types and unique microenvironment, thereby maximizing the protein synthesis efficiency of each mRNA transcript.

Other RNA-based antigen expression vectors

The current instability of mRNA presents significant limitations in its application as a therapeutic agent, prompting recent research to focus on the exploration of alternative RNA vectors. Besides linear mRNA, several other types of RNA vectors have advanced quickly (). One such promising vector is the self-amplifying RNA (saRNA), which is derived from a virus and carries the RNA polymerase sequence. This saRNA exhibits self-amplification and translation capabilities, enabling it to elicit a detectable protective immune response with only minimal quantities.Citation53 This characteristic suggests that saRNA could be effectively applied in cancer vaccines.Citation54 Several preclinical studies exploring this potential are already underway. For instance, a comparative study demonstrated that saRNA targeting the human papillomavirus (HPV) exhibited stronger anti-tumor effects compared to both unmodified and modified non-replicating mRNAs.Citation55 However, it is currently unclear whether RNA polymerase can trigger an immune response in vivo. Furthermore, the larger size of saRNA may pose new challenges for existing delivery systems. One potential strategy to address this involves spatially segregating the sequence encoding RNA polymerase from the sequence of the target protein.

Figure 1. Different types of RNA expression vectors used in mRNA cancer vaccines.

Figure 1. Different types of RNA expression vectors used in mRNA cancer vaccines.

Circular RNA (circRNA), with its covalent circular structure, offers enhanced stability over mRNA due to its resistance to RNase. This characteristic provides distinct advantages for RNA vaccines.Citation56,Citation57 The circRNA vaccine platform developed by Qu et al. has shown protective efficacy against multiple COVID-19 variants, contributing valuable insights to the development of circRNA-based cancer vaccines.Citation58 Studies have demonstrated that circRNA molecules encoding antigens or cytokines are more effective in tumor suppression than mRNA vaccines.Citation59,Citation60 Additionally, Amaya et al. highlighted circRNA’s role as an adjuvant in enhancing anti-tumor effects.Citation61 However, the precise mechanisms behind these effects are yet to be fully understood. In a recent study, Huang et al. showed that circFAM53B, a tumor-specific circRNA, initiates anti-tumor immunity by expressing a specific peptide, underlining circRNA’s potential in immunotherapy.Citation62

Despite these developments, challenges remain in circRNA synthesis, including suboptimal cyclization efficiency and high costs of reagents like enzymes. Nonetheless, circRNA is emerging as a promising class of RNA-based therapeutics and vaccines with significant future potential.

A recent advancement involves hybridizing short double-stranded RNA (dsRNA) with single-stranded mRNA to form a dendritic, comb-like structure, serving both as an antigen expression vector and immune adjuvant. This method allows precise control over immune stimulation strength in mRNA vaccines by manipulating dsRNA’s length, sequence, and quantity.Citation63 This comb-structured RNA design, applicable to various mRNA vaccines under development, is anticipated to significantly enhance their efficacy and safety.

Delivery platforms

A crucial component of mRNA therapy is the delivery system. Common delivery systems include protamine, lipoplex (LPX),Citation64 lipopolyplex (LPP),Citation65 virus-like particles (VLPs),Citation66 dendritic cell,Citation67 and LNPs.Citation68 Due to its most advanced status in current application, this review primarily focuses on the application of LNP technology.

LNPs typically consist of cationic or ionizable lipids, helper lipids, cholesterol, and PEGylated lipids.Citation69 Cationic or ionizable lipids, positively charged in acidic conditions, bind negatively charged mRNA through electrostatic interactions. In endosomes, these lipids protonate, facilitating mRNA escape for cytoplasmic antigen expression.Citation70 Cholesterol improves LNP stability, aiding endocytosis and endosomal escape.Citation71 while Phospholipids promote mRNA encapsulation and stability, while PEGylation prevents in vivo protein-LNP interactions.Citation72

Originally developed for delivering siRNA,Citation73 LNP technology has been adapted for mRNA encapsulation and delivery.Citation74 While offering efficient mRNA packaging and biocompatibility, most LNPs accumulate in the liver post-administration, potentially leading to hepatotoxicity.Citation75 The current focus in LNP-mRNA cancer vaccines is on targeting strategies for selective mRNA delivery to specific organs or cells.

Recent advancements in LNP targeting include passive or active methods to tumors.Citation76 Effective anti-tumor immunity hinges on delivering antigen-encoding mRNA to antigen-presenting cells (APCs), with LNPs designed to target immune cells expected to bolster anti-tumor responses and minimize systemic side effects.Citation8 Subcutaneous injections near lymph nodes, as a passive targeting approach, have proven effective in various studies.Citation77–79 However, subcutaneously administered LNPs can also accumulate in organs like the liver and spleen, potentially impacting vaccine efficacy and safety.Citation80

Modifying lipid structure of LNP is another targeting strategy, as LNPs naturally interact with plasma proteins in vivo, forming a “protein corona” that alters their interactions with cells and organs.Citation81 Preclinical studies show that these modifications can significantly affect mRNA delivery and LNP targeting.Citation82–84 Lipid 113-O12B, for example, specifically targets lymph nodes and is uptaken by a significant proportion of dendritic cells and macrophages, inducing robust immunity and, when combined with anti-PD1 antibody, achieving a 40% complete tumor remission rate.Citation85 However, in vivo screening has limitations, including the ethical concerns of animal use and the inability of in vitro screening to accurately predict in vivo delivery efficiency.Citation86 Moreover, the specific mechanism of these lipids remains unclear, and the limited understanding of the interaction between LNPs and biological components hampers the prediction of the in vivo targeting behavior of novel LNPs.

Regulating the internal and/or external charges of LNPs is crucial for targeting.Citation87,Citation88 SORT nanoparticles, developed for tissue-specific mRNA delivery and CRISPR/Cas gene editing, interact with serum proteins that bind to receptors on target organ cells, allowing selective organ targeting.Citation89 However, reliance on endogenous patient proteins for targeting may lead to off-target effects and inter-individual variability.Citation90,Citation91

Passive targeting of LNPs is popular for its simplicity and broad applicability, but it lacks specificity. Active targeting, while more specific and efficient, involves higher costs and potential immunogenicity risks. Targeted LNPs, therefore, represent a promising avenue in mRNA delivery, with future research needed to optimize both passive and active strategies to enhance LNP performance and safety in mRNA and drug delivery.

