Skip to main content
Erschienen in: Journal of Translational Medicine 1/2023

Open Access 01.12.2023 | Review

Advancing personalized medicine in brain cancer: exploring the role of mRNA vaccines

verfasst von: Feng Lin, Emma Z. Lin, Misa Anekoji, Thomas E. Ichim, Joyce Hu, Francesco M. Marincola, Lawrence D. Jones, Santosh Kesari, Shashaanka Ashili

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2023

Abstract

Advancing personalized medicine in brain cancer relies on innovative strategies, with mRNA vaccines emerging as a promising avenue. While the initial use of mRNA vaccines was in oncology, their stunning success in COVID-19 resulted in widespread attention, both positive and negative. Regardless of politically biased opinions, which relate more to the antigenic source than form of delivery, we feel it is important to objectively review this modality as relates to brain cancer. This class of vaccines trigger robust immune responses through MHC-I and MHC-II pathways, in both prophylactic and therapeutic settings. The mRNA platform offers advantages of rapid development, high potency, cost-effectiveness, and safety. This review provides an overview of mRNA vaccine delivery technologies, tumor antigen identification, combination therapies, and recent therapeutic outcomes, with a particular focus on brain cancer. Combinatorial approaches are vital to maximizing mRNA cancer vaccine efficacy, with ongoing clinical trials exploring combinations with adjuvants and checkpoint inhibitors and even adoptive cell therapy. Efficient delivery, neoantigen identification, preclinical studies, and clinical trial results are highlighted, underscoring mRNA vaccines' potential in advancing personalized medicine for brain cancer. Synergistic combinatorial therapies play a crucial role, emphasizing the need for continued research and collaboration in this area.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Personalized medicine aims to revolutionize healthcare by providing tailored treatments based on an individual’s unique characteristics. Genetic information of the host and target plays a crucial role in determining disease susceptibility and treatment response [1, 2]. By utilizing genomic analysis, biomarker identification, risk assessment, tailored treatment strategies, and continuous monitoring, personalized medicine seeks to improve patient outcomes while minimizing adverse effects and reducing healthcare costs. In the field of brain cancer, personalized medicine holds great promise due to the complexity and heterogeneity of tumors, as well as individual variations in treatment response.
Brain cancer poses a significant threat to health, with approximately 1% of annual cancer diagnoses in the United States attributed to this condition. The rapid growth and invasion of malignant tumors in the brain or nearby structures can disrupt vital brain functions. Treatment options for brain cancer, including surgery, radiation therapy, and chemotherapy, vary in effectiveness depending on tumor type, location, and stage. However, challenges such as the difficulty of accessing and removing tumors in critical brain regions without damaging healthy tissue [3], and the presence of blood–brain barrier (BBB) limit the efficacy of current treatment approaches. Furthermore, brain tumors often exhibit heterogeneity, with different cell populations having varying sensitivities to therapies. These factors necessitate personalized treatment strategies tailored to each patient’s unique circumstances.
Immunotherapy has emerged as a promising approach for brain cancer treatment, harnessing the body's innate and adaptive defenses to seek and destroy transformed cells. Immune checkpoint inhibitors, such as pembrolizumab and nivolumab, have shown some success in treating certain types of brain cancer [4, 5] by blocking proteins that inhibit immune responses, thereby enhancing the ability of immune cells to recognize and attack cancer cells. Other immunotherapeutic approaches, including tumor and biomarker-based vaccines [6], monoclonal antibody drugs [7], and chimeric antigen receptor (CAR) T cell therapies are also being investigated [8, 9]. These immunotherapies aim to directly target cancer biomarkers, induce tumor regression, and modulate the suppressive tumor microenvironment, all with the goal of improving overall survival rates. However, several challenges hinder the effectiveness of immunotherapy for brain cancer. The presence of the BBB restricts immune cell access to the tumor site, potentially diminishing treatment efficacy [10]. Additionally, the immunosuppressive brain microenvironment and tumor heterogeneity further complicate the activation of immune response [11]. A deeper understanding of the intricate interactions between the immune system and brain cancer cells is crucial for the development of more effective and personalized treatment options.
Cancer vaccines, which educate the immune system about cancer cells to enable their recognition and elimination, represent an attractive form of immunotherapy. Prophylactic cancer vaccines against hepatitis B (HBV)-related liver cancer and human papilloma virus (HPV)-related cervical cancer and head and neck cancer have already been approved by the U.S. FDA [12]. Therapeutic cancer vaccine have been extensively studied in both preclinical and clinical settings, with Bacillus Calmette-Guerin (BCG) and Sipuleucel-T (Provenge) being antigen nonspecific and antigen specific FDA-approved examples. Recent studies, including a phase 3 clinical trial of a dendritic cell vaccine have reported success at prolonging survival in glioblastoma (GBM) patients [13]. Chen et al. developed a novel cancer cell-based therapy by modifying tumor cells to release immunomodulatory agents and induce cancer cell apoptosis. The engineered therapeutic tumor cells (ThTCs) effectively eliminated glioblastoma tumors and promoted antitumor immune responses in mice, offering a potential immunotherapy for glioblastoma tumor and other solid tumors [14]. Other type of vaccines such as personalized or shared neoantigen peptide vaccines for brain cancer have been obtaining promising results [15]. Advances in single-cell and RNA sequencing technologies have facilitated the identification of cancer biomarkers, allowing for the development of personalized neoantigen vaccines for brain cancer. These vaccines target specific neoantigens recognized by T cells, offering a highly immunogenic approach for personalized vaccination [16]. Overall, targeting either tumor cells or benign cells within the cancer cellular network should aim to enhance, either directly or indirectly, immunogenic cell death. This is likely the primary driver that transforms a poorly immunogenic tumor into an immune active, immune infiltrated phenotype that is more responsive to immunotherapy [17, 18].
To induce specific immune responses again tumor cells, therapeutic vaccines must activate broad immune responses, particularly specific cytotoxic T cell response. Genetic vaccines, including DNA and RNA vaccines, have the ability to prime both T and B cell responses, activating cellular and humoral immune responses and inducing an adaptive immune response to the encoded target antigen [19]. An example of this are studies demonstrating DNA-based therapeutic vaccines targeting HPV exhibit impressive efficacy in phase II clinical trials and are progressing to phase III [20, 21]. Compared to DNA vaccines, mRNA vaccines are believed to have a better safety profile and a more adaptable production process [22, 23]. The recent approvals of mRNA vaccines for COVID-19 have further highlighted their potential of mRNA vaccine. Notably, COVID-19 mRNA vaccination induced significant antibody [24, 25] and memory B cell responses against full-length SARS-CoV2 spike protein and the spike receptor binding domain (RBD) [25, 26], along with robust T cell response [27, 28]. Encouraging results from preclinical studies and clinical trials with mRNA cancer vaccines and mRNA infectious disease vaccines have inspired biotechnological and pharmaceutical industries to focus on this promising area of research [29, 30].
This review aims to explore the factors, combinations, and optimizations necessary to maximize the efficacy of mRNA cancer vaccines and delivery systems. It will also discuss the clinical applications of mRNA vaccines in advancing personalized medicine for brain cancer. Additionally, the review will consider future considerations for transitioning mRNA vaccines from the laboratory to the clinic. By shedding light on the role of mRNA vaccines in brain cancer treatment, we aim to contribute to the growing body of knowledge and provide valuable insight into the potential of mRNA-based immunotherapies for improving brain cancer treatment outcomes.

mRNA vaccine delivery technology, route, and adjuvant exploration

mRNA is intrinsically not stable, but over the past decades, several technological innovations have enabled mRNA to be a more favorable vaccine platform. Developments in modification of mRNA backbone and untranslated regions by codon optimization as well as innovative selection of internal ribosome entry sites have resulted in creation of molecules with increased stability and RNase resistance, as well as augmented translation efficiency [31, 32]. Introduction of efficient non-viral in vivo delivery of mRNA has been achieved by formulating mRNA into lipid nanoparticles (LNPs) using peptides, cationic nano-emulsions, lipids, and polymers [31, 33, 34], and the technology has been reviewed by Hou et al. and others [3538]. Alternatively, viral like particles may provide the specificity that maybe lacking with LNPs [39, 40].
Ongoing improvements and optimizations to the technology are underway. Given that the dendritic cell (DC) resides at the center of the immunological universe, being the only cell capable of activating naïve T cells, one unique approach involves specifically targeting mRNA to these cells. Zhang et al. developed a minimalist nanovaccine with C1 LNPs that could efficiently deliver mRNA into dendritic cells with simultaneous Toll-like receptor 4 (TLR4) activation and induced robust T cell activation [41]. In a different variation on the theme, Verbeke et al. [42] developed mRNA Galsomes, a nanoparticle platform co-delivering mRNA, glycolipid antigen, and α-GC to antigen-presenting cells after intravenous administration. In mice, mRNA Galsomes triggered a robust tumor-specific immune response. Tumor growth decreased, achieving complete rejection in 40% of animals. Combining mRNA Galsomes with a PD-L1 checkpoint inhibitor reduced tumor outgrowth and increased survival, highlighting their potential in cancer immunotherapy. Fornaguera et al. created poly(beta aminoesters) polymer nanoparticles as efficient mRNA delivery systems, with a formulation protocol for disease prevention and cure using mRNA vaccines [43]. Fan YN et al. investigated cationic lipid-assisted nanoparticles (CLAN) for mRNA vaccine delivery. CLAN effectively stimulated dendritic cell maturation and activated antigen-specific T cells in vitro and in vivo. Intravenous immunization with CLAN containing mRNA encoding ovalbumin resulted in a robust OVA-specific T-cell response and inhibited tumor growth in an E·G7-OVA lymphoma model [44]. Similarly, Mai et al. [45] used positively charged protamine to concentrate mRNA and form a stable polycation-mRNA complex. Encapsulating this complex with cationic liposomes (LPC) resulted in enhanced dendritic cell maturation and potent anti-tumor immune response in vitro and in vivo.
These targeted delivery systems have been utilized via alternative rounds of administration for example, intranasal immunization of mice with cationic LPC containing mRNA encoding cytokeratin 19 resulted in a robust cellular immune response and inhibited tumor growth in a Lewis lung cancer model. Persano et al. developed a lipopolyplex mRNA vaccine comprising a poly-(β-amino ester) polymer mRNA core enclosed within an EDOPC/DOPE/DSPE-PEG lipid shell. This vaccine enters dendritic cells, stimulates interferon-β and interleukin-12 expression through Toll-like receptor 7/8 signaling. In a mouse tumor model with B16-OVA lung metastatic tumors, treatment with the lipopolyplex mRNA led to over 90% reduction in tumor nodules [46]. Haabeth et al. developed charge-altering releasable transporters (CARTs) for efficient mRNA vaccine delivery. CARTs target antigen presenting cells (APCs) in lymphoid organs via intravenous injection and local APCs via subcutaneous injection in a mouse model. Co-formulating CARTs with mRNA and Toll-like receptor ligands enables transfection and activation of target cells, leading to an effective immune response capable of treating and curing mice with large, established tumors [47]. Overall, these studies appear to be a “second generation” in the development of mRNA therapeutics in that they are targeting specific immune cells. Further understanding of dendritic cell subsets will allow for additional improvements in efficacy of this approach.
Dendritic cells naturally respond to recognition of immunological adjuvants by enhancement of antigen presenting activity. Accordingly, administration of various adjuvants may increase efficacy of mRNA activity through increasing dendritic cell number and activity. Hess et al. found genetic adjuvant can boost immunogenicity of mRNA vaccine. Co-delivering GM-CSF mRNA with tumor-associated antigen mRNA significantly enhances the cytotoxic T cell response in animal tumor models [48]. Li et al. developed CpG2018B, activating immune responses through TLR9. CpG2018B and an mRNA-based neoantigen cancer vaccine were encapsulated in lipid nanoparticles (LNPs) and intratumorally administered to melanoma mouse models. Combing CpG and the mRNA vaccine enhanced the antitumor effect [49]. A novel vaccine design exploits its dual role in adaptive and innate immunity. Combing free and protamine-complexed mRNA, it induces balanced adaptive immune responses with antigen-specific CD4 + and CD8 + T cells, as well as sustains memory responses. In a mouse tumor model, the two-component mRNA vaccine demonstrates robust antitumor activity against OVA-expressing tumor cells, serving as both preventive and therapeutic intervention [50].
Leveraging of various nanotechnologies has also been used to augment mRNA immune stimulatory activity. For example, a novel adjuvant-pulsed mRNA vaccine nanoparticle has been developed, containing ovalbumin-coded mRNA and a palmitic acid-modified TLR7/8 agonist (C16-R848). The lipid-PEG-coated NP with C16-R848 adjuvant significantly enhances OVA-specific CD8 + T cells expansion and infiltration into the tumor sites. In mice models, this approach effectively induces anti-tumor immunity against OVA-expressing lymphoma and prostate cancer. Administered before tumor engraftment, the vaccine prevents tumor growth by 84% compared to the control, while post-engraftment administration results in a 60% reduction in tumor growth [51].Van Lint et al. formulated a TriMix mRNA adjuvant vaccine that encodes CD70, CD40 ligand, and a constitutively active Toll-like receptor 4. Intra-tumoral delivery stimulated antitumor T-cell responses in animal models, resulting in systemic therapeutic antitumor immunity [52]. Bialkowski et al. demonstrated that intra-lymphatic E7-TriMix mRNA vaccine induced CD8 ( +) T lymphocyte migration into the tumor and controlled tumor growth effectively. Combining mRNA immunization with cisplatin treatment led to complete regression of clinically relevant tumors [53].
The recent advancements have elevated the field of mRNA therapeutics from just a novel method of antigen delivery to a practical treatment modality with practical ability to modulate immunity to tumors which conventionally have been seen as poorly immunogenic. With the tools now available to break antigen-specific tolerance found in cancers, the selection of appropriate antigens has become paramount.

