Skip to main content

Main menu

  • Home
  • About
    • About CBM
    • Editorial Board
    • Announcement
  • Articles
    • Ahead of print
    • Current Issue
    • Archive
    • Collections
    • Cover Story
  • For Authors
    • Instructions for Authors
    • Resources
    • Submit a Manuscript
  • For Reviewers
    • Become a Reviewer
    • Instructions for Reviewers
    • Resources
    • Outstanding Reviewer
  • Subscription
  • Alerts
    • Email Alerts
    • RSS Feeds
    • Table of Contents
  • Contact us
  • Other Publications
    • cbm

User menu

  • My alerts

Search

  • Advanced search
Cancer Biology & Medicine
  • Other Publications
    • cbm
  • My alerts
Cancer Biology & Medicine

Advanced Search

 

  • Home
  • About
    • About CBM
    • Editorial Board
    • Announcement
  • Articles
    • Ahead of print
    • Current Issue
    • Archive
    • Collections
    • Cover Story
  • For Authors
    • Instructions for Authors
    • Resources
    • Submit a Manuscript
  • For Reviewers
    • Become a Reviewer
    • Instructions for Reviewers
    • Resources
    • Outstanding Reviewer
  • Subscription
  • Alerts
    • Email Alerts
    • RSS Feeds
    • Table of Contents
  • Contact us
  • Follow cbm on Twitter
  • Visit cbm on Facebook
OtherPerspective
Open Access

Immunological and metabolic optimization of tumor neoantigen vaccines

Xiafeng Wang, Zhangping Huang, Lin Peng, Shuoxi Xu, Jianfeng Huang and Ji Wang
Cancer Biology & Medicine September 2025, 20250445; DOI: https://doi.org/10.20892/j.issn.2095-3941.2025.0445
Xiafeng Wang
Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 440100, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Xiafeng Wang
  • For correspondence: wangxf58{at}mail.sysu.edu.cn huangjf85{at}mail.sysu.edu.cn wangj683{at}mail.sysu.edu.cn
Zhangping Huang
Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 440100, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lin Peng
Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 440100, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shuoxi Xu
Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 440100, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jianfeng Huang
Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 440100, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Jianfeng Huang
  • For correspondence: wangxf58{at}mail.sysu.edu.cn huangjf85{at}mail.sysu.edu.cn wangj683{at}mail.sysu.edu.cn
Ji Wang
Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 440100, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Ji Wang
  • For correspondence: wangxf58{at}mail.sysu.edu.cn huangjf85{at}mail.sysu.edu.cn wangj683{at}mail.sysu.edu.cn
  • Article
  • Info & Metrics
  • References
  • PDF
Loading

Tumor initiation and progression are highly intricate biological processes, and mutation-driven tumorigenesis is a primary underlying cause. Personalized cancer vaccines have been developed to exploit these specific mutations, particularly in the form of tumor neoantigens, to induce immune responses, particularly the activation of CD8+ T cells, which can attack malignant cells. Since tumor mutations result in protein sequence alterations distinct from those in normal tissues, therapies that precisely target these alterations could, in principle, confer effective tumor control while minimizing off-target effects.

Neoantigen-based therapies have garnered considerable scientific and clinical attention in recent years, as evidenced by more than 100 active or completed clinical trials. To date, a diverse array of neoantigen-targeting vaccine platforms have advanced to clinical trials, including peptide, DNA, RNA, and viral vector-based vaccines, each with their own advantages and limitations (Table 1). Notably, more than 50% of these trials have used synthetic long peptides (SLPs) in combination with adjuvants, typically poly-ICLC. Although these platforms can induce CD4+ T cell immune responses, their inherently low immunogenicity for CD8+ T cells remains a major challenge. Typically, neoantigen-specific CD8+ T cells are either undetectable or present at very low frequencies, accounting for 0.1% of the total CD8+ T cells in the peripheral blood. This proportion is far below the 10%–20% rate achievable with highly immunogenic vaccines such as the yellow fever and vaccinia vaccine1. Although the magnitude of CD8+ T cells is not the sole factor determining clinical outcomes, it is foundational to therapeutic efficacy. Sufficient CD8+ T cells are necessary for the effectiveness of other therapies aimed at reversing exhaustion and the immunosuppressive tumor microenvironment (TME).