Classifying mRNA cancer vaccines

mRNA-based cancer vaccines that encode viral antigens

The development of mRNA-based cancer vaccines targeting viral antigens is an emerging field. These vaccines are particularly relevant for malignancies associated with viruses such as Human Papillomavirus (HPV), Hepatitis B (HBV), Epstein-Barr (EBV), and HIV, which are implicated in cervical, liver, and nasopharyngeal cancers, among others.Citation92–94 Traditional vaccines against these viruses have predominantly been prophylactic.Citation95–97 However, mRNA-based vaccines offer a therapeutic approach by stimulating the body’s immune system to recognize and attack cells expressing these viral antigens.Citation1 For example, a recent study showed that a messenger RNA-HPV therapeutic vaccine (mHTV) exhibited strong immunogenicity and anti-tumor effects in mice and non-human primates, positioning mHTV as a promising therapeutic and prophylactic vaccine for HPV.Citation98

The flexibility of mRNA vaccine platforms is particularly valuable in personalized medicine. Customizing vaccines to match individual patient’s viral antigen profiles can address the challenge posed by the evolving nature of viral oncogenes, offering more effective treatment. HIV, known for its rapid mutation and diversity, presents a significant challenge in vaccine development.Citation99 An mRNA-based HIV vaccine induced both humoral and cellular immune responses in mice and primates, with Rhesus monkeys showing a 79% reduction in SHIV exposure after receiving priming and booster vaccines.Citation100 A recent phase I clinical trial of the mRNA vaccine eOD-GT8 60mer against HIV showed good safety and induced broad neutralizing antibodies (bnAbs) in 97% of participants (35 out of 36), demonstrating the potential of mRNA vaccines in HIV prevention.Citation101 Additionally, mRNA vaccines offer a novel approach for chronic virus infections such as HBV.Citation102 A mRNA-based vaccine for treating Hepatitis B virus-associated hepatocellular carcinoma (HCC) (NCT05738447) is currently undergoing clinical trial.

EBV is linked to various cancers, including nasopharyngeal carcinoma, Hodgkin’s lymphoma, and gastric cancer.Citation103 The complex nature of EBV and its interactions with the host immune system present significant challenges for vaccine development. Notably, EBV’s latency within host cells renders it difficult for the immune system to detect and eradicate. To date, no vaccines have been approved for EBV infection, and the development of mRNA vaccines for EBV remains in the preclinical stage, though they have shown promising potential.Citation104,Citation105

Despite the potential of mRNA vaccines to prevent viral infections linked to cancer, vaccines targeting cancer-related viruses predominantly remain in preclinical stages. This is due to challenges such as viral mutation, immune evasion, and immune tolerance stemming from long-term infection.Citation106–108 Future research needs to focus on overcoming these hurdles to develop more effective mRNA vaccines against these viruses.

mRNA vaccines encoding tumor antigens

Tumor-associated antigens (TAAs)

TAAs are a class of antigens that are expressed at significantly higher levels in tumors compared to normal tissues. This overexpression designates them as prime targets for cancer vaccines. These vaccines, customizable based on individual TAAs, offer enhanced efficacy and reduced side effects compared to traditional therapies like chemotherapy or radiotherapy.Citation109 This personalized approach marks a significant advancement in oncology, presenting a more targeted and patient-specific treatment strategy. Some of the well-known TAAs are listed in the .

Table 4. A list of well-studied TAAs for different cancer types.

Despite their potential, mRNA vaccines targeting TAAs have limitations. TAAs, being non-mutated self-antigens, can lead to poor T cell responses and immune tolerance in clinical immunotherapy.Citation121 Additionally, TAA expression in normal cells might cause collateral damage. These vaccines may also be less effective in cancers where TAAs have low expression levels or mutations.Citation122 Strategies to overcome these limitations include co-delivering cancer antigens with immune-stimulatory molecules, enhancing immune activation in the tumor microenvironment, and combining vaccines with conventional treatments like chemotherapy or radiotherapy.Citation123–126 BNT111, targeting four common TAAs (NY-ESO-1, MAGE-A3, Tyrosinase, TPTE), is exemplary. In over 90% of cutaneous melanoma patients, BNT111 induced novel and enhanced preexisting immune responses against these antigens (Phase I clinical trial, NCT02410733). A phase II trial (BNT111–01, NCT04526899) is underway, combining BNT111 with Cemiplimab (an anti-PD1 antibody made by Regeneron Inc.) for advanced unresectable stage III or IV melanoma patients unresponsive to anti-PD-1 therapies.

In mRNA vaccines encoding TAAs, priority should be given to TAAs exclusively expressed in tumor cells, minimizing expression in normal cells. Comprehensive studies on TAAs’ expression patterns across tumors and normal tissues are crucial for identifying safer and more effective targets.

Tumor-specific antigens (TSAs)

Tumor-specific antigens (TSAs) are proteins or peptide fragments uniquely expressed or significantly overexpressed in cancer cells compared to normal cells. These antigens result from various oncogenic processes, including mutations, abnormal gene expression, and post-translational modifications.Citation1 Neoantigens are presented on the surface of cancer cells via Major Histocompatibility Complex (MHC) molecules. These neoantigens are absent in normal tissues, making them ideal targets for immunotherapeutic strategies. Their uniqueness to individual tumors circumvents the issues of tolerance and autoimmunity often associated with targeting shared tumor antigens. In 2014, Tran et al. successfully treated a bile duct cancer patient with ERBB2IP-specific CD4+ cells, achieving complete tumor regression,Citation127 demonstrating TSAs’ potential in inducing anti-tumor responses. They also found gene mutations in 9 out of 10 gastrointestinal tumor patients,Citation128 which indicates the widespread presence of tumor heterogeneity, with TSAs varying among patients even within the same tumor type. The development of personalized mRNA vaccines involves the identification and selection of neoantigens through advanced sequencing and bioinformatics approaches. Next-generation sequencing (NGS), high throughput screening and machine learning algorithms have facilitated TSA prediction by identifying mutations and aberrant transcription/translation events in individual tumor genome exons.Citation129–132

The clinical potential of personalized mRNA neoantigen vaccines is underpinned by their ability to induce robust and specific immune responses. Early-phase clinical trials have shown promising results, with evidence of vaccine-induced T cell responses and potential clinical benefits (). Ugur Sahin et al., identified mutations most likely to provoke immune responses in 13 melanoma patients and developed mRNA encoding up to 10 novel antigens. The clinical trial showed that among 13 patients, 8 had no recurrence within a year after vaccination, and of the remaining 5, 2 experienced tumor shrinkage, with 1 achieving complete remission combined with PD-1 antibody treatment.Citation133 mRNA-4157 and BNT122 are two typical personalized mRNA vaccines underway for clinical trials. mRNA-4157, encoding a repertoire of 34 antigens targeting unique mutations in individual patients’ tumor DNA, is undergoing clinical trials for melanoma, non-small cell lung cancer, and other solid tumors. The clinical trial testing mRNA-4157 combined with pembrolizumab (anti-PD1 made by Merck Inc.) in head and neck squamous cell carcinoma patients showed a 50% overall response rate and a 90% disease control rate. Two phase IIb clinical trials combining mRNA-4157 and pembrolizumab for high-risk melanoma patients reduced the risk of recurrence or death by 44% and 49%, respectively, suggesting more pronounced benefits with longer treatment durations.Citation134 The combination therapy of mRNA-4157 and pembrolizumab has advanced to Phase III clinical trials (NCT05933577), marking it as the first mRNA cancer vaccine to reach this stage.