mRNA vaccine for cancer

mRNA is a single-stranded macromolecule transcribed from DNA in the cell nuclei. Once transcribed, this endogenous mRNA is read by ribosomes to translate the genetic information into proteins in the cytoplasm [54]. Leveraging this natural process, scientists have synthesized mRNA for vaccines. The mRNA vaccine has been utilized in cancer treatment to prompt cells to produce cancer-specific proteins/neoantigens [55]. These neoantigens can induce an immune response against cancer cells, making mRNA vaccine a promising approach in cancer immunotherapy. It has rapidly become recognized that mRNA vaccines possess multiple advantages compared to traditional approaches [5557]. Firstly, mRNA vaccines can be customized to target specific tumor antigens or neoantigens unique to each patient's cancer cells through a process that involves tumor removal, sequencing to identify these unique antigens, and then developing a personalized vaccine. While promising, this approach can be prolonged and costly, with biotech estimates placing the cost per patient at around 350,000 USD. Alternatively, there are mRNA strategies that prompt immunogenic cell death, directly releasing tumor antigens from the cancer cells. These methods not only expedite the process but also allow for the creation of tailored vaccines as a subsequent step after tumor extraction. The personalized nature of mRNA vaccines facilitates a precisely target immune response against cancer cells, ensuring minimal impact on healthy cells. While the development and manufacturing of these vaccines are swift, enabling rapid adjustment to ever-changing tumor traits and mutations, there’s a caveat: tumor sequencing and epitope definition for each patient might introduce delays. Still, the versatility of mRNA vaccine is evident in their compatibility with other cancer therapies. When paired with treatments like chemotherapy or checkpoint inhibitors, they can significantly amplify the efficacy. This synergy with established treatments could enhance overall therapeutic outcomes. Moreover, by eliciting both cellular and humoral immune defenses, mRNA vaccines may offer a robust, long-term shield against cancer cells, potentially diminishing the odds of a recurrence.
The initial clinical utilization of mRNA vaccines was in oncology in which numerous clinical trials have been conducted to evaluate the safety and efficacy of mRNA vaccines in various types of cancer [58, 59]. Early clinical results have shown promising outcomes, with evidence of immune responses against tumor cells and some instances of tumor shrinkage. Findings from the phase IIb KEYNOTE-942 trial indicate that a personalized mRNA cancer vaccine, mRNA-4157/V940, combined with pembrolizumab, improved recurrence-free survival in high-risk melanoma patients regardless of tumor mutational burden. The vaccine encodes patient-specific tumor neoantigens, stimulating an immune response [59]. The approach shows promise for melanoma and potentially other cancers such as pancreatic cancer [60].
Research efforts are also focused on refining delivery methods, stability, and immunogenicity specifically for cancer applications. As mRNA vaccine technology advances and more data is gathered from clinical trials, it is expected that the potential of mRNA vaccines for cancer treatment will continue to be explored and optimized. The ability to customize vaccines based on the individual genetic profile of each patient's tumor offers a promising avenue for personalized cancer therapy. However, further research and larger-scale clinical studies are required to determine the full potential and integration of mRNA vaccines into standard cancer treatment protocols. Several challenges need to be addressed through ongoing research and development. These challenges include optimizing delivery systems, stabilizing mRNA molecules, and modulating immunogenicity to maximize the potential of mRNA vaccines in cancer immunotherapy.

mRNA cancer vaccine clinical trials

Clinical trials exploring mRNA cancer vaccines are underway, transforming the standard of case in oncology treatment. These trials leverage mRNA technology to activate the immune system against cancer cells, offering personalized medicine for improved outcomes. With diverse vaccine options targeting different malignancies, researchers are diligently evaluating their efficacy and safety. These groundbreaking trials hold immense potential to transform the landscape of cancer therapy, offering hope for cancer patients. The related trials both completed and ongoing have been listed in Table 1.
Table 1
mRNA vaccine clinical trials for cancer
mRNA Vaccine
Adjuvant/intervention
Indication
Phase
Trial number
Sponsor
Reference
Personalized mRNA vaccine (Autogene Cevumeran)
Atezolizumab
Advanced tumors including pancreatic cancer
I
NCT03289962
Genentech, (collaborator: BioNTech)
[60]
Atezolizumab, mFOLFIRINOX
Pancreatic cancer
II
NCT05968326
 
Personalized mRNA vaccine
N/A
Esophageal cancer, NSCLC
I
NCT03908671
Stemirna Therapeutics
 
Sintilimab
Advanced solid tumor
I
NCT05949775
 
Personalzed mRNA vaccine SW1115C3
N/A
Solid tumor
I
NCT05198752
 
Personalized mRNA vaccine
N/A
Esophageal, gastric, pancreatic, and colon cancers
I
NCT03468244
Changhai Hospital, Shanghai
 
Personalized mRNA vaccine
Sintilimab
Liver cancer
I
NCT05761717
Shanghai Zhongshan Hospital
 
Personalized neoantigen mRNA vaccine
N/A
Solid tumor
I
NCT05359354
Peking Union Medical College Hospital
 
EBV mRNA vaccine
N/A
EBV-positive advanced malignant tumors
I
NCT05714748
West China Hospital
 
HBV mRNA vaccine
N/A
HBV-positive liver cancer
I
NCT05738447
Peking Union Medical College Hospital
 
BNT113 (HPV16 E6/E7 mRNA- LPX vaccine)
Pembrolizumab
HPV16 + _head and neck cancer, PDL1 postive
II
NCT04534205
BioNTech SE
 
Neoantigen mRNA vaccine ABOR2014(IPM511)
NA
Liver cancer
I
NCT05981066
Peking Union Medical College Hospital
 
mRNA neoantigen tumor vaccine
Combination with PD-1/L1
Gastric cancer, esophageal cancer, liver cancer
I
NCT05192460
NeoCure
 
DCs loaded with mRNA from tumor tissue, hTERT and Survivin
N/A
Prostate cancer
I/II
NCT01197625
Oslo university hospital
 
CV9103 mRNA
 
Prostate cancer
I/II
NCT00831467
CureVac
[65]
BNT112 containing mRNA targeting 5 antigens expressed in prostate cancder (RNA-LPX)
Cemiplimab
Prostate cancer
I
NCT04382898
BioNTech SE
 
Tumor mRNA-lipid particles(LPs)
N/A
Lung cancer
I
NCT05660408
University of Florida
 
mRNA vaccine BI1361849 (CV9202)
Anti-PDL1 Durvalumab
Anti-CTLA3 Tremelimumab
NSCLC
Results posted on Clinicaltrials.gov
I/II
NCT03164772
Ludwig Institute for cancer research
 
V941(mRNA-5671/V941)
Pembrolizumab
NSCLC, colorectal cancer or pancreatic cancer
I/II
NCT03948763
Completed
Merck Sharp & Dohme LLC
 
mRNA-4157
Pembrolizumab
Solid tumor
I
NCT03313778
ModernaTX, Inc
 
WT1 mRNA loaded DCs
Combined with Chemotherapy
Malignant pleural mesothelioma
I/II
NCT02649829
University Hospital, Antwerp
 
TAA-loaded DCs
N/A
Colorectal cancer
I/II
NCT01885702
Radboud University
 
CEA mRNA and peptide loaded DCs (electroporated)
 
Colorectal cancer
I
NCT00228189
Radbound Universtity Medical Cener
[71]
Personalized primary tumor mRNA loaded DOTAP liposome vaccine
Anti-PD1
Melanoma
I
NCT05264974
University of Florida
 
mRNA-4157
Pembrolizumab
Melanoma
II
NCT03897881
Moderna TX, Inc
[74]
DCs-loaded with TAA mRNA
N/A
Melanoma
I
NCT01456104
Memorial Sloan Kettering Cancer Center
 
TriMix-DC (mRNA-loaded DCs)
 
Melanoma
I
NCT01066390
Universitair Ziekenhuis Brussel
[66, 67]
Tumor mRNA transfected DCs
IL-2
Melanoma
I/II
NCT01278940
Olso University Hospital
[70]
mRNA encoding Melan-A, MAGE-A1, MAGE-A3, Survivin, GP100 and Tyrosinase
GM-CSF s.c
Melanoma
I/II
NCT00204607
University Hospital Tuebingen
[63]
NCI-4650 mRNA vaccine (personalized)
 
Melanoma or epithelial cancer
I/II
NCT03480152
NCI
[16]
CT7, MAGE-A3 and WT1 mRNA-electroporated Langerhans cells (LCs)
 
Multiple myeloma
I
NCT01995708
Memorial Sloan Kettering Cancer Center
[73]
WT1 mRNA-DCs
N/A
AML
II
NCT01686334
Zwi Berneman
 
WT1 mRNA -DCs
N/A
Myelodysplastic syndromes, AML
I/II
NCT03083054
University of Campinas, Brazil
 
WT1 mRNA-DCs
 
AML
I/II
NCT00834002
University Hospital, Antwerp
[72]
GRNVAC1 mRNA encoding hTERT-LAMP1 -DCs
 
AML
II
NCT00510133
Asterias Biotherapeutics, Inc
 
mRNA plus lysate-loaded DCs
Pegylated-interferon alpha-2A and Filgrastim
Cutaneous Angiosarcoma
I
NCT05799612
MD Anderson Cancer Center
 
CMV-DC (CMV pp65-LAMP mRNA-loaded DC)
Tetanus toxoid
GBM
I/II
NCT00639639
Duke University
 
DCs transfected with mRNA from tumor stem cells, surviving and hTERT
Adjuvant temozolomide
GBM
II/III
NCT03548571
Oslo University Hospital
 
CMV pp65-LAMP mRNA-pulsed autologous DC vaccine
N/A
GBM
I
NCT02366728
Duke University
 
WT1 mRNA-loaded DC vaccine
Temozolomide
Glioma
I/II
NCT04911621
University Hospital, Antwerp
 
Personalized primary tumor mRNA and full length of pp65-LAMP (RNA-LPs)
N/A
pHGG and GBM
I
NCT04573140
University of Florida
 
CV09050101 mRNA
N/A
GBM or astrocytoma
I
NCT05938387
CureVac
 
CMV-pp65-LAMP mRNA- loaded DCs
Varillumab and Temozolomide
GBM
II
NCT03688178
Duke University
 
CMV pp65-LAMP mRNA-loaded DCs
CMV-ALT, GM-CSF
GBM
I/II
NCT00639639
Duke University
[76, 77]
CMV mRNA DCs
Nivolumab
GBM
I/II
NCT02529072
Duke University
 
pp65-shLAMP mRNA-DCs
Td, and GM-CSF
GBM
II
NCT02465268
Immunomic Therapeutics, Inc
 
WT1 mRNA-loaded DCs
Temozolomide
GBM
I/II
NCT02649582
Unviersity Hospital, Antwerp
 
Tumor stem cell derived mRNA-transfected DCs
Combined with standard treatment
GBM
I/II
NCT00846456
Oslo University Hospital
[75]
Personalized mRNA-DCs
 