View this table:
  • View inline
  • View popup
Table 1

Comparison of neoantigen vaccine platforms

This Perspective reviews current advances and ongoing challenges (Table 2) in the development of tumor neoantigen vaccines. Specifically, the efficacy of tumor vaccines is influenced by factors within the TME, such as metabolism, whose crucial role has been progressively elucidated in recent years; this aspect is also summarized and discussed. Finally, strategies for enhancing tumor neoantigen vaccines are described (Figure 1).

View this table:
  • View inline
  • View popup
Table 2

Obstacles to therapeutic neoantigen vaccine development

Figure 1
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1

Strategies to overcome the low immunogenicity of neoantigen vaccines focus on 5 key areas: antigen design, prediction methods, adjuvant development, delivery system optimization, and metabolic regulation. 1) Target trunk mutations, or shared canonical or non-canonical antigens, optimize antigen length and screen neoantigens to induce epitope spreading. 2) Whole exome sequencing (WES), ribosome profiling (RiboSeq), immunopeptidomics, and mass spectrometry-based detection methods are integrated to screen for preferable neoantigens. 3) Novel multifunctional adjuvants or combinations of existing adjuvants are developed. 4) The composition of the delivery system is optimized, targeted molecules and multi-level targeting are explored, and new delivery materials with high safety are developed. 5) Metabolic regulation promotes antigen presentation, and reverses tumor suppressor environments and T cell exhaustion. (Created with BioRender.com).

Sources of neoantigens

Tumor neoantigens originate primarily from mutation-dependent tumor antigens. Because of tumors’ intrinsic heterogeneity, substantial variability exists in neoantigen expression, thus leading to inconsistent neoantigen presentation. Therefore, the identification of so-called “trunk mutations” that prominently control tumor proliferation has become critical. However, a major challenge arises from the high similarity between these trunk mutations and their corresponding wild-type sequences. During T cell development, neoantigen-specific T cell receptors (TCRs) often exhibit inherently low affinity for such neoantigens with minimally altered peptide sequences due to negative selection. Even when high-affinity TCRs do exist, the absolute abundance of such T cells remains extremely low2. This mechanistic insight helps explain why neoantigen-specific CD8+ T cell responses are often undetected or are observed at only very low levels in patients, although the possible induction of immune responses against such antigens may occur.

Beyond mutation-driven antigens, an alternative source of tumor neoantigens comes from mutation-independent mechanisms involving antigens derived from non-coding genomic regions. Their generation can occur through various pathways, such as the translation of circular RNAs, initiation from selective non-AUG start codons, and ribosomal frameshift errors3. Because some of these antigens exhibit substantial tumor specificity, they might potentially provide a rich source for novel immunotherapeutic targets. Notably, a recent study has revealed that such cryptic neoantigens can elicit effective cytotoxic responses against pancreatic cancer organoids4. However, notable challenges lie in the heterogeneous expression of these non-canonical antigen epitopes across tumor clones, coupled with their dispensability for tumor cell survival. These characteristics render them inherently susceptible to immune editing and facilitate vaccine escape in the absence of robust epitope spreading. Consequently, although targeting this “dark proteome” offers compelling opportunities, its overall clinical utility remains to be rigorously validated.

Neoantigen identification and design

The advent of next-generation sequencing technologies has made large-scale genetic analysis of individual tumors and patients feasible. Coupled with advances in whole-exome sequencing and RNA sequencing, a wide range of bioinformatics pipelines and MHC-peptide binding prediction algorithms have been developed for the prospective identification of tumor neoantigens. Some predictive tools integrate multiple considerations, such as antigen cleavage, antigen transport, cellular presentation, and TCR recognition. In recent years, new artificial intelligence technologies have also been applied to enhance accuracy.

Despite these technical advancements, the practical effectiveness of these bioinformatics approaches remains limited. In some cases, less than 2.7% of predicted neoantigens are ultimately recognized by patient-derived T cells5. Similarly, the Tumor Neoantigen Selection Alliance (TESLA) global consortium has reported that, even with