Table 5. Selected representative mRNA vaccine clinical trials with published results.

Personalized mRNA vaccines offer promising treatments for cancers resistant to current immunotherapy, such like pancreatic ductal adenocarcinoma (PDAC), which shows resistance to immune checkpoint inhibitors (ICIs).Citation135 The abundance of neoantigens in pancreatic cancer makes neoantigen-based mRNA vaccines a viable therapeutic strategy. BNT122 employs mRNA to express 20 novel antigens in PDAC patients. When combined with chemotherapy and immune checkpoint therapy, BNT122 exhibits potential in decelerating the recurrence in PDAC patients, with a follow-up global randomized trial (IMCODE 003, BNT122) forthcoming.Citation136

TSA mRNA vaccines represent a highly personalized approach, tailoring each vaccine to an individual’s tumor antigen. Combined with immune checkpoint inhibitors, these vaccines offer new avenues for treating diseases long challenging to medical science.

mRNA-encoded immunomodulators

The immunosuppressive tumor microenvironment (TME) is characterized by limited T cell infiltration or an abundance of immune inhibitory cells, contributing to resistance against current therapies. Cytokines, crucial for regulating intercellular communication among immune cells, can efficiently transduce immune signals and orchestrate robust immune responses against target antigens. The delivery of cytokines, co-stimulating ligands, or other immune regulatory factors via mRNA can effectively remodel the TME and enhance tumor sensitivity to various immunotherapies.

Innovative approaches include the delivery of mRNA encoding the tumor suppressor p53 to liver cancer cells using nanoparticles, combined with anti-PD-1 monoclonal antibodies. This strategy has shown effectiveness in modulating the TME and achieving anti-tumor effects.Citation137 Additionally, previous studies have exploited mRNA encoding caTLR4 and co-stimulatory ligands CD70 and CD40L to stimulate immune responses in APCs. The followed enhancement in T cell functionality has been demonstrated to combat tumors effectively and suppress early resectable breast cancer lesions in mice.Citation138 mRNA-2752, an mRNA vaccine encoding three immune regulatory factors, is currently under Phase I and Phase II clinical trials to assess its safety and tolerability, both as a standalone treatment and in combination with fixed-dose durvalumab (anti-PDL1 antibody made by AstraZeneca Inc.) in patients with advanced malignancies.Citation129

Conclusions and future perspectives

The successful deployment of COVID-19 vaccines has significantly accelerated the advancement of mRNA technology in oncology. Utilizing innovative RNA-based vectors and targeted strategies, new mRNA vaccine platforms are being developed to target an extensive range of tumor antigens. Notably, the progress in personalized mRNA vaccine marks a considerable breakthrough in cancer treatment. These vaccines show enhanced potential in clinical trials, particularly when combined with existing therapies, including ICI and traditional treatments.

Despite comprehensive development, mRNA cancer vaccines still confront multiple challenges. Further research is essential to refine the development and application of these vaccines. Continued improvements in vaccine design, delivery technologies, and rapid identification of tumor neoantigens are crucial, positioning mRNA vaccines as a promising tool in personalized cancer therapy.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

This work was supported by the National Natural Science Foundation of China [Grant No. 32370923, 81972692].