GBM
I/II
NCT02709616
Guangdong 999 Brain Hospital
[78]
The first mRNA vaccine clinical trial for cancer was conducted in renal cell carcinoma (RCC). The trial aimed to induce T-cell responses against multiple tumor-associated antigens (TAAs) to improve overall survival. Conducted between 2003 and 2005, investigators administered an intradermal mRNA-based vaccine encoding six TAAs (MUC1, CEA, Her2/neu, telomerase, survivin, and MAGE-A1) to 30 metastatic RCC patients. Results revealed that long-term survivors exhibited immunological responses to the antigens, establishing a clear link between survival and immune responses to TAAs encoded by the naked mRNA vaccine [61, 62]. In a phase I/II trial, Weide et al. [63] admonished protamine-stabilized mRNAs coding for Melan-A, Tyrosinase, gp100, Mage-A1, Mage-A3, and Survivin to 21 metastatic melanoma patients. Ten patients received keyhole limpet hemocyanin (KLH) with the vaccine, and GM-CSF was used as an adjuvant. KLH patients experienced decreased Foxp3 + /CD4 + regulatory T cells, while non-KLH patients had reduced myeloid suppressor cells (CD11b + HLA-DR lo monocytes). Two of four evaluated patients showed increased vaccine specific T cells. One of 7 patients with measurable disease showed a complete response.
Several similar studies were reviewed by Bidram et al. [64], highlighting the associated challenges. They emphasized the potential of combining mRNA vaccines with immune-checkpoint blockade to enhance cell-mediated immunity. Viability of this approach was observed in other tumors, for example, the mRNA vaccines CV9103 and CV9104 were developed for prostate cancer treatment [65]. They utilize RNActive® formulation by CureVac (Tubingen, Germany), enabling optimized translational activity and immune stimulation. CV9103, the precursor to CV9104, targets four prostate-specific antigens: PSA, PSMA, PSCA, and STEAP. Phase I and II trials of CV9103 demonstrated safe administration, high immunogenicity rates, and favorable clinical outcomes, including prolonged stabilization of PSA levels. CV9104, with additional mRNA molecules encoding PAP and MUC1, showed antigen-specific immune responses in 5 out of 6 patients, eliciting immune changes in CD4 + and CD8 + T cells [65].
Utilizing mRNA in DC-based cancer vaccines shows promise due to DCs role in linking innate and adaptive immune responses. Van Nuffel et al. loaded the mRNAs of gp100, MAGE-A3, and MAGE- C2 into the DCs via the pST1-SIINFEKL vector, resulting in significant levels of functional CD4 + and CD8 + cells in melanoma patients [66, 67]. Since the peptides from loaded mRNAs must be coupled with the MHC compound to mount the anti-tumoral immune responses, the chimeric lysosome-associated membrane protein-1 (LAMP-1) with the tumoral antigen can facilitate this process [68]. Similarly, Su et al. have shown that the mRNA of tumoral antigen/LAMP-1 can enhance the peptide/MHC-I presentation and the anti-tumoral immune responses in affected patients [69]. However, Kyte et al. [70] vaccinated melanoma patients with autologous tumor-mRNA loaded DCs in a phase I/II trial. The study showed the DC-vaccine was safe, feasible, and induced in vivo T-cell response against transfected tumor-mRNA antigens. Lesterhuis et al. [71] compared CEA mRNA-DC with CEA peptide loaded DC vaccine in 16 colorectal cancer patients. CEA peptide-specific T cells were detected in 8 of 11 patients in the peptide group, but none in the mRNA group, possibly due to different antigen processing pathways or T-cell repertoires. Van de Velde et al. [72] reported a non-randomized single-center study for acute myeloid leukemia in 50 patients. The study demonstrated better survival in patients treated with allogeneic hematopoietic stem cell transplantation (HCT) and Wilms’ tumor protein (WT1) mRNA transfected DCs vaccine compared to control groups. Chung et al. [73] conducted a phase 1 vaccine trial using autologous Langerhans-type dendritic cells (LCs) electroporated with CT7, MAGE-A3, and Wilms tumor 1 (WT1) mRNA in multiple myeloma patients after autologous stem cell transplantation. The study demonstrated the vaccine's safety and ability to induce antigen-specific immune reactivity.
Therapeutic cancer vaccinations have traditionally focused on differentiation antigens, cancer-testis antigens, and overexpressed antigens. Recent studies have highlighted the recognition of neoantigens by T cells, providing a highly specific and immunogenic target for personalized vaccination. Cafri et al. developed a process utilizing tumor-infiltrating lymphocytes to identify immunogenic mutations in patients' tumors. These validated neoantigens and predicted neoepitopes were then combined into a single mRNA construct for vaccination in metastatic gastrointestinal cancer patients. Their Phase I/II trial demonstrated the safety of the vaccine and induced mutation-specific T cell responses against predicted neoepitopes not previously detected [16]. In a phase I trial, researchers used a personalized mRNA neoantigen vaccine, Autogene Cevumeran (a patient specific vaccine incorporating a maximum of 20 neoantigens per patient), in combination with atezolizumab and chemotherapy to treat pancreatic ductal adenocarcinoma (PDAC) patients. The vaccine induced neoantigen-specific T cells in 8 out of 16 patients and correlated with longer recurrence-free survival, independent of patients' immune fitness. This combination treatment shows promise in delaying PDAC recurrence [60], and is gearing up for a subsequent phase II trial. In a phase 2 trial, the combination of mRNA-4157 (V940) and pembrolizumab showed reduced risk of death or developing distant metastasis in high-risk melanoma patients compared to pembrolizumab alone. The combination induced high response rates and improved distant metastasis-free survival. The study demonstrated the potential of personalized mRNA vaccines in improving outcomes for melanoma patients [74].
Clinical trials with mRNA-based vaccines in brain cancer have shown to be promising. Vik-Mo et al. conducted a clinical study with cancer stem cell (CSCs)-mRNA loaded DCs vaccination in glioblastoma patients’ post-radio-chemotherapy. An immune response was identified in all seven patients, with no adverse autoimmune events. Progression-free survival was significantly longer in vaccinated patients compared to controls (Vik-Mo et al. [75]). Cytomegalovirus pp65 is expressed in > 90% of GBM specimens but not surrounding normal brain, it is an ideal tumor-specific target. Vaccination with of pp65 mRNA-pulsed DCs in patients with GBM can extend survival [76], especially when followed by dose-intensified (DI) TMZ and adjuvant GM-CSF. Despite increased Treg proportions, patients receiving pp65- DCs showed long-term progression-free survival and overall survival [77]. Zhu et al. [78] reported a GBM case treated with a combination immunotherapy of tumor-associated antigen (TAAs) and neoantigen mRNA DC vaccines, anti-PD-1, poly I:C, and cyclophosphamide integrated with standard chemoradiation therapy. The patient remained disease-free for 69 months with no immunotherapy-related adverse events, indicating the safety and feasibility of this integrated combination immunotherapy for long-term treatment in this patient.
In summary, an increasing number of mRNA cancer vaccine clinical trials are emerging. Our online search for studies relating to cancer and mRNA vaccines revealed over 80 clinical trials. Of these, 36 are currently active or under recruiting participants.

mRNA cancer vaccine combination with checkpoint inhibitors and CAR-T cell therapy (preclinical)

A potentially groundbreaking approach in cancer treatment would combine mRNA-based vaccines and checkpoint inhibitors, revolutionizing personalized medicine. This strategy leverages the immune system to destroy cancer cells, offering unprecedented precision and effectiveness. The marriage of these therapies is reshaping oncology and inspiring hope in patients and researchers.
In a preclinical study, Wang et al. [79] engineered a cancer vaccine utilizing mRNA and immune checkpoint-blocking siRNA delivered through lipid-coated calcium phosphate nanoparticles. This innovative vaccine elicited robust immune responses, effectively impeding melanoma growth in mice. Simultaneous administration of PD-L1 siRNA and the mRNA vaccine synergistically amplified T cell activation, resulting in remarkable suppression of tumor growth and metastasis. In parallel, Liu et al. employed nanoparticles (NPs) for delivering an mRNA vaccine targeting the tumor antigen MUC1 in a triple-negative breast cancer (TNBC) 4T1 animal model. They combined the mRNA vaccine with an anti-CTLA-4 monoclonal antibody to augment the anti-tumor effects. The findings revealed that the NP-based mRNA vaccine successfully induced a potent and specific in vivo cytotoxic T lymphocyte response against TNBC 4T1 cells. Furthermore, the combination immunotherapy of the vaccine and anti-CTLA-4 monoclonal antibody exhibited a significantly enhanced anti-tumor immune response compared to either the vaccine or monoclonal antibody alone [80]. Chimeric antigen receptor (CAR) technologies have revolutionized the field of hematological malignancies, however, have had minimal success in solid tumors. One innovative approach to increase efficacy is boosting through vaccination. This process involves DCs recruitment, increased tumor antigen uptake, and priming of anti-tumor T cells. CAR-T-derived IFN-gamma plays a crucial role in promoting antigen spreading, enabling complete responses even with antigen-negative tumors. Vaccine boosting is a clinically viable strategy against solid tumors [81].
These advancements, when coupled with the principles of personalized medicine guided by various omics strategies, hold immense potential for revolutionizing glioma therapy. Furthermore, exploring novel immunotherapy approaches, particularly in the context of GBM, may hold the key to enhancing current immune checkpoint inhibitor therapies, considering the historical resistance of GBM to such interventions.

The role of mRNA vaccines in brain cancer

The role of mRNA vaccines in brain cancer is an area of active research and development. Although mRNA vaccines have shown promise in other disease areas, their specific application for brain cancer is still in the early stages.
Due to their potency in stimulation of immunity, as well as the precedent of one FDA approved immunotherapy (Provenge), dendritic cells (DCs) have been a key focus in neoantigen and/or mRNA vaccine research for brain cancer. DC-based vaccines have been demonstrated to be effective to cancers including brain cancer glioma. A recent Phase 3 clinical study investigated the efficacy of adding a tumor lysate-based DC vaccine called DCVax-L to standard treatment for glioblastoma. The results indicated that adding DCVax-L extended the survival of patients with newly diagnosed and recurrent glioblastoma compared to those who received standard treatment alone. This suggests that DCVax-L could be a promising addition to current glioblastoma treatments [13]. A few studies of mRNA-based vaccines have been reported. The receptor for hyaluronan-mediated motility (RHAMM), an overexpressed antigen in brain tumors, has been recently identified as an immunogenic antigen through serological screening of cDNA expression libraries. Researchers investigated the therapeutic potential of DCs transfected with RHAMM mRNA in a glioma model. Mice treated with the vaccine showed significantly prolonged survival compared to control groups. Immunohistochemical analysis demonstrated an increased presence of CD4 + , CD8 + , and CD25 + activated T lymphocytes in the vaccinated group. These findings underscore the therapeutic potential of this approach in treating malignant glioma [82]. In a mouse glioma model, researchers explored the potential of DCs vaccination with IL13ra2 mRNA, a target for glioma immunotherapy. DCs were transfected with Il13ra2 mRNA using a constructed plasmid and injected into the glioma model. The group treated with DCs transfected with Il13ra2 mRNA exhibited a significant improvement in survival rate and increased numbers of CD4 + and CD8 + T lymphocytes. These results emphasize the potential of DC vaccination with Il13ra2 mRNA as a treatment approach for malignant glioma [83]. In a groundbreaking study, a DC vaccine with cancer stem cell (CSCs) mRNA was administered to seven patients with glioblastoma. Following radio-chemotherapy, the vaccine successfully induced an immune response in all patients, leading to a 2.9 times longer progression-free survival compared to controls. These promising results highlight the safety, tolerability, and potential of the vaccine to extend progression-free survival in glioblastoma patients [75]. A study evaluated the combination of dose-intensified temozolomide (DI-TMZ) and DCs pulsed with pp65 lysosome-associated membrane glycoprotein mRNA in glioblastoma patients. Following treatment, there was an increase in pp65 cellular responses, leading to long-term progression-free survival (PFS) and overall survival (OS) exceeding historical controls. Despite an increase in Treg proportions, the combination showed potential benefits in glioblastoma treatment [77].
The long-term durability and prevention of tumor relapse with mRNA vaccines in brain cancer remain under investigation. Nonetheless, ongoing research and advancements stemming from previous preclinical and clinical studies in mRNA vaccine technology hold promise.