References

  • Sahin U, Türeci Ö. Personalized vaccines for cancer immunotherapy. Science. 2018;359(6382):1355–12. doi:10.1126/science.aar7112.
  • Fiedler K, Lazzaro S, Lutz J, Rauch S, Heidenreich R. mRNA Cancer vaccines. Recent Results Cancer Res, 2016;209:61–85. doi:10.1007/978-3-319-42934-2_5.
  • Weissman D, Karikó K. mRNA: fulfilling the promise of gene therapy. Mol Ther. 2015;23(9):1416–17. doi:10.1038/mt.2015.138.
  • Lin MJ, Svensson-Arvelund J, Lubitz GS, Marabelle A, Melero I, Brown BD, Brody JD. Cancer vaccines: the next immunotherapy frontier. Nat Cancer. 2022;3(8):911–26. doi:10.1038/s43018-022-00418-6.
  • Pardi N, Hogan MJ, Weissman D. Recent advances in mRNA vaccine technology. Curr Opin Immunol. 2020;65:14–20. doi:10.1016/j.coi.2020.01.008.
  • Baden LR, El Sahly HM, Essink B, Kotloff K, Frey S, Novak R, Diemert D, Spector SA, Rouphael N, Creech CB, et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med. 2021;384(5):403–16. doi:10.1056/NEJMoa2035389.
  • Corbett KS, Edwards DK, Leist SR, Abiona OM, Boyoglu-Barnum S, Gillespie RA, Himansu S, Schäfer A, Ziwawo CT, DiPiazza AT, et al. SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness. Nature. 2020;586(7830):567–71. doi:10.1038/s41586-020-2622-0.
  • Miao L, Zhang Y, Huang L. mRNA vaccine for cancer immunotherapy. Mol Cancer. 2021;20(1):41. doi:10.1186/s12943-021-01335-5.
  • Polack FP, Thomas SJ, Kitchin N, Absalon J, Gurtman A, Lockhart S, Perez JL, Pérez Marc G, Moreira ED, Zerbini C, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med. 2020;383(27):2603–15. doi:10.1056/NEJMoa2034577.
  • Eygeris Y, Gupta M, Kim J, Sahay G. Chemistry of lipid nanoparticles for RNA delivery. Acc Chem Res. 2022;55(1):2–12. doi:10.1021/acs.accounts.1c00544.
  • Wolff JA, Malone RW, Williams P, Chong W, Acsadi G, Jani A, Felgner PL. Direct gene transfer into mouse muscle in vivo. Science. 1990;247(4949 Pt 1):1465–8. doi:10.1126/science.1690918.
  • Martinon F, Krishnan S, Lenzen G, Magné R, Gomard E, Guillet JG, Lévy JP, Meulien P. Induction of virus-specific cytotoxic T lymphocytes in vivo by liposome-entrapped mRNA. Eur J Immunol. 1993;23(7):1719–22. doi:10.1002/eji.1830230749.
  • Boczkowski D, Nair SK, Snyder D, Gilboa E. Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J Exp Med. 1996;184(2):465–72. doi:10.1084/jem.184.2.465.
  • Alexopoulou L, Holt AC, Medzhitov R, Flavell RA. Recognition of double-stranded RNA and activation of NF-kappaB by toll-like receptor 3. Nature. 2001;413(6857):732–8. doi:10.1038/35099560.
  • Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science. 2004;303(5663):1529–31. doi:10.1126/science.1093616.
  • Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S, Lipford G, Wagner H, Bauer S. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science. 2004;303(5663):1526–29. doi:10.1126/science.1093620.
  • Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, Matsumoto M, Hoshino K, Wagner H, Takeda K, et al. A Toll-like receptor recognizes bacterial DNA. Nature. 2000;408(6813):740–45. doi:10.1038/35047123.
  • Karikó K, Buckstein M, Ni H, Weissman D. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity. 2005;23(2):165–75. doi:10.1016/j.immuni.2005.06.008.
  • Karikó K, Muramatsu H, Welsh FA, Ludwig J, Kato H, Akira S, Weissman D. Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stability. Mol Ther. 2008;16(11):1833–40. doi:10.1038/mt.2008.200.
  • Cullis PR, Chonn A. Recent advances in liposome technologies and their applications for systemic gene delivery. Adv Drug Deliv Rev. 1998;30(1–3):73–83. doi:10.1016/s0169-409x(97)00108-7.
  • Pardi N, Tuyishime S, Muramatsu H, Kariko K, Mui BL, Tam YK, Madden TD, Hope MJ, Weissman D. Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes. J Control Release. 2015;217:345–51. doi:10.1016/j.jconrel.2015.08.007.
  • Blass E, Ott PA. Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat Rev Clin Oncol. 2021;18(4):215–29. doi:10.1038/s41571-020-00460-2.
  • Bae H, Coller J. Codon optimality-mediated mRNA degradation: linking translational elongation to mRNA stability. Mol Cell. 2022;82(8):1467–76. doi:10.1016/j.molcel.2022.03.032.
  • Hanson G, Coller J. Codon optimality, bias and usage in translation and mRNA decay. Nat Rev Mol Cell Biol. 2018;19(1):20–30. doi:10.1038/nrm.2017.91.
  • Shanmugasundaram M, Senthilvelan A, Kore AR. Recent advances in modified Cap Analogs: synthesis, biochemical properties, and mRNA based vaccines. Chem Rec. 2022;22(8):e202200005. doi:10.1002/tcr.202200005.
  • Ramanathan A, Robb GB, Chan SH. mRNA capping: biological functions and applications. Nucleic Acids Res. 2016;44(16):7511–26. doi:10.1093/nar/gkw551.
  • Ohno H, Akamine S, Mochizuki M, Hayashi K, Akichika S, Suzuki T, Saito H. Versatile strategy using vaccinia virus-capping enzyme to synthesize functional 5’ cap-modified mRNAs. Nucleic Acids Res. 2023;51(6):e34. doi:10.1093/nar/gkad019.
  • Nwokeoji AO, Chou T, Nwokeoji EA. Low resource integrated platform for production and analysis of capped mRNA. ACS Synth Biol. 2023;12(1):329–39. doi:10.1021/acssynbio.2c00609.
  • Chen Y, Guo D. Molecular mechanisms of coronavirus RNA capping and methylation. Virol Sin. 2016;31(1):3–11. doi:10.1007/s12250-016-3726-4.
  • Henderson JM, Ujita A, Hill E, Yousif-Rosales S, Smith C, Ko N, McReynolds T, Cabral CR, Escamilla-Powers JR, Houston ME. Cap 1 messenger RNA synthesis with Co-transcriptional CleanCap(®) analog by in vitro transcription. Curr Protoc. 2021;1(2):e39. doi:10.1002/cpz1.39.
  • Mockey M, Gonçalves C, Dupuy FP, Lemoine FM, Pichon C, Midoux P. mRNA transfection of dendritic cells: synergistic effect of ARCA mRNA capping with Poly(A) chains in cis and in trans for a high protein expression level. Biochem Biophys Res Commun. 2006;340(4):1062–8. doi:10.1016/j.bbrc.2005.12.105.
  • Despic V, Jaffrey SR. mRNA ageing shapes the Cap2 methylome in mammalian mRNA. Nature. 2023;614(7947):358–66. doi:10.1038/s41586-022-05668-z.
  • Schuster SL, Hsieh AC. The untranslated regions of mRnas in cancer. Trends Cancer. 2019;5(4):245–62. doi:10.1016/j.trecan.2019.02.011.
  • Kozak M. Leader length and secondary structure modulate mRNA function under conditions of stress. Mol Cell Biol. 1988;8(7):2737–44. doi:10.1128/MCB.8.7.2737.
  • Kozak M. Circumstances and mechanisms of inhibition of translation by secondary structure in eucaryotic mRNAs. Mol Cell Biol. 1989;9(11):5134–142. doi:10.1128/MCB.9.11.5134.
  • Trepotec Z, Aneja MK, Geiger J, Hasenpusch G, Plank C, Rudolph C. Maximizing the translational yield of mRNA therapeutics by minimizing 5’-UTRs. Tissue Eng Part A. 2019;25(1–2):69–79. doi:10.1089/ten.TEA.2017.0485.
  • Andreassi C, Riccio A. To localize or not to localize: mRNA fate is in 3‘UTR ends. Trends Cell Biol. 2009;19(9):465–74. doi:10.1016/j.tcb.2009.06.001.
  • Jia L, Qian SB. Therapeutic mRNA engineering from head to Tail. Acc Chem Res. 2021;54(23):4272–82. doi:10.1021/acs.accounts.1c00541.
  • Mayr C. Regulation by 3′-untranslated regions. Annu Rev Genet. 2017;51(1):171–94. doi:10.1146/annurev-genet-120116-024704.
  • Mayr C. What are 3′ UTRs doing? Cold Spring Harb Perspect Biol. 2019;11(10):a034728. doi:10.1101/cshperspect.a034728.
  • Wang C, Zhang Y, Dong Y. Lipid nanoparticle-mRNA formulations for therapeutic applications. Acc Chem Res. 2021;54(23):4283–93. doi:10.1021/acs.accounts.1c00550.
  • Castillo-Hair SM, Seelig G. Machine learning for designing next-generation mRNA therapeutics. Acc Chem Res. 2022;55(1):24–34. doi:10.1021/acs.accounts.1c00621.
  • Sample PJ, Wang B, Reid DW, Presnyak V, McFadyen IJ, Morris DR, Seelig G. Human 5’ UTR design and variant effect prediction from a massively parallel translation assay. Nat Biotechnol. 2019;37(7):803–9. doi:10.1038/s41587-019-0164-5.
  • Presnyak V, Alhusaini N, Chen YH, Martin S, Morris N, Kline N, Olson S, Weinberg D, Baker KE, Graveley BR, et al. Codon optimality is a major determinant of mRNA stability. Cell. 2015;160(6):1111–24. doi:10.1016/j.cell.2015.02.029.
  • Kudla G, Lipinski L, Caffin F, Helwak A, Zylicz M, Hurst LD. High guanine and cytosine content increases mRNA levels in mammalian cells. PLoS Biol. 2006;4(6):e180. doi:10.1371/journal.pbio.0040180.
  • Mauger DM, Cabral BJ, Presnyak V, Su SV, Reid DW, Goodman B, Link K, Khatwani N, Reynders J, Moore MJ, et al. mRNA structure regulates protein expression through changes in functional half-life. Proc Natl Acad Sci USA. 2019;116(48):24075–83. doi:10.1073/pnas.1908052116.
  • Wayment-Steele HK, Kim DS, Choe CA, Nicol JJ, Wellington-Oguri R, Watkins AM, Parra Sperberg RA, Huang PS, Participants E, Das R. Theoretical basis for stabilizing messenger RNA through secondary structure design. Nucleic Acids Res. 2021;49(18):10604–17. doi:10.1093/nar/gkab764.
  • Zhang H, Zhang L, Lin A, Xu C, Li Z, Liu K, Liu B, Ma X, Zhao F, Jiang H, et al. Algorithm for optimized mRNA design improves stability and immunogenicity. Nature. 2023;621(7978):396–403. doi:10.1038/s41586-023-06127-z.
  • Trepotec Z, Geiger J, Plank C, Aneja MK, Rudolph C. Segmented poly(A) tails significantly reduce recombination of plasmid DNA without affecting mRNA translation efficiency or half-life. RNA. 2019;25(4):507–18. doi:10.1261/rna.069286.118.
  • Xia X. Detailed dissection and critical evaluation of the Pfizer/BioNTech and moderna mRNA vaccines. Vaccines. 2021;9(7):734. doi:10.3390/vaccines9070734.
  • Li CY, Liang Z, Hu Y, Zhang H, Setiasabda KD, Li J, Ma S, Xia X, Kuang Y. Cytidine-containing tails robustly enhance and prolong protein production of synthetic mRNA in cell and in vivo. Mol Ther Nucleic Acids. 2022;30:300–10. doi:10.1016/j.omtn.2022.10.003.
  • Mostafa D, Takahashi A, Yanagiya A, Yamaguchi T, Abe T, Kureha T, Kuba K, Kanegae Y, Furuta Y, Yamamoto T, et al. Essential functions of the CNOT7/8 catalytic subunits of the CCR4-NOT complex in mRNA regulation and cell viability. RNA Biol. 2020;17(3):403–16. doi:10.1080/15476286.2019.1709747.
  • McKay PF, Hu K, Blakney AK, Samnuan K, Brown JC, Penn R, Zhou J, Bouton CR, Rogers P, Polra K, et al. Self-amplifying RNA SARS-CoV-2 lipid nanoparticle vaccine candidate induces high neutralizing antibody titers in mice. Nat Commun. 2020;11(1):3523. doi:10.1038/s41467-020-17409-9.
  • Pollock KM, Cheeseman HM, Szubert AJ, Libri V, Boffito M, Owen D, Bern H, O’Hara J, McFarlane LR, Lemm NM, et al. Safety and immunogenicity of a self-amplifying RNA vaccine against COVID-19: COVAC1, a phase I, dose-ranging trial. EClinicalMedicine. 2022;44:101262. doi:10.1016/j.eclinm.2021.101262.
  • Ramos da Silva J, Bitencourt Rodrigues K, Formoso Pelegrin G, Silva Sales N, Muramatsu H, de Oliveira Silva M, Porchia B, Moreno ACR, Aps L, Venceslau-Carvalho AA, et al. Single immunizations of self-amplifying or non-replicating mRNA-LNP vaccines control HPV-associated tumors in mice. Sci Transl Med. 2023;15(686):eabn3464. doi:10.1126/scitranslmed.abn3464.
  • Liu X, Zhang Y, Zhou S, Dain L, Mei L, Zhu G. Circular RNA: an emerging frontier in RNA therapeutic targets, RNA therapeutics, and mRNA vaccines. J Control Release. 2022;348:84–94. doi:10.1016/j.jconrel.2022.05.043.
  • Obi P, Chen YG. The design and synthesis of circular RNAs. Methods. 2021;196:85–103. doi:10.1016/j.ymeth.2021.02.020.
  • Qu L, Yi Z, Shen Y, Lin L, Chen F, Xu Y, Wu Z, Tang H, Zhang X, Tian F, et al. Circular RNA vaccines against SARS-CoV-2 and emerging variants. Cell. 2022;185(10):1728–44.e1716. doi:10.1016/j.cell.2022.03.044.
  • Li H, Peng K, Yang K, Ma W, Qi S, Yu X, He J, Lin X, Yu G. Circular RNA cancer vaccines drive immunity in hard-to-treat malignancies. Theranostics. 2022;12(14):6422–36. doi:10.7150/thno.77350.