Novel tumor antigen identification in brain cancer for mRNA vaccine

To address the urgent need for effective treatments for brain cancer, particularly GBM, a highly lethal brain tumor despite existing treatment options, it is crucial to develop novel immune stimulatory interventions such as mRNA-based therapies. To achieve this, identifying new therapeutic targets or biomarkers is essential. Cell-surface proteins hold significant promise as targets due to their accessibility, involvement in critical signaling pathways, and dysregulation in cancer. Not only can they serve as potential targets for mRNA vaccine strategies, but they also have the potential to be utilized in chimeric antigen receptor (CAR) based immunotherapy approaches.
Brain tumors, including GBM, often exhibit molecular and genetic heterogeneity, meaning there are different subpopulations of cells within the tumor. Designing mRNA vaccines that can effectively target all tumor cell populations poses a challenge due to the diverse antigen profiles. Identifying the most appropriate antigens or neoantigens to target with mRNA vaccines requires thorough characterization of the patient's tumor. Obtaining accurate and comprehensive information about the specific antigens expressed by the tumor cells can be challenging, potentially limiting the efficacy of personalized mRNA vaccines.
Recent studies used bioinformatics analysis to identify potential neoantigens in brain cancer based on abnormal alternative splicing. Novel tumor antigens can be identified in certain cancer type of patients based on genomic, clinical, and mRNA sequence data from their normal and tumor tissues, the neoantigens have been listed in Table 2. In one study seeking to identify novel tumor antigens, Ye et al. analyzed 529 patient with low grade glioma (LGG) tumor samples and clinical data, and selected mutated, up-regulated, prognosis- and immune-related genes. They identified four potential tumor antigens restricted to LGG. These antigens, FCGBP, FLNC, TLR7, and CSF2RA were useful for development of mRNA cancer vaccines [84]. By using the same technology and analyses, Ye et al. further identified ARPC1B and HK3 as potential mRNA antigens for developing GBM mRNA vaccination, and the patients in IS2 were considered the most suitable population for vaccination in GBM [85]. PTBP1 and SLC39A1 were selected as antigens for LGG, and MMP9 and SLC16A3 were identified as antigens for GBM [86]. Zhou et al. identified KDR, COL1A2, and SAMD9 are potential antigens for developing mRNA vaccines against diffuse glioma [87]. TP53, IDH1, C3 and TCF12 four overexpressed and mutated tumor antigens were identified to be associated with poor prognosis and infiltration of antigen presenting cells in glioma patients. The four neoantigens are effective antigens for the development of anti-glioma mRNA vaccines. [88]. Zhong et al. analyzed over 1000 patients’ RNA-seq data and clinical information and identified four glioma antigens, namely ANXA5, FKBP10, MSN, and PYGL, to be associated with superior prognoses. They can also be used as potential antigens for anti-glioma mRNA vaccine production, specifically for patients in immune subtypes 2 and 3 [89]. GBM-associated surfaceome has been used to identify new therapeutic targets for GBM, and 11 specific potential targets found for potential immunotherapy such as mRNA strategy [90]. The immune subtypes of GBM were identified, and a potential neoantigen (LRP1) and patients suitable for vaccination were identified. These findings offer guidance for GBM immunotherapy development. Five potential tumor neoantigens (LRP1, TCF12, DERL3, WIPI2, and TSHZ3) were identified in GBM by selecting genes both with abnormal alternative splicing (upregulated) and gene frameshift mutations, in which LRP1 was significantly associated with APCs [91]. In another study, nine potential tumor antigens were identified, and four immune subtypes with distinct characteristics were validated. ADAMTSL4, COL6A1, CTSL, CYTH4, EGFLAM, LILRB2, MPZL2, SAA2, and LSP1 are candidate antigens, and IS1 and IS2 subtypes are suited for mRNA vaccination and immune checkpoint inhibitors, respectively. This study provides a framework for GBM immunotherapy strategies [92].
Table 2
Novel antigens identified in brain cancer suitable for mRNA vaccine
Brain cancer
Antigens
Reference
Low grade glioma (LGG)
FCGBP, FLNC, TLR7, CSF2RA
[84]
PTBP1, SLC39A1
[86]
Diffuse glioma
KDR, COL1A2, SAMD9
[87]
GBM and glioma
ARPC1B, HK3
[85]
MMP9, SLC16A3
[86]
TP53, IDH1, C3, TCF12
[88]
ANXA5, FKBP10, MSN, PYGL
[89]
LRP1, TCF12, DERL3, WIPI2, TSHZ3
[90]
ADAMTSL4, COL6A1, CTSL, CYTH4, EGFLAM, LILRB2, MPZL2, SAA2, and LSP1
[92]
NAT1, FRRS1, GTF2H2C, BRCA2, GRAP, NR5A2, ABCB4, ZNF90, ERCC6L, ZNF813, and IS1/2A/2B
[93]
ARHGAP9, ARHGAP30, CLEC7A, MAN2B1, ARPC1B and PLB1
[94]
Through the integration of RNA sequencing expression data and somatic mutation data from TCGA-GBM, TCGA-LGG, and CGGA datasets, ten biomarkers (NAT1, FRRS1, GTF2H2C, BRCA2, GRAP, NR5A2, ABCB4, ZNF90, ERCC6L, and ZNF813) have been identified as potential antigens suitable for glioma mRNA vaccine. Additionally, IS1/2A/2B have been determined to be suitable for mRNA vaccination [93]. Ultilizing various methods such as Gene Expression Profiling Interactive Analysis (GEPIA), cBioPortal, TIMER, RNA-seq data analysis, clustering analysis, graph learning-based dimensional reduction, and weighted gene co-expression network analysis (WGCNA), six overexpressed and mutated tumor antigens (ARHGAP9, ARHGAP30, CLEC7A, MAN2B1, ARPC1B and PLB1) have been identified as potential targets for developing anti-GBM mRNA vaccine. Furthermore, three immune subtypes of GBMs have been identified, with GBMs from the IS3 subtype showing a higher likelihood of benefiting from vaccination. The study also used WGCNA to identify potential vaccination biomarkers by clustering immune-related genes [94].
The comprehensive identification of tumor antigens in brain cancer, as detailed above, underscores the remarkable capabilities of contemporary "omics" technology. These advancements, harnessed for antigen discovery, not only bolster the potential of mRNA vaccine development for aggressive brain cancers but also pave the way for personalized, targeted immunotherapies. The integration of multifaceted analytical approaches has unearthed a wealth of potential targets, emphasizing the promising horizon of immunotherapeutic strategies for gliomas and other brain malignancies.

Future directions

Combination therapies involving mRNA vaccines hold significant potential for enhancing the treatment of brain cancer. By combining mRNA vaccines with other treatment modalities, synergistic effects can be achieved to improve outcomes. Here are some potential combination strategies:
  • Immune Checkpoint Inhibitors: Immune checkpoint inhibitors, such as antibodies targeting PD-1/PD-L1 or CTLA-4, have shown promise in enhancing immune responses against cancer cells. In tandem with mRNA vaccines, they can intensify the vaccine-induced immune response, overcoming immune suppression in the tumor microenvironment.
  • Radiation Therapy: Radiation therapy is a standard treatment for brain cancer. Combining mRNA vaccines with radiation therapy can have a dual effect. Radiation therapy can induce tumor cell death, releasing antigens that can be targeted by the mRNA vaccine. Additionally, radiation therapy can also modulate the tumor microenvironment, potentially enhancing the effectiveness of the mRNA vaccine-induced immune response.
  • Targeted Therapies: Targeted therapies, such as tyrosine kinase inhibitors or specific molecular inhibitors, are used to inhibit specific signaling pathways in cancer cells. Combining mRNA vaccines with targeted therapies can potentially enhance the vaccine-induced immune response while simultaneously targeting specific genetic alterations in the tumor cells.
  • Chemotherapy: Chemotherapy drugs are commonly used in the treatment of brain cancer. Combining mRNA vaccines with chemotherapy can potentially increase the immune response against cancer cells while the chemotherapy targets rapidly dividing tumor cells.
  • Adoptive Cell Therapy: Adoptive cell therapy involves the infusion of immune cells, such as CAR-T cells, that are genetically engineered to recognize and attack cancer cells. Combining mRNA vaccines with adoptive cell therapy can further enhance the anti-tumor immune response by priming the infused immune cells to recognize specific tumor antigens.
It is important to note that the choice and sequence of combination therapies should be carefully considered, considering the specific characteristics of the tumor and the patient's condition. As clinical trials progress, the focus remains on delineating the most efficacious combinational regimens. Here are some key areas that require further development:
  • Biomarker Identification: Identifying and validating biomarkers crucial for predicting responses to mRNA vaccines and combination therapies can streamline patient selection, ensuring treatments are optimized for those with the highest likelihood of benefiting.
  • Immunosuppressive Microenvironment: Given the brain tumor’s natural immunosuppressive nature, strategies to reverse or modulate this microenvironment will be key in enhancing mRNA vaccine effectiveness.
  • Vaccine Design: Optimizing mRNA vaccine design, including the choice of antigens, adjuvants, and delivery systems, will be fundamental in boosting their potency against brain cancer.
  • Combination Scheduling: Understanding the optimal scheduling and dosing of combination therapies is crucial. The timing of administering mRNA vaccines in relation to other treatments can influence their synergistic effects.
  • Patient Monitoring and Holistic Care: Incorporating advanced imaging and diagnostic tools is essential for closely monitoring tumor responses and adjusting treatments, with real-time monitoring aiding in the early detection of potential side effects and complications. Furthermore, understanding the emotional and social challenges of brain cancer and giving all-around care, including mental support, is key to improving the patient's life.
The effective application of mRNA vaccines for brain cancer treatment demands a multifaceted approach, encompassing biomarker research, environmental modulation, optimized vaccine design, strategic combination scheduling, and a comprehensive, patient-centric care model.

Conclusion

mRNA vaccines have the potential to revolutionize personalized medicine for brain cancer by targeting specific tumor antigens or neoantigens. They can stimulate a strong immune response while sparing healthy tissues. Benefits include durable immune responses, tumor-specific targeting, and adaptability for multiple antigens. Challenges include overcoming the blood–brain barrier, identifying optimal antigens, navigating the tumor microenvironment, and ensuring vaccine stability and scalability. Research and technological advancements are needed to refine delivery systems, understand tumor biology, optimize vaccine design, and develop effective combination therapies. Collaboration among researchers, clinicians, and industry partners is crucial for advancing personalized medicine in brain cancer using mRNA vaccines.

Acknowledgements

Not available.

Declarations

Not available.
All authors have read and approved the final manuscript and agree to its submission to JTM.

Competing interests

The authors assume complete responsibility for the content of this article and affirm that there are no conflicts of interest related to this publication. Details of the authors' employment affiliations can be found on the cover page. This review article was independently initiated and has not received any financial support. The perspectives and opinions articulated in this paper reflect those of the authors exclusively. None of the authors have affiliations or financial engagements with any entity possessing a monetary stake in or conflict with the topics or materials elaborated upon in this manuscript. This encompasses employment, consultancies, honoraria, stock or option ownership, expert testimonies, grants, patents (either received or pending), or royalties.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Abul-Husn NS, Owusu Obeng A, Sanderson SC, Gottesman O, Scott SA. Implementation and utilization of genetic testing in personalized medicine. Pharmgenomics Pers Med. 2014;7:227–40.PubMedPubMedCentral Abul-Husn NS, Owusu Obeng A, Sanderson SC, Gottesman O, Scott SA. Implementation and utilization of genetic testing in personalized medicine. Pharmgenomics Pers Med. 2014;7:227–40.PubMedPubMedCentral
2.
Zurück zum Zitat Strianese O, Rizzo F, Ciccarelli M, Galasso G, D’Agostino Y, Salvati A, Del Giudice C, Tesorio P, Rusciano MR. Precision and personalized medicine: how genomic approach improves the management of cardiovascular and neurodegenerative disease. Genes. 2020;11:747.PubMedPubMedCentralCrossRef Strianese O, Rizzo F, Ciccarelli M, Galasso G, D’Agostino Y, Salvati A, Del Giudice C, Tesorio P, Rusciano MR. Precision and personalized medicine: how genomic approach improves the management of cardiovascular and neurodegenerative disease. Genes. 2020;11:747.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Aldape K, Brindle KM, Chesler L, Chopra R, Gajjar A, Gilbert MR, Gottardo N, Gutmann DH, Hargrave D, Holland EC, et al. Challenges to curing primary brain tumours. Nat Rev Clin Oncol. 2019;16:509–20.PubMedPubMedCentralCrossRef Aldape K, Brindle KM, Chesler L, Chopra R, Gajjar A, Gilbert MR, Gottardo N, Gutmann DH, Hargrave D, Holland EC, et al. Challenges to curing primary brain tumours. Nat Rev Clin Oncol. 2019;16:509–20.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Marei HE, Hasan A, Pozzoli G, Cenciarelli C. Cancer immunotherapy with immune checkpoint inhibitors (ICIs): potential, mechanisms of resistance, and strategies for reinvigorating T cell responsiveness when resistance is acquired. Cancer Cell Int. 2023;23:64.PubMedPubMedCentralCrossRef Marei HE, Hasan A, Pozzoli G, Cenciarelli C. Cancer immunotherapy with immune checkpoint inhibitors (ICIs): potential, mechanisms of resistance, and strategies for reinvigorating T cell responsiveness when resistance is acquired. Cancer Cell Int. 2023;23:64.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Arrieta VA, Dmello C, McGrail DJ, Brat DJ, Lee-Chang C, Heimberger AB, Chand D, Stupp R, Sonabend AM. Immune checkpoint blockade in glioblastoma: from tumor heterogeneity to personalized treatment. J Clin Invest. 2023;133:e163447.PubMedPubMedCentralCrossRef Arrieta VA, Dmello C, McGrail DJ, Brat DJ, Lee-Chang C, Heimberger AB, Chand D, Stupp R, Sonabend AM. Immune checkpoint blockade in glioblastoma: from tumor heterogeneity to personalized treatment. J Clin Invest. 2023;133:e163447.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Lin F, Anekoji M, Ichim TE, Pingle S, Patro KCP, Jones LD, Datta S, Kesari S, Ashili S. Immunotherapetuic peptide vaccine approaches to glioma. J Cancer Res Rev Reports. 2021;3:1–11.CrossRef Lin F, Anekoji M, Ichim TE, Pingle S, Patro KCP, Jones LD, Datta S, Kesari S, Ashili S. Immunotherapetuic peptide vaccine approaches to glioma. J Cancer Res Rev Reports. 2021;3:1–11.CrossRef
7.
Zurück zum Zitat Mullard A. FDA approves 100th monoclonal antibody product. Nat Rev Drug Discov. 2021;20:491–5.PubMedCrossRef Mullard A. FDA approves 100th monoclonal antibody product. Nat Rev Drug Discov. 2021;20:491–5.PubMedCrossRef
8.
Zurück zum Zitat Mullard A. FDA approves first BCMA-targeted CAR-T cell therapy. Nat Rev Drug Discov. 2021;20:332.PubMed Mullard A. FDA approves first BCMA-targeted CAR-T cell therapy. Nat Rev Drug Discov. 2021;20:332.PubMed
9.
Zurück zum Zitat Mullard A. FDA approves fourth CAR-T cell therapy. Nat Rev Drug Discov. 2021;20:166.PubMed Mullard A. FDA approves fourth CAR-T cell therapy. Nat Rev Drug Discov. 2021;20:166.PubMed
10.
11.
Zurück zum Zitat DeCordova S, Shastri A, Tsolaki AG, Yasmin H, Klein L, Singh SK, Kishore U. Molecular heterogeneity and immunosuppressive microenvironment in glioblastoma. Front Immunol. 2020;11:1402.PubMedPubMedCentralCrossRef DeCordova S, Shastri A, Tsolaki AG, Yasmin H, Klein L, Singh SK, Kishore U. Molecular heterogeneity and immunosuppressive microenvironment in glioblastoma. Front Immunol. 2020;11:1402.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Guo C, Manjili MH, Subjeck JR, Sarkar D, Fisher PB, Wang XY. Therapeutic cancer vaccines: past, present, and future. Adv Cancer Res. 2013;119:421–75.PubMedPubMedCentralCrossRef Guo C, Manjili MH, Subjeck JR, Sarkar D, Fisher PB, Wang XY. Therapeutic cancer vaccines: past, present, and future. Adv Cancer Res. 2013;119:421–75.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Liau LM, Ashkan K, Brem S, Campian JL, Trusheim JE, Iwamoto FM, Tran DD, Ansstas G, Cobbs CS, Heth JA, et al. Association of autologous tumor lysate-loaded dendritic cell vaccination with extension of survival among patients with newly diagnosed and recurrent glioblastoma: a phase 3 prospective externally controlled cohort trial. JAMA Oncol. 2023;9:112–21.PubMedCrossRef Liau LM, Ashkan K, Brem S, Campian JL, Trusheim JE, Iwamoto FM, Tran DD, Ansstas G, Cobbs CS, Heth JA, et al. Association of autologous tumor lysate-loaded dendritic cell vaccination with extension of survival among patients with newly diagnosed and recurrent glioblastoma: a phase 3 prospective externally controlled cohort trial. JAMA Oncol. 2023;9:112–21.PubMedCrossRef
14.
Zurück zum Zitat Chen KS, Reinshagen C, Van Schaik TA, Rossignoli F, Borges P, Mendonca NC, Abdi R, Simon B, Reardon DA, Wakimoto H, Shah K. Bifunctional cancer cell-based vaccine concomitantly drives direct tumor killing and antitumor immunity. Sci Transl Med. 2023;15:4778.CrossRef Chen KS, Reinshagen C, Van Schaik TA, Rossignoli F, Borges P, Mendonca NC, Abdi R, Simon B, Reardon DA, Wakimoto H, Shah K. Bifunctional cancer cell-based vaccine concomitantly drives direct tumor killing and antitumor immunity. Sci Transl Med. 2023;15:4778.CrossRef
15.
Zurück zum Zitat Lin F, Ichim T, Pingle S, Patro KCP, Loren M, Jones LD, Datta S, Kesari S, Ashili S. Immunotherapeutic peptide vaccine approaches to glioma. J Cancer Res Rev Reports. 2021;3:1–11.CrossRef Lin F, Ichim T, Pingle S, Patro KCP, Loren M, Jones LD, Datta S, Kesari S, Ashili S. Immunotherapeutic peptide vaccine approaches to glioma. J Cancer Res Rev Reports. 2021;3:1–11.CrossRef
16.
Zurück zum Zitat Cafri G, Gartner JJ, Zaks T, Hopson K, Levin N, Paria BC, Parkhurst MR, Yossef R, Lowery FJ, Jafferji MS, et al. mRNA vaccine-induced neoantigen-specific T cell immunity in patients with gastrointestinal cancer. J Clin Invest. 2020;130:5976–88.PubMedPubMedCentralCrossRef Cafri G, Gartner JJ, Zaks T, Hopson K, Levin N, Paria BC, Parkhurst MR, Yossef R, Lowery FJ, Jafferji MS, et al. mRNA vaccine-induced neoantigen-specific T cell immunity in patients with gastrointestinal cancer. J Clin Invest. 2020;130:5976–88.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Turan T, Kannan D, Patel M, Matthew Barnes J, Tanlimco SG, Lu R, Halliwill K, Kongpachith S, Kline DE, Hendrickx W, et al. Immune oncology, immune responsiveness and the theory of everything. J Immunother Cancer. 2018;6:50.PubMedPubMedCentralCrossRef Turan T, Kannan D, Patel M, Matthew Barnes J, Tanlimco SG, Lu R, Halliwill K, Kongpachith S, Kline DE, Hendrickx W, et al. Immune oncology, immune responsiveness and the theory of everything. J Immunother Cancer. 2018;6:50.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Galluzzi L, Kepp O, Hett E, Kroemer G, Marincola FM. Immunogenic cell death in cancer: concept and therapeutic implications. J Transl Med. 2023;21:162.PubMedPubMedCentralCrossRef Galluzzi L, Kepp O, Hett E, Kroemer G, Marincola FM. Immunogenic cell death in cancer: concept and therapeutic implications. J Transl Med. 2023;21:162.PubMedPubMedCentralCrossRef
19.
20.
Zurück zum Zitat Trimble CL, Morrow MP, Kraynyak KA, Shen X, Dallas M, Yan J, Edwards L, Parker RL, Denny L, Giffear M, et al. Safety, efficacy, and immunogenicity of VGX-3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2b trial. Lancet. 2015;386:2078–88.PubMedPubMedCentralCrossRef Trimble CL, Morrow MP, Kraynyak KA, Shen X, Dallas M, Yan J, Edwards L, Parker RL, Denny L, Giffear M, et al. Safety, efficacy, and immunogenicity of VGX-3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2b trial. Lancet. 2015;386:2078–88.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Bagarazzi ML, Yan J, Morrow MP, Shen X, Parker RL, Lee JC, Giffear M, Pankhong P, Khan AS, Broderick KE, et al. Immunotherapy against HPV16/18 generates potent TH1 and cytotoxic cellular immune responses. Sci Transl Med. 2012;4:155ra138.PubMedPubMedCentralCrossRef Bagarazzi ML, Yan J, Morrow MP, Shen X, Parker RL, Lee JC, Giffear M, Pankhong P, Khan AS, Broderick KE, et al. Immunotherapy against HPV16/18 generates potent TH1 and cytotoxic cellular immune responses. Sci Transl Med. 2012;4:155ra138.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Iavarone C, O’Hagan DT, Yu D, Delahaye NF, Ulmer JB. Mechanism of action of mRNA-based vaccines. Expert Rev Vaccines. 2017;16:871–81.PubMedCrossRef Iavarone C, O’Hagan DT, Yu D, Delahaye NF, Ulmer JB. Mechanism of action of mRNA-based vaccines. Expert Rev Vaccines. 2017;16:871–81.PubMedCrossRef
23.
Zurück zum Zitat Wadhwa A, Aljabbari A, Lokras A, Foged C, Thakur A. Opportunities and challenges in the delivery of mRNA-based vaccines. Pharmaceutics. 2020;12:102.PubMedPubMedCentralCrossRef Wadhwa A, Aljabbari A, Lokras A, Foged C, Thakur A. Opportunities and challenges in the delivery of mRNA-based vaccines. Pharmaceutics. 2020;12:102.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Racine-Brzostek SE, Yee JK, Sukhu A, Qiu Y, Rand S, Barone PD, Hao Y, Yang HS, Meng QH, Apple FS, et al. Rapid, robust, and sustainable antibody responses to mRNA COVID-19 vaccine in convalescent COVID-19 individuals. JCI Insight. 2021;6:e151477.PubMedPubMedCentralCrossRef Racine-Brzostek SE, Yee JK, Sukhu A, Qiu Y, Rand S, Barone PD, Hao Y, Yang HS, Meng QH, Apple FS, et al. Rapid, robust, and sustainable antibody responses to mRNA COVID-19 vaccine in convalescent COVID-19 individuals. JCI Insight. 2021;6:e151477.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Goel RR, Apostolidis SA, Painter MM, Mathew D, Pattekar A, Kuthuru O, Gouma S, Kuri-Cervantes L, Meng W, Adamski S, et al. Longitudinal analysis reveals distinct antibody and memory B cell responses in SARS-CoV2 naive and recovered individuals following mRNA vaccination. MedRxiv. 2021;6:eabi6950. Goel RR, Apostolidis SA, Painter MM, Mathew D, Pattekar A, Kuthuru O, Gouma S, Kuri-Cervantes L, Meng W, Adamski S, et al. Longitudinal analysis reveals distinct antibody and memory B cell responses in SARS-CoV2 naive and recovered individuals following mRNA vaccination. MedRxiv. 2021;6:eabi6950.
26.
Zurück zum Zitat Campo F, Venuti A, Pimpinelli F, Abril E, Blandino G, Conti L, De Virgilio A, De Marco F, Di Noia V, Di Domenico EG, et al. Antibody persistence 6 months post-vaccination with BNT162b2 among health care workers. Vaccines. 2021;9:1125.PubMedPubMedCentralCrossRef Campo F, Venuti A, Pimpinelli F, Abril E, Blandino G, Conti L, De Virgilio A, De Marco F, Di Noia V, Di Domenico EG, et al. Antibody persistence 6 months post-vaccination with BNT162b2 among health care workers. Vaccines. 2021;9:1125.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Lederer K, Castano D, Gomez Atria D, Oguin TH 3rd, Wang S, Manzoni TB, Muramatsu H, Hogan MJ, Amanat F, Cherubin P, et al. SARS-CoV-2 mRNA vaccines foster potent antigen-specific germinal center responses associated with neutralizing antibody generation. Immunity. 2020;53(1281–1295): e1285. Lederer K, Castano D, Gomez Atria D, Oguin TH 3rd, Wang S, Manzoni TB, Muramatsu H, Hogan MJ, Amanat F, Cherubin P, et al. SARS-CoV-2 mRNA vaccines foster potent antigen-specific germinal center responses associated with neutralizing antibody generation. Immunity. 2020;53(1281–1295): e1285.
28.
Zurück zum Zitat Sahin U, Muik A, Derhovanessian E, Vogler I, Kranz LM, Vormehr M, Baum A, Pascal K, Quandt J, Maurus D, et al. COVID-19 vaccine BNT162b1 elicits human antibody and TH1 T cell responses. Nature. 2020;586:594–9.PubMedCrossRef Sahin U, Muik A, Derhovanessian E, Vogler I, Kranz LM, Vormehr M, Baum A, Pascal K, Quandt J, Maurus D, et al. COVID-19 vaccine BNT162b1 elicits human antibody and TH1 T cell responses. Nature. 2020;586:594–9.PubMedCrossRef
30.
Zurück zum Zitat Chakraborty C, Sharma AR, Bhattacharya M, Lee SS. From COVID-19 to cancer mRNA vaccines: moving from bench to clinic in the vaccine landscape. Front Immunol. 2021;12: 679344.PubMedPubMedCentralCrossRef Chakraborty C, Sharma AR, Bhattacharya M, Lee SS. From COVID-19 to cancer mRNA vaccines: moving from bench to clinic in the vaccine landscape. Front Immunol. 2021;12: 679344.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Koh EY, Ho SC, Mariati A, Song Z, Bi X, Bardor M, Yang Y. An internal ribosome entry site (IRES) mutant library for tuning expression level of multiple genes in mammalian cells. PLoS ONE. 2013;8:e82100.PubMedPubMedCentralCrossRef Koh EY, Ho SC, Mariati A, Song Z, Bi X, Bardor M, Yang Y. An internal ribosome entry site (IRES) mutant library for tuning expression level of multiple genes in mammalian cells. PLoS ONE. 2013;8:e82100.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat To KKW, Cho WCS. An overview of rational design of mRNA-based therapeutics and vaccines. Expert Opin Drug Discov. 2021;16:1307–17.PubMedCrossRef To KKW, Cho WCS. An overview of rational design of mRNA-based therapeutics and vaccines. Expert Opin Drug Discov. 2021;16:1307–17.PubMedCrossRef