  • Yang J, Zhu J, Sun J, Chen Y, Du Y, Tan Y, Wu L, Zhai M, Wei L, Li N, et al. Intratumoral delivered novel circular mRNA encoding cytokines for immune modulation and cancer therapy. Mol Ther Nucleic Acids. 2022;30:184–97. doi:10.1016/j.omtn.2022.09.010.
  • Amaya L, Grigoryan L, Li Z, Lee A, Wender PA, Pulendran B, Chang HY. Circular RNA vaccine induces potent T cell responses. Proc Natl Acad Sci USA. 2023;120(20):e2302191120. doi:10.1073/pnas.2302191120.
  • Huang D, Zhu X, Ye S, Zhang J, Liao J, Zhang N, Zeng X, Wang J, Yang B, Zhang Y, et al. Tumour circular RNAs elicit anti-tumour immunity by encoding cryptic peptides. Nature. 2023. doi:10.1038/s41586-023-06834-7.
  • Tockary TA, Abbasi S, Matsui-Masai M, Hayashi A, Yoshinaga N, Boonstra E, Wang Z, Fukushima S, Kataoka K, Uchida S. Comb-structured mRNA vaccine tethered with short double-stranded RNA adjuvants maximizes cellular immunity for cancer treatment. Proc Natl Acad Sci USA. 2023;120(29):e2214320120. doi:10.1073/pnas.2214320120.
  • Kranz LM, Diken M, Haas H, Kreiter S, Loquai C, Reuter KC, Meng M, Fritz D, Vascotto F, Hefesha H, et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature. 2016;534(7607):396–401. doi:10.1038/nature18300.
  • Tan Y, Lu S, Wang B, Duan X, Zhang Y, Peng X, Li H, Lin A, Zhan Z, Liu X. Single-cell transcriptome atlas reveals protective characteristics of COVID-19 mRNA vaccine. J Med Virol. 2023;95(1):e28161. doi:10.1002/jmv.28161.
  • Mohsen MO, Zha L, Cabral-Miranda G, Bachmann MF. Major findings and recent advances in virus-like particle (VLP)-based vaccines. Semin Immunol. 2017;34:123–32. doi:10.1016/j.smim.2017.08.014.
  • Ding Z, Li Q, Zhang R, Xie L, Shu Y, Gao S, Wang P, Su X, Qin Y, Wang Y, et al. Personalized neoantigen pulsed dendritic cell vaccine for advanced lung cancer. Sig Transduct Target Ther. 2021;6(1):26. doi:10.1038/s41392-020-00448-5.
  • Wang Z, Ma W, Fu X, Qi Y, Zhao Y, Zhang S. Development and applications of mRNA treatment based on lipid nanoparticles. Biotechnol Adv. 2023;65:108130. doi:10.1016/j.biotechadv.2023.108130.
  • Kiaie SH, Majidi Zolbanin N, Ahmadi A, Bagherifar R, Valizadeh H, Kashanchi F, Jafari R. Recent advances in mRNA-LNP therapeutics: immunological and pharmacological aspects. J Nanobiotechnol. 2022;20(1):276. doi:10.1186/s12951-022-01478-7.
  • Kauffman KJ, Webber MJ, Anderson DG. Materials for non-viral intracellular delivery of messenger RNA therapeutics. J Control Release. 2016;240:227–34. doi:10.1016/j.jconrel.2015.12.032.
  • Patel S, Ashwanikumar N, Robinson E, Xia Y, Mihai C, Griffith JP 3rd, Hou S, Esposito AA, Ketova T, Welsher K, et al. Naturally-occurring cholesterol analogues in lipid nanoparticles induce polymorphic shape and enhance intracellular delivery of mRNA. Nat Commun. 2020;11(1):983. doi:10.1038/s41467-020-14527-2.
  • Semple SC, Akinc A, Chen J, Sandhu AP, Mui BL, Cho CK, Sah DW, Stebbing D, Crosley EJ, Yaworski E, et al. Rational design of cationic lipids for siRNA delivery. Nat Biotechnol. 2010;28(2):172–6. doi:10.1038/nbt.1602.
  • Adams D, Gonzalez-Duarte A, O’Riordan WD, Yang CC, Ueda M, Kristen AV, Tournev I, Schmidt HH, Coelho T, Berk JL, et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med. 2018;379(1):11–21. doi:10.1056/NEJMoa1716153.
  • Zong Y, Lin Y, Wei T, Cheng Q. Lipid nanoparticle (LNP) enables mRNA delivery for cancer therapy. Adv Mater. 2023a;35(51):e2303261. doi:10.1002/adma.202303261.
  • Zong Y, Lin Y, Wei T, Cheng Q. Lipid nanoparticle (LNP) enables mRNA delivery for cancer therapy. Adv Mater. 2023b:e2303261. doi:10.1002/adma.202303261.
  • Kon E, Ad-El N, Hazan-Halevy I, Stotsky-Oterin L, Peer D. Targeting cancer with mRNA-lipid nanoparticles: key considerations and future prospects. Nat Rev Clin Oncol. 2023;20(11):739–54. doi:10.1038/s41571-023-00811-9.
  • Nakamura-Nishimura Y, Shinkuma S, Miyagawa F, Haredy A, Gomi Y, Yamanishi K, Asada H. Immunogenicity of varicella-zoster virus vaccine by different routes of administration: Comparable vaccination efficacy of one-fifth dose intradermal vaccination to conventional subcutaneous vaccination. J Dermatol Sci. 2022;106(2):86–92. doi:10.1016/j.jdermsci.2022.04.001.
  • Sahin U, Karikó K, Türeci Ö. mRNA-based therapeutics–developing a new class of drugs. Nat Rev Drug Discov. 2014;13(10):759–80. doi:10.1038/nrd4278.
  • Zhang H, You X, Wang X, Cui L, Wang Z, Xu F, Li M, Yang Z, Liu J, Huang P, et al. Delivery of mRNA vaccine with a lipid-like material potentiates antitumor efficacy through Toll-like receptor 4 signaling. Proc Natl Acad Sci USA. 2021;118(6). doi:10.1073/pnas.2005191118.
  • Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines - a new era in vaccinology. Nat Rev Drug Discov. 2018;17(4):261–79. doi:10.1038/nrd.2017.243.
  • Tenzer S, Docter D, Kuharev J, Musyanovych A, Fetz V, Hecht R, Schlenk F, Fischer D, Kiouptsi K, Reinhardt C, et al. Rapid formation of plasma protein corona critically affects nanoparticle pathophysiology. Nat Nanotechnol. 2013;8(10):772–81. doi:10.1038/nnano.2013.181.
  • Qiu M, Tang Y, Chen J, Muriph R, Ye Z, Huang C, Evans J, Henske EP, Xu Q. Lung-selective mRNA delivery of synthetic lipid nanoparticles for the treatment of pulmonary lymphangioleiomyomatosis. Proc Natl Acad Sci USA. 2022;119(8). doi:10.1073/pnas.2116271119.
  • Yang Z, Liu Y, Zhao K, Jing W, Gao L, Dong X, Wang Y, Han M, Shi C, Tang C, et al. Dual mRNA co-delivery for in situ generation of phagocytosis-enhanced CAR macrophages augments hepatocellular carcinoma immunotherapy. J Control Release. 2023;360:718–33. doi:10.1016/j.jconrel.2023.07.021.
  • Zhao X, Chen J, Qiu M, Li Y, Glass Z, Xu Q. Imidazole-based synthetic lipidoids for in vivo mRNA delivery into primary T lymphocytes. Angew Chem Int Ed Engl. 2020;59(45):20083–89. doi:10.1002/anie.202008082.