34.
Zurück zum Zitat Oberli MA, Reichmuth AM, Dorkin JR, Mitchell MJ, Fenton OS, Jaklenec A, Anderson DG, Langer R, Blankschtein D. Lipid nanoparticle assisted mRNA delivery for potent cancer immunotherapy. Nano Lett. 2017;17:1326–35.PubMedCrossRef Oberli MA, Reichmuth AM, Dorkin JR, Mitchell MJ, Fenton OS, Jaklenec A, Anderson DG, Langer R, Blankschtein D. Lipid nanoparticle assisted mRNA delivery for potent cancer immunotherapy. Nano Lett. 2017;17:1326–35.PubMedCrossRef
35.
Zurück zum Zitat Hou X, Zaks T, Langer R, Dong Y. Lipid nanoparticles for mRNA delivery. Nat Rev Mater. 2021;6:1–17.CrossRef Hou X, Zaks T, Langer R, Dong Y. Lipid nanoparticles for mRNA delivery. Nat Rev Mater. 2021;6:1–17.CrossRef
37.
Zurück zum Zitat Park KS, Sun X, Aikins ME, Moon JJ. Non-viral COVID-19 vaccine delivery systems. Adv Drug Deliv Rev. 2021;169:137–51.PubMedCrossRef Park KS, Sun X, Aikins ME, Moon JJ. Non-viral COVID-19 vaccine delivery systems. Adv Drug Deliv Rev. 2021;169:137–51.PubMedCrossRef
38.
Zurück zum Zitat Zeng C, Zhang C, Walker PG, Dong Y. Formulation and delivery technologies for mRNA vaccines. Curr Top Microbiol Immunol. 2020;440:71–110. Zeng C, Zhang C, Walker PG, Dong Y. Formulation and delivery technologies for mRNA vaccines. Curr Top Microbiol Immunol. 2020;440:71–110.
39.
Zurück zum Zitat Rohovie MJ, Nagasawa M, Swartz JR. Virus-like particles: Next-generation nanoparticles for targeted therapeutic delivery. Bioeng Transl Med. 2017;2:43–57.PubMedPubMedCentralCrossRef Rohovie MJ, Nagasawa M, Swartz JR. Virus-like particles: Next-generation nanoparticles for targeted therapeutic delivery. Bioeng Transl Med. 2017;2:43–57.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Tan JS, Jaffar Ali MNB, Gan BK, Tan WS. Next-generation viral nanoparticles for targeted delivery of therapeutics: Fundamentals, methods, biomedical applications, and challenges. Expert Opin Drug Deliv. 2023;20:955–78.PubMedCrossRef Tan JS, Jaffar Ali MNB, Gan BK, Tan WS. Next-generation viral nanoparticles for targeted delivery of therapeutics: Fundamentals, methods, biomedical applications, and challenges. Expert Opin Drug Deliv. 2023;20:955–78.PubMedCrossRef
41.
Zurück zum Zitat 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 U S A. 2021;118:e2005191118.PubMedPubMedCentralCrossRef 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 U S A. 2021;118:e2005191118.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Verbeke R, Lentacker I, Breckpot K, Janssens J, Van Calenbergh S, De Smedt SC, Dewitte H. Broadening the message: a nanovaccine co-loaded with messenger RNA and alpha-GalCer induces antitumor immunity through conventional and natural killer T cells. ACS Nano. 2019;13:1655–69.PubMed Verbeke R, Lentacker I, Breckpot K, Janssens J, Van Calenbergh S, De Smedt SC, Dewitte H. Broadening the message: a nanovaccine co-loaded with messenger RNA and alpha-GalCer induces antitumor immunity through conventional and natural killer T cells. ACS Nano. 2019;13:1655–69.PubMed
43.
Zurück zum Zitat Fornaguera C, Diaz-Caballero M, Garcia-Fernandez C, Olmo L, Stampa-Lopez Pinto M, Navalon-Lopez M, Guerra-Rebollo M, Borros S. Synthesis and characterization of mRNA-loaded Poly(Beta Aminoesters) nanoparticles for vaccination purposes. J Vis Exp. 2021. https://doi.org/10.3791/62889.CrossRefPubMed Fornaguera C, Diaz-Caballero M, Garcia-Fernandez C, Olmo L, Stampa-Lopez Pinto M, Navalon-Lopez M, Guerra-Rebollo M, Borros S. Synthesis and characterization of mRNA-loaded Poly(Beta Aminoesters) nanoparticles for vaccination purposes. J Vis Exp. 2021. https://​doi.​org/​10.​3791/​62889.CrossRefPubMed
44.
Zurück zum Zitat Fan YN, Li M, Luo YL, Chen Q, Wang L, Zhang HB, Shen S, Gu Z, Wang J. Cationic lipid-assisted nanoparticles for delivery of mRNA cancer vaccine. Biomater Sci. 2018;6:3009–18.PubMedCrossRef Fan YN, Li M, Luo YL, Chen Q, Wang L, Zhang HB, Shen S, Gu Z, Wang J. Cationic lipid-assisted nanoparticles for delivery of mRNA cancer vaccine. Biomater Sci. 2018;6:3009–18.PubMedCrossRef
45.
Zurück zum Zitat Mai Y, Guo J, Zhao Y, Ma S, Hou Y, Yang J. Intranasal delivery of cationic liposome-protamine complex mRNA vaccine elicits effective anti-tumor immunity. Cell Immunol. 2020;354: 104143.PubMedCrossRef Mai Y, Guo J, Zhao Y, Ma S, Hou Y, Yang J. Intranasal delivery of cationic liposome-protamine complex mRNA vaccine elicits effective anti-tumor immunity. Cell Immunol. 2020;354: 104143.PubMedCrossRef
46.
Zurück zum Zitat Persano S, Guevara ML, Li Z, Mai J, Ferrari M, Pompa PP, Shen H. Lipopolyplex potentiates anti-tumor immunity of mRNA-based vaccination. Biomaterials. 2017;125:81–9.PubMedPubMedCentralCrossRef Persano S, Guevara ML, Li Z, Mai J, Ferrari M, Pompa PP, Shen H. Lipopolyplex potentiates anti-tumor immunity of mRNA-based vaccination. Biomaterials. 2017;125:81–9.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Haabeth OAW, Blake TR, McKinlay CJ, Waymouth RM, Wender PA, Levy R. mRNA vaccination with charge-altering releasable transporters elicits human T cell responses and cures established tumors in mice. Proc Natl Acad Sci USA. 2018;115:E9153–61.PubMedPubMedCentralCrossRef Haabeth OAW, Blake TR, McKinlay CJ, Waymouth RM, Wender PA, Levy R. mRNA vaccination with charge-altering releasable transporters elicits human T cell responses and cures established tumors in mice. Proc Natl Acad Sci USA. 2018;115:E9153–61.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Hess PR, Boczkowski D, Nair SK, Snyder D, Gilboa E. 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;55:672–83.PubMedCrossRef Hess PR, Boczkowski D, Nair SK, Snyder D, Gilboa E. 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;55:672–83.PubMedCrossRef
49.
Zurück zum Zitat Li Q, Ren J, Liu W, Jiang G, Hu R. CpG oligodeoxynucleotide developed to activate primate immune responses promotes antitumoral effects in combination with a neoantigen-based mRNA cancer vaccine. Drug Des Devel Ther. 2021;15:3953–63.PubMedPubMedCentralCrossRef Li Q, Ren J, Liu W, Jiang G, Hu R. CpG oligodeoxynucleotide developed to activate primate immune responses promotes antitumoral effects in combination with a neoantigen-based mRNA cancer vaccine. Drug Des Devel Ther. 2021;15:3953–63.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Fotin-Mleczek M, Duchardt KM, Lorenz C, Pfeiffer R, Ojkic-Zrna S, Probst J, Kallen KJ. Messenger RNA-based vaccines with dual activity induce balanced TLR-7 dependent adaptive immune responses and provide antitumor activity. J Immunother. 2011;34:1–15.PubMedCrossRef Fotin-Mleczek M, Duchardt KM, Lorenz C, Pfeiffer R, Ojkic-Zrna S, Probst J, Kallen KJ. Messenger RNA-based vaccines with dual activity induce balanced TLR-7 dependent adaptive immune responses and provide antitumor activity. J Immunother. 2011;34:1–15.PubMedCrossRef
51.
Zurück zum Zitat Islam MA, Rice J, Reesor E, Zope H, Tao W, Lim M, Ding J, Chen Y, Aduluso D, Zetter BR, et al. Adjuvant-pulsed mRNA vaccine nanoparticle for immunoprophylactic and therapeutic tumor suppression in mice. Biomaterials. 2021;266: 120431.PubMedCrossRef Islam MA, Rice J, Reesor E, Zope H, Tao W, Lim M, Ding J, Chen Y, Aduluso D, Zetter BR, et al. Adjuvant-pulsed mRNA vaccine nanoparticle for immunoprophylactic and therapeutic tumor suppression in mice. Biomaterials. 2021;266: 120431.PubMedCrossRef
52.
Zurück zum Zitat 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;4:146–56.PubMedCrossRef 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;4:146–56.PubMedCrossRef
53.
Zurück zum Zitat Bialkowski L, van Weijnen A, Van der Jeught K, Renmans D, Daszkiewicz L, Heirman C, Stange G, Breckpot K, Aerts JL, Thielemans K. Intralymphatic mRNA vaccine induces CD8 T-cell responses that inhibit the growth of mucosally located tumours. Sci Rep. 2016;6:22509.PubMedPubMedCentralCrossRef Bialkowski L, van Weijnen A, Van der Jeught K, Renmans D, Daszkiewicz L, Heirman C, Stange G, Breckpot K, Aerts JL, Thielemans K. Intralymphatic mRNA vaccine induces CD8 T-cell responses that inhibit the growth of mucosally located tumours. Sci Rep. 2016;6:22509.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Maruggi G, Zhang C, Li J, Ulmer JB, Yu D. mRNA as a transformative technology for vaccine development to control infectious diseases. Mol Ther. 2019;27:757–72.PubMedPubMedCentralCrossRef Maruggi G, Zhang C, Li J, Ulmer JB, Yu D. mRNA as a transformative technology for vaccine development to control infectious diseases. Mol Ther. 2019;27:757–72.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Liu C, Shi Q, Huang X, Koo S, Kong N, Tao W. mRNA-based cancer therapeutics. Nat Rev Cancer. 2023;23:526–43.PubMedCrossRef Liu C, Shi Q, Huang X, Koo S, Kong N, Tao W. mRNA-based cancer therapeutics. Nat Rev Cancer. 2023;23:526–43.PubMedCrossRef
59.
Zurück zum Zitat mRNA Vaccine slows melanoma recurrence. Cancer Discov 2023; 13:1278 mRNA Vaccine slows melanoma recurrence. Cancer Discov 2023; 13:1278
60.
Zurück zum Zitat 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;618:144–50.PubMedPubMedCentralCrossRef 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;618:144–50.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Rittig SM, Haentschel M, Weimer KJ, Heine A, Muller MR, Brugger W, Horger MS, Maksimovic O, Stenzl A, Hoerr I, et al. Long-term survival correlates with immunological responses in renal cell carcinoma patients treated with mRNA-based immunotherapy. Oncoimmunology. 2016;5: e1108511.PubMedCrossRef Rittig SM, Haentschel M, Weimer KJ, Heine A, Muller MR, Brugger W, Horger MS, Maksimovic O, Stenzl A, Hoerr I, et al. Long-term survival correlates with immunological responses in renal cell carcinoma patients treated with mRNA-based immunotherapy. Oncoimmunology. 2016;5: e1108511.PubMedCrossRef
62.
Zurück zum Zitat Rittig SM, Haentschel M, Weimer KJ, Heine A, Muller MR, Brugger W, Horger MS, Maksimovic O, Stenzl A, Hoerr I, et al. Intradermal vaccinations with RNA coding for TAA generate CD8+ and CD4+ immune responses and induce clinical benefit in vaccinated patients. Mol Ther. 2011;19:990–9.PubMedCrossRef Rittig SM, Haentschel M, Weimer KJ, Heine A, Muller MR, Brugger W, Horger MS, Maksimovic O, Stenzl A, Hoerr I, et al. Intradermal vaccinations with RNA coding for TAA generate CD8+ and CD4+ immune responses and induce clinical benefit in vaccinated patients. Mol Ther. 2011;19:990–9.PubMedCrossRef
63.
Zurück zum Zitat Weide B, Pascolo S, Scheel B, Derhovanessian E, Pflugfelder A, Eigentler TK, Pawelec G, Hoerr I, Rammensee HG, Garbe C. Direct injection of protamine-protected mRNA: results of a phase 1/2 vaccination trial in metastatic melanoma patients. J Immunother. 2009;32:498–507.PubMedCrossRef Weide B, Pascolo S, Scheel B, Derhovanessian E, Pflugfelder A, Eigentler TK, Pawelec G, Hoerr I, Rammensee HG, Garbe C. Direct injection of protamine-protected mRNA: results of a phase 1/2 vaccination trial in metastatic melanoma patients. J Immunother. 2009;32:498–507.PubMedCrossRef
64.
Zurück zum Zitat Bidram M, Zhao Y, Shebardina NG, Baldin AV, Bazhin AV, Ganjalikhany MR, Zamyatnin AA Jr, Ganjalikhani-Hakemi M. mRNA-Based cancer vaccines: a therapeutic strategy for the treatment of melanoma patients. Vaccines. 2021;9:1060.PubMedPubMedCentralCrossRef Bidram M, Zhao Y, Shebardina NG, Baldin AV, Bazhin AV, Ganjalikhany MR, Zamyatnin AA Jr, Ganjalikhani-Hakemi M. mRNA-Based cancer vaccines: a therapeutic strategy for the treatment of melanoma patients. Vaccines. 2021;9:1060.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Rausch S, Schwentner C, Stenzl A, Bedke J. mRNA vaccine CV9103 and CV9104 for the treatment of prostate cancer. Hum Vaccin Immunother. 2014;10:3146–52.PubMedPubMedCentralCrossRef Rausch S, Schwentner C, Stenzl A, Bedke J. mRNA vaccine CV9103 and CV9104 for the treatment of prostate cancer. Hum Vaccin Immunother. 2014;10:3146–52.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Van Nuffel AM, Benteyn D, Wilgenhof S, Pierret L, Corthals J, Heirman C, van der Bruggen P, Coulie PG, Neyns B, Thielemans K, Bonehill A. Dendritic cells loaded with mRNA encoding full-length tumor antigens prime CD4+ and CD8+ T cells in melanoma patients. Mol Ther. 2012;20:1063–74.PubMedPubMedCentralCrossRef Van Nuffel AM, Benteyn D, Wilgenhof S, Pierret L, Corthals J, Heirman C, van der Bruggen P, Coulie PG, Neyns B, Thielemans K, Bonehill A. Dendritic cells loaded with mRNA encoding full-length tumor antigens prime CD4+ and CD8+ T cells in melanoma patients. Mol Ther. 2012;20:1063–74.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Van Nuffel AM, Benteyn D, Wilgenhof S, Corthals J, Heirman C, Neyns B, Thielemans K, Bonehill A. Intravenous and intradermal TriMix-dendritic cell therapy results in a broad T-cell response and durable tumor response in a chemorefractory stage IV-M1c melanoma patient. Cancer Immunol Immunother. 2012;61:1033–43.PubMedCrossRef Van Nuffel AM, Benteyn D, Wilgenhof S, Corthals J, Heirman C, Neyns B, Thielemans K, Bonehill A. Intravenous and intradermal TriMix-dendritic cell therapy results in a broad T-cell response and durable tumor response in a chemorefractory stage IV-M1c melanoma patient. Cancer Immunol Immunother. 2012;61:1033–43.PubMedCrossRef
68.