  • Chen J, Ye Z, Huang C, Qiu M, Song D, Li Y, Xu Q. Lipid nanoparticle-mediated lymph node-targeting delivery of mRNA cancer vaccine elicits robust CD8(+) T cell response. Proc Natl Acad Sci USA. 2022;119(34):e2207841119. doi:10.1073/pnas.2207841119.
  • Paunovska K, Sago CD, Monaco CM, Hudson WH, Castro MG, Rudoltz TG, Kalathoor S, Vanover DA, Santangelo PJ, Ahmed R, et al. A direct comparison of in vitro and in vivo nucleic acid delivery mediated by hundreds of nanoparticles reveals a weak correlation. Nano Lett. 2018;18(3):2148–57. doi:10.1021/acs.nanolett.8b00432.
  • Gustafson HH, Holt-Casper D, Grainger DW, Ghandehari H. Nanoparticle uptake: the phagocyte problem. Nano Today. 2015;10(4):487–510. doi:10.1016/j.nantod.2015.06.006.
  • Kowalski PS, Capasso Palmiero U, Huang Y, Rudra A, Langer R, Anderson DG. Ionizable amino-polyesters synthesized via ring opening polymerization of tertiary amino-alcohols for tissue selective mRNA delivery. Adv Mater. 2018;30(34):e1801151. doi:10.1002/adma.201801151.
  • Cheng Q, Wei T, Farbiak L, Johnson LT, Dilliard SA, Siegwart DJ. Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR-Cas gene editing. Nat Nanotechnol. 2020;15(4):313–20. doi:10.1038/s41565-020-0669-6.
  • Dilliard SA, Cheng Q, Siegwart DJ. On the mechanism of tissue-specific mRNA delivery by selective organ targeting nanoparticles. Proc Natl Acad Sci USA. 2021;118(52). doi:10.1073/pnas.2109256118.
  • Igyártó BZ, Jacobsen S, Ndeupen S. Future considerations for the mRNA-lipid nanoparticle vaccine platform. Curr Opin Virol. 2021;48:65–72. doi:10.1016/j.coviro.2021.03.008.
  • Martin D, Gutkind JS. Human tumor-associated viruses and new insights into the molecular mechanisms of cancer. Oncogene. 2008;Suppl 27(S2):S31–42. doi:10.1038/onc.2009.351.
  • Pietropaolo V, Prezioso C, Moens U. Role of virus-induced host cell epigenetic changes in cancer. Int J Mol Sci. 2021;22(15):8346. doi:10.3390/ijms22158346.
  • Saha A, Kaul R, Murakami M, Robertson ES. Tumor viruses and cancer biology: modulating signaling pathways for therapeutic intervention. Cancer Biol Ther. 2010;10(10):961–78. doi:10.4161/cbt.10.10.13923.
  • Cheng L, Wang Y, Du J. Human papillomavirus vaccines: an updated review. Vaccines. 2020;8(3):391. doi:10.3390/vaccines8030391.
  • Pattyn J, Hendrickx G, Vorsters A, Van Damme P. Hepatitis B vaccines. J Infect Dis. 2021;224(12 Suppl 2):S343–51. doi:10.1093/infdis/jiaa668.
  • Zhai L, Tumban E. Gardasil-9: a global survey of projected efficacy. Antiviral Res. 2016;130:101–09. doi:10.1016/j.antiviral.2016.03.016.
  • Lee S, Yoon H, Hong SH, Kwon SP, Hong JJ, Kwak HW, Park HJ, Yoo S, Bae SH, Park HJ, et al. mRNA-HPV vaccine encoding E6 and E7 improves therapeutic potential for HPV-mediated cancers via subcutaneous immunization. J Med Virol. 2023;95(12):e29309. doi:10.1002/jmv.29309.
  • Barouch DH, Whitney JB, Moldt B, Klein F, Oliveira TY, Liu J, Stephenson KE, Chang HW, Shekhar K, Gupta S, et al. Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys. Nature. 2013;503(7475):224–8. doi:10.1038/nature12744.
  • Zhang P, Narayanan E, Liu Q, Tsybovsky Y, Boswell K, Ding S, Hu Z, Follmann D, Lin Y, Miao H, et al. A multiclade env-gag VLP mRNA vaccine elicits tier-2 HIV-1-neutralizing antibodies and reduces the risk of heterologous SHIV infection in macaques. Nat Med. 2021;27(12):2234–45. doi:10.1038/s41591-021-01574-5.
  • Leggat DJ, Cohen KW, Willis JR, Fulp WJ, deCamp AC, Kalyuzhniy O, Cottrell CA, Menis S, Finak G, Ballweber-Fleming L, et al. Vaccination induces HIV broadly neutralizing antibody precursors in humans. Science 2022;378(6623):eadd6502. doi:10.1126/science.add6502.
  • Chen B, Chen Y, Li J, Wang C, Song W, Wen Y, Lin J, Wu Y, Ying T, Zheng C. A single dose of anti-HBsAg antibody-encoding mRNA-LNPs suppressed HBsAg expression: a potential cure of chronic hepatitis B virus infection. mBio. 2022;13(4):e0161222. doi:10.1128/mbio.01612-22.
  • Young LS, Yap LF, Murray PG. Epstein-Barr virus: more than 50 years old and still providing surprises. Nat Rev Cancer. 2016;16(12):789–802. doi:10.1038/nrc.2016.92.
  • Guo M, Duan X, Peng X, Jin Z, Huang H, Xiao W, Zheng Q, Deng Y, Fan N, Chen K, et al. A lipid-based LMP2-mRNA vaccine to treat nasopharyngeal carcinoma. Nano Res. 2023;16(4):5357–67. doi:10.1007/s12274-022-5254-x.
  • van Zyl DG, Mautner J, Delecluse HJ. Progress in EBV Vaccines. Front Oncol. 2019;9:104. doi:10.3389/fonc.2019.00104.
  • Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-van der Meer DM, Vloon AP, Essahsah F, Fathers LM, Offringa R, Drijfhout JW, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med. 2009;361(19):1838–47. doi:10.1056/NEJMoa0810097.
  • Shi Y, Zheng M. Hepatitis B virus persistence and reactivation. BMJ. 2020;370:m2200. doi:10.1136/bmj.m2200.
  • Zajac AJ, Blattman JN, Murali-Krishna K, Sourdive DJ, Suresh M, Altman JD, Ahmed R. Viral immune evasion due to persistence of activated T cells without effector function. J Exp Med. 1998;188(12):2205–13. doi:10.1084/jem.188.12.2205.
  • Buonaguro L, Tagliamonte M. Selecting target antigens for cancer vaccine development. Vaccines. 2020;8(4):615. doi:10.3390/vaccines8040615.
  • Gold P, Freedman SO. Demonstration of tumor-specific antigens in human colonic carcinomata by immunological tolerance and absorption techniques. J Exp Med. 1965;121(3):439–62. doi:10.1084/jem.121.3.439.
  • Ménard S, Pupa SM, Campiglio M, Tagliabue E. Biologic and therapeutic role of HER2 in cancer. Oncogene. 2003;22(42):6570–78. doi:10.1038/sj.onc.1206779.
  • Park JW, Hong K, Kirpotin DB, Meyer O, Papahadjopoulos D, Benz CC. Anti-HER2 immunoliposomes for targeted therapy of human tumors. Cancer Lett. 1997;118(2):153–60. doi:10.1016/s0304-3835(97)00326-1.
  • van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den Eynde B, Knuth A, Boon T. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science. 1991;254(5038):1643–47. doi:10.1126/science.1840703.
  • Gendler SJ, Lancaster CA, Taylor-Papadimitriou J, Duhig T, Peat N, Burchell J, Pemberton L, Lalani EN, Wilson D. Molecular cloning and expression of human tumor-associated polymorphic epithelial mucin. J Biol Chem. 1990;265(25):15286–93. doi:10.1016/S0021-9258(18)77254-2.
  • Wang MC, Valenzuela LA, Murphy GP, Chu TM. Purification of a human prostate specific antigen. J Urol. 2017;197(2s):S148–52. doi:10.1016/j.juro.2016.10.100.
  • Chen YT, Scanlan MJ, Sahin U, Türeci O, Gure AO, Tsang S, Williamson B, Stockert E, Pfreundschuh M, Old LJ. A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci USA. 1997;94(5):1914–18. doi:10.1073/pnas.94.5.1914.
  • Oji Y, Ogawa H, Tamaki H, Oka Y, Tsuboi A, Kim EH, Soma T, Tatekawa T, Kawakami M, Asada M, et al. Expression of the Wilms’ tumor gene WT1 in solid tumors and its involvement in tumor cell growth. Jpn J Cancer Res. 1999;90(2):194–204. doi:10.1111/j.1349-7006.1999.tb00733.x.
  • Crandall BF, Lau HL. Alpha-fetoprotein: a review. Crit Rev Clin Lab Sci. 1981;15(2):127–85. doi:10.3109/10408368109105870.
  • Nakatsura T, Yoshitake Y, Senju S, Monji M, Komori H, Motomura Y, Hosaka S, Beppu T, Ishiko T, Kamohara H, et al. Glypican-3, overexpressed specifically in human hepatocellular carcinoma, is a novel tumor marker. Biochem Biophys Res Commun. 2003;306(1):16–25. doi:10.1016/s0006-291x(03)00908-2.
  • Ribas A, Butterfield LH, McBride WH, Jilani SM, Bui LA, Vollmer CM, Lau R, Dissette VB, Hu B, Chen AY, et al. Genetic immunization for the melanoma antigen MART-1/Melan-A using recombinant adenovirus-transduced murine dendritic cells. Cancer Res. 1997;57:2865–9.
  • Hollingsworth RE, Jansen K. Turning the corner on therapeutic cancer vaccines. NPJ Vaccines. 2019;4(1):7. doi:10.1038/s41541-019-0103-y.
  • He Q, Gao H, Tan D, Zhang H, Wang JZ. mRNA cancer vaccines: advances, trends and challenges. Acta Pharm Sin B. 2022;12(7):2969–89. doi:10.1016/j.apsb.2022.03.011.
  • Gubin MM, Zhang X, Schuster H, Caron E, Ward JP, Noguchi T, Ivanova Y, Hundal J, Arthur CD, Krebber WJ, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature. 2014;515(7528):577–81. doi:10.1038/nature13988.
  • Matsushita H, Vesely MD, Koboldt DC, Rickert CG, Uppaluri R, Magrini VJ, Arthur CD, White JM, Chen YS, Shea LK, et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature. 2012;482(7385):400–404. doi:10.1038/nature10755.
  • Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, Lin JC, Teer JK, Cliften P, Tycksen E, et al. Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med. 2013;19(6):747–52. doi:10.1038/nm.3161.
  • Sahin U, Oehm P, Derhovanessian E, Jabulowsky RA, Vormehr M, Gold M, Maurus D, Schwarck-Kokarakis D, Kuhn AN, Omokoko T, et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature. 2020;585(7823):107–12. doi:10.1038/s41586-020-2537-9.
  • Tran E, Turcotte S, Gros A, Robbins PF, Lu YC, Dudley ME, Wunderlich JR, Somerville RP, Hogan K, Hinrichs CS, et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science. 2014;344(6184):641–45. doi:10.1126/science.1251102.
  • Tran E, Ahmadzadeh M, Lu YC, Gros A, Turcotte S, Robbins PF, Gartner JJ, Zheng Z, Li YF, Ray S, et al. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science. 2015;350(6266):1387–90. doi:10.1126/science.aad1253.
  • Bassani-Sternberg M, Chong C, Guillaume P, Solleder M, Pak H, Gannon PO, Kandalaft LE, Coukos G, Gfeller D, Hertz T. Deciphering HLA-I motifs across HLA peptidomes improves neo-antigen predictions and identifies allostery regulating HLA specificity. PLoS Comput Biol. 2017;13(8):e1005725. doi:10.1371/journal.pcbi.1005725.
  • Kim S, Kim HS, Kim E, Lee MG, Shin EC, Paik S, Kim S. Neopepsee: accurate genome-level prediction of neoantigens by harnessing sequence and amino acid immunogenicity information. Ann Oncol. 2018;29(4):1030–6. doi:10.1093/annonc/mdy022.
  • Segal NH, Parsons DW, Peggs KS, Velculescu V, Kinzler KW, Vogelstein B, Allison JP. Epitope landscape in breast and colorectal cancer. Cancer Res. 2008;68(3):889–92. doi:10.1158/0008-5472.Can-07-3095.
  • Xu H, Zheng X, Zhang S, Yi X, Zhang T, Wei Q, Li H, Ai J. Tumor antigens and immune subtypes guided mRNA vaccine development for kidney renal clear cell carcinoma. Mol Cancer. 2021;20(1):159. doi:10.1186/s12943-021-01465-w.
  • Sahin U, Derhovanessian E, Miller M, Kloke BP, Simon P, Löwer M, Bukur V, Tadmor AD, Luxemburger U, Schrörs B, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547(7662):222–6. doi:10.1038/nature23003.
  • mRNA Vaccine Slows Melanoma Recurrence. Cancer Discov. 2023;13(6):1278. doi:10.1158/2159-8290.Cd-nb2023-0028.
  • Hsu SK, Jadhao M, Liao WT, Chang WT, Hung CT, Chiu CC. Culprits of PDAC resistance to gemcitabine and immune checkpoint inhibitor: tumour microenvironment components. Front Mol Biosci. 2022;9:1020888. doi:10.3389/fmolb.2022.1020888.
  • Rojas LA, Sethna Z, Soares KC, Olcese C, Pang N, Patterson E, Lihm J, Ceglia N, Guasp P, Chu A, et al. Personalized RNA neoantigen vaccines stimulate T cells in pancreatic cancer. Nature. 2023;618(7963):144–50. doi:10.1038/s41586-023-06063-y.
  • Xiao Y, Chen J, Zhou H, Zeng X, Ruan Z, Pu Z, Jiang X, Matsui A, Zhu L, Amoozgar Z, et al. Combining p53 mRNA nanotherapy with immune checkpoint blockade reprograms the immune microenvironment for effective cancer therapy. Nat Commun. 2022;13(1):758. doi:10.1038/s41467-022-28279-8.
  • Van Lint S, Renmans D, Broos K, Goethals L, Maenhout S, Benteyn D, Goyvaerts C, Du Four S, Van der Jeught K, Bialkowski L, et al. Intratumoral delivery of TriMix mRNA results in T-cell activation by cross-presenting dendritic cells. Cancer Immunol Res. 2016;4(2):146–56. doi:10.1158/2326-6066.Cir-15-0163.