Zurück zum Zitat Su Z, Dannull J, Yang BK, Dahm P, Coleman D, Yancey D, Sichi S, Niedzwiecki D, Boczkowski D, Gilboa E, Vieweg J. Telomerase mRNA-transfected dendritic cells stimulate antigen-specific CD8+ and CD4+ T cell responses in patients with metastatic prostate cancer. J Immunol. 2005;174:3798–807.PubMedCrossRef Su Z, Dannull J, Yang BK, Dahm P, Coleman D, Yancey D, Sichi S, Niedzwiecki D, Boczkowski D, Gilboa E, Vieweg J. Telomerase mRNA-transfected dendritic cells stimulate antigen-specific CD8+ and CD4+ T cell responses in patients with metastatic prostate cancer. J Immunol. 2005;174:3798–807.PubMedCrossRef
69.
Zurück zum Zitat Su Z, Vieweg J, Weizer AZ, Dahm P, Yancey D, Turaga V, Higgins J, Boczkowski D, Gilboa E, Dannull J. Enhanced induction of telomerase-specific CD4(+) T cells using dendritic cells transfected with RNA encoding a chimeric gene product. Cancer Res. 2002;62:5041–8.PubMed Su Z, Vieweg J, Weizer AZ, Dahm P, Yancey D, Turaga V, Higgins J, Boczkowski D, Gilboa E, Dannull J. Enhanced induction of telomerase-specific CD4(+) T cells using dendritic cells transfected with RNA encoding a chimeric gene product. Cancer Res. 2002;62:5041–8.PubMed
70.
Zurück zum Zitat Kyte JA, Mu L, Aamdal S, Kvalheim G, Dueland S, Hauser M, Gullestad HP, Ryder T, Lislerud K, Hammerstad H, Gaudernack G. Phase I/II trial of melanoma therapy with dendritic cells transfected with autologous tumor-mRNA. Cancer Gene Ther. 2006;13:905–18.PubMedCrossRef Kyte JA, Mu L, Aamdal S, Kvalheim G, Dueland S, Hauser M, Gullestad HP, Ryder T, Lislerud K, Hammerstad H, Gaudernack G. Phase I/II trial of melanoma therapy with dendritic cells transfected with autologous tumor-mRNA. Cancer Gene Ther. 2006;13:905–18.PubMedCrossRef
71.
Zurück zum Zitat Lesterhuis WJ, De Vries IJ, Schreibelt G, Schuurhuis DH, Aarntzen EH, De Boer A, Scharenborg NM, Van De Rakt M, Hesselink EJ, Figdor CG, et al. Immunogenicity of dendritic cells pulsed with CEA peptide or transfected with CEA mRNA for vaccination of colorectal cancer patients. Anticancer Res. 2010;30:5091–7.PubMed Lesterhuis WJ, De Vries IJ, Schreibelt G, Schuurhuis DH, Aarntzen EH, De Boer A, Scharenborg NM, Van De Rakt M, Hesselink EJ, Figdor CG, et al. Immunogenicity of dendritic cells pulsed with CEA peptide or transfected with CEA mRNA for vaccination of colorectal cancer patients. Anticancer Res. 2010;30:5091–7.PubMed
72.
Zurück zum Zitat Van de Velde AL, Beutels P, Smits EL, Van Tendeloo VF, Nijs G, Anguille S, Verlinden A, Gadisseur AP, Schroyens WA, Dom S, et al. Medical costs of treatment and survival of patients with acute myeloid leukemia in Belgium. Leuk Res. 2016;46:26–9.PubMedCrossRef Van de Velde AL, Beutels P, Smits EL, Van Tendeloo VF, Nijs G, Anguille S, Verlinden A, Gadisseur AP, Schroyens WA, Dom S, et al. Medical costs of treatment and survival of patients with acute myeloid leukemia in Belgium. Leuk Res. 2016;46:26–9.PubMedCrossRef
73.
Zurück zum Zitat Chung DJ, Sharma S, Rangesa M, DeWolf S, Elhanati Y, Perica K, Young JW. Langerhans dendritic cell vaccine bearing mRNA-encoded tumor antigens induces antimyeloma immunity after autotransplant. Blood Adv. 2022;6:1547–58.PubMedPubMedCentralCrossRef Chung DJ, Sharma S, Rangesa M, DeWolf S, Elhanati Y, Perica K, Young JW. Langerhans dendritic cell vaccine bearing mRNA-encoded tumor antigens induces antimyeloma immunity after autotransplant. Blood Adv. 2022;6:1547–58.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Seymour C. mRNA vaccine/Pembrolizumab Reduces Metastasis/Death Risk in Melanoma. American Society of Clinical Oncology Annual Meeting (ASCO) 2023, June 19th 2023. Seymour C. mRNA vaccine/Pembrolizumab Reduces Metastasis/Death Risk in Melanoma. American Society of Clinical Oncology Annual Meeting (ASCO) 2023, June 19th 2023.
75.
Zurück zum Zitat Vik-Mo EO, Nyakas M, Mikkelsen BV, Moe MC, Due-Tonnesen P, Suso EM, Saeboe-Larssen S, Sandberg C, Brinchmann JE, Helseth E, et al. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother. 2013;62:1499–509.PubMedPubMedCentralCrossRef Vik-Mo EO, Nyakas M, Mikkelsen BV, Moe MC, Due-Tonnesen P, Suso EM, Saeboe-Larssen S, Sandberg C, Brinchmann JE, Helseth E, et al. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother. 2013;62:1499–509.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Mitchell DA, Batich KA, Gunn MD, Huang MN, Sanchez-Perez L, Nair SK, Congdon KL, Reap EA, Archer GE, Desjardins A, et al. Tetanus toxoid and CCL3 improve dendritic cell vaccines in mice and glioblastoma patients. Nature. 2015;519:366–9.PubMedPubMedCentralCrossRef Mitchell DA, Batich KA, Gunn MD, Huang MN, Sanchez-Perez L, Nair SK, Congdon KL, Reap EA, Archer GE, Desjardins A, et al. Tetanus toxoid and CCL3 improve dendritic cell vaccines in mice and glioblastoma patients. Nature. 2015;519:366–9.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Batich KA, Reap EA, Archer GE, Sanchez-Perez L, Nair SK, Schmittling RJ, Norberg P, Xie W, Herndon JE 2nd, Healy P, et al. Long-term survival in glioblastoma with cytomegalovirus pp65-targeted vaccination. Clin Cancer Res. 2017;23:1898–909.PubMedPubMedCentralCrossRef Batich KA, Reap EA, Archer GE, Sanchez-Perez L, Nair SK, Schmittling RJ, Norberg P, Xie W, Herndon JE 2nd, Healy P, et al. Long-term survival in glioblastoma with cytomegalovirus pp65-targeted vaccination. Clin Cancer Res. 2017;23:1898–909.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Zhu P, Li SY, Ding J, Fei Z, Sun SN, Zheng ZH, Wei D, Jiang J, Miao JL, Li SZ, et al. Combination immunotherapy of glioblastoma with dendritic cell cancer vaccines, anti-PD-1 and poly I:C. J Pharm Anal. 2023;13:616–24.PubMedPubMedCentralCrossRef Zhu P, Li SY, Ding J, Fei Z, Sun SN, Zheng ZH, Wei D, Jiang J, Miao JL, Li SZ, et al. Combination immunotherapy of glioblastoma with dendritic cell cancer vaccines, anti-PD-1 and poly I:C. J Pharm Anal. 2023;13:616–24.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Wang Y, Zhang L, Xu Z, Miao L, Huang L. mRNA vaccine with antigen-specific checkpoint blockade induces an enhanced immune response against established melanoma. Mol Ther. 2018;26:420–34.PubMedCrossRef Wang Y, Zhang L, Xu Z, Miao L, Huang L. mRNA vaccine with antigen-specific checkpoint blockade induces an enhanced immune response against established melanoma. Mol Ther. 2018;26:420–34.PubMedCrossRef
80.
Zurück zum Zitat Liu L, Wang Y, Miao L, Liu Q, Musetti S, Li J, Huang L. Combination immunotherapy of MUC1 mRNA nano-vaccine and CTLA-4 blockade effectively inhibits growth of triple negative breast cancer. Mol Ther. 2018;26:45–55.PubMedCrossRef Liu L, Wang Y, Miao L, Liu Q, Musetti S, Li J, Huang L. Combination immunotherapy of MUC1 mRNA nano-vaccine and CTLA-4 blockade effectively inhibits growth of triple negative breast cancer. Mol Ther. 2018;26:45–55.PubMedCrossRef
81.
Zurück zum Zitat Ma L, Hostetler A, Morgan DM, Maiorino L, Sulkaj I, Whittaker CA, Neeser A, Pires IS, Yousefpour P, Gregory J, et al. Vaccine-boosted CAR T crosstalk with host immunity to reject tumors with antigen heterogeneity. Cell. 2023;186:3148–65.PubMedPubMedCentralCrossRef Ma L, Hostetler A, Morgan DM, Maiorino L, Sulkaj I, Whittaker CA, Neeser A, Pires IS, Yousefpour P, Gregory J, et al. Vaccine-boosted CAR T crosstalk with host immunity to reject tumors with antigen heterogeneity. Cell. 2023;186:3148–65.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Amano T, Kajiwara K, Yoshikawa K, Morioka J, Nomura S, Fujisawa H, Kato S, Fujii M, Fukui M, Hinoda Y, Suzuki M. Antitumor effects of vaccination with dendritic cells transfected with modified receptor for hyaluronan-mediated motility mRNA in a mouse glioma model. J Neurosurg. 2007;106:638–45.PubMedCrossRef Amano T, Kajiwara K, Yoshikawa K, Morioka J, Nomura S, Fujisawa H, Kato S, Fujii M, Fukui M, Hinoda Y, Suzuki M. Antitumor effects of vaccination with dendritic cells transfected with modified receptor for hyaluronan-mediated motility mRNA in a mouse glioma model. J Neurosurg. 2007;106:638–45.PubMedCrossRef
83.
Zurück zum Zitat Saka M, Amano T, Kajiwara K, Yoshikawa K, Ideguchi M, Nomura S, Fujisawa H, Kato S, Fujii M, Ueno K, et al. Vaccine therapy with dendritic cells transfected with Il13ra2 mRNA for glioma in mice. J Neurosurg. 2010;113:270–9.PubMedCrossRef Saka M, Amano T, Kajiwara K, Yoshikawa K, Ideguchi M, Nomura S, Fujisawa H, Kato S, Fujii M, Ueno K, et al. Vaccine therapy with dendritic cells transfected with Il13ra2 mRNA for glioma in mice. J Neurosurg. 2010;113:270–9.PubMedCrossRef
84.
Zurück zum Zitat Ye L, Wang L, Yang J, Hu P, Zhang C, Tong S, Liu Z, Tian D. Identification of tumor antigens and immune subtypes in lower grade gliomas for mRNA vaccine development. J Transl Med. 2021;19:352.PubMedPubMedCentralCrossRef Ye L, Wang L, Yang J, Hu P, Zhang C, Tong S, Liu Z, Tian D. Identification of tumor antigens and immune subtypes in lower grade gliomas for mRNA vaccine development. J Transl Med. 2021;19:352.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Ye L, Wang L, Yang J, Hu P, Zhang C, Tong S, Liu Z, Tian D. Identification of tumor antigens and immune landscape in glioblastoma for mRNA vaccine development. Front Genet. 2021;12: 701065.PubMedPubMedCentralCrossRef Ye L, Wang L, Yang J, Hu P, Zhang C, Tong S, Liu Z, Tian D. Identification of tumor antigens and immune landscape in glioblastoma for mRNA vaccine development. Front Genet. 2021;12: 701065.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Wang ZL, Huang RY, Han B, Wu F, Sun ZY, Li GZ, Zhang W, Zhao Z, Liu X. Identification of tumor-associated antigens and immune subtypes of lower-grade glioma and glioblastoma for mRNA vaccine development. Chin Neurosurg J. 2022;8:34.PubMedPubMedCentralCrossRef Wang ZL, Huang RY, Han B, Wu F, Sun ZY, Li GZ, Zhang W, Zhao Z, Liu X. Identification of tumor-associated antigens and immune subtypes of lower-grade glioma and glioblastoma for mRNA vaccine development. Chin Neurosurg J. 2022;8:34.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Zhou Q, Yan X, Zhu H, Xin Z, Zhao J, Shen W, Yin W, Guo Y, Xu H, Zhao M, et al. Identification of three tumor antigens and immune subtypes for mRNA vaccine development in diffuse glioma. Theranostics. 2021;11:9775–90.PubMedPubMedCentralCrossRef Zhou Q, Yan X, Zhu H, Xin Z, Zhao J, Shen W, Yin W, Guo Y, Xu H, Zhao M, et al. Identification of three tumor antigens and immune subtypes for mRNA vaccine development in diffuse glioma. Theranostics. 2021;11:9775–90.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Ma S, Ba Y, Ji H, Wang F, Du J, Hu S. Recognition of tumor-associated antigens and immune subtypes in glioma for mRNA vaccine development. Front Immunol. 2021;12: 738435.PubMedPubMedCentralCrossRef Ma S, Ba Y, Ji H, Wang F, Du J, Hu S. Recognition of tumor-associated antigens and immune subtypes in glioma for mRNA vaccine development. Front Immunol. 2021;12: 738435.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Zhong H, Liu S, Cao F, Zhao Y, Zhou J, Tang F, Peng Z, Li Y, Xu S, Wang C, et al. Dissecting tumor antigens and immune subtypes of glioma to develop mRNA vaccine. Front Immunol. 2021;12: 709986.PubMedPubMedCentralCrossRef Zhong H, Liu S, Cao F, Zhao Y, Zhou J, Tang F, Peng Z, Li Y, Xu S, Wang C, et al. Dissecting tumor antigens and immune subtypes of glioma to develop mRNA vaccine. Front Immunol. 2021;12: 709986.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Rose M, Cardon T, Aboulouard S, Hajjaji N, Kobeissy F, Duhamel M, Fournier I, Salzet M. Surfaceome proteomic of glioblastoma revealed potential targets for immunotherapy. Front Immunol. 2021;12: 746168.PubMedPubMedCentralCrossRef Rose M, Cardon T, Aboulouard S, Hajjaji N, Kobeissy F, Duhamel M, Fournier I, Salzet M. Surfaceome proteomic of glioblastoma revealed potential targets for immunotherapy. Front Immunol. 2021;12: 746168.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Deng Z, Zhan P, Yang K, Liu L, Liu J, Gao W. Identification of personalized neoantigen-based vaccines and immune subtype characteristic analysis of glioblastoma based on abnormal alternative splicing. Am J Cancer Res. 2022;12:3581–600.PubMedPubMedCentral Deng Z, Zhan P, Yang K, Liu L, Liu J, Gao W. Identification of personalized neoantigen-based vaccines and immune subtype characteristic analysis of glioblastoma based on abnormal alternative splicing. Am J Cancer Res. 2022;12:3581–600.PubMedPubMedCentral
92.
Zurück zum Zitat Wu C, Qin C, Long W, Wang X, Xiao K, Liu Q. Tumor antigens and immune subtypes of glioblastoma: the fundamentals of mRNA vaccine and individualized immunotherapy development. J Big Data. 2022;9:92.PubMedPubMedCentralCrossRef Wu C, Qin C, Long W, Wang X, Xiao K, Liu Q. Tumor antigens and immune subtypes of glioblastoma: the fundamentals of mRNA vaccine and individualized immunotherapy development. J Big Data. 2022;9:92.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Chen Z, Wang X, Yan Z, Zhang M. Identification of tumor antigens and immune subtypes of glioma for mRNA vaccine development. Cancer Med. 2022;11:2711–26.PubMedPubMedCentralCrossRef Chen Z, Wang X, Yan Z, Zhang M. Identification of tumor antigens and immune subtypes of glioma for mRNA vaccine development. Cancer Med. 2022;11:2711–26.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Lin H, Wang K, Xiong Y, Zhou L, Yang Y, Chen S, Xu P, Zhou Y, Mao R, Lv G, et al. Identification of tumor antigens and immune subtypes of glioblastoma for mRNA vaccine development. Front Immunol. 2022;13: 773264.PubMedPubMedCentralCrossRef Lin H, Wang K, Xiong Y, Zhou L, Yang Y, Chen S, Xu P, Zhou Y, Mao R, Lv G, et al. Identification of tumor antigens and immune subtypes of glioblastoma for mRNA vaccine development. Front Immunol. 2022;13: 773264.PubMedPubMedCentralCrossRef
Metadaten
Titel
Advancing personalized medicine in brain cancer: exploring the role of mRNA vaccines
verfasst von
Feng Lin
Emma Z. Lin
Misa Anekoji
Thomas E. Ichim
Joyce Hu
Francesco M. Marincola
Lawrence D. Jones
Santosh Kesari
Shashaanka Ashili
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2023
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-023-04724-0

Weitere Artikel der Ausgabe 1/2023

Journal of Translational Medicine 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Schadet Ärger den Gefäßen?

14.05.2024 Arteriosklerose Nachrichten

In einer Studie aus New York wirkte sich Ärger kurzfristig deutlich negativ auf die Endothelfunktion gesunder Probanden aus. Möglicherweise hat dies Einfluss auf die kardiovaskuläre Gesundheit.

Intervallfasten zur Regeneration des Herzmuskels?

14.05.2024 Herzinfarkt Nachrichten

Die Nahrungsaufnahme auf wenige Stunden am Tag zu beschränken, hat möglicherweise einen günstigen Einfluss auf die Prognose nach akutem ST-Hebungsinfarkt. Darauf deutet eine Studie an der Uniklinik in Halle an der Saale hin.

Klimaschutz beginnt bei der Wahl des Inhalators

14.05.2024 Klimawandel Podcast

Auch kleine Entscheidungen im Alltag einer Praxis können einen großen Beitrag zum Klimaschutz leisten. Die neue Leitlinie zur "klimabewussten Verordnung von Inhalativa" geht mit gutem Beispiel voran, denn der Wechsel vom klimaschädlichen Dosieraerosol zum Pulverinhalator spart viele Tonnen CO2. Leitlinienautor PD Dr. Guido Schmiemann erklärt, warum nicht nur die Umwelt, sondern auch Patientinnen und Patienten davon profitieren.

Zeitschrift für Allgemeinmedizin, DEGAM

Typ-2-Diabetes und Depression folgen oft aufeinander

14.05.2024 Typ-2-Diabetes Nachrichten

Menschen mit Typ-2-Diabetes sind überdurchschnittlich gefährdet, in den nächsten Jahren auch noch eine Depression zu entwickeln – und umgekehrt. Besonders ausgeprägt ist die Wechselbeziehung laut GKV-Daten bei jüngeren Erwachsenen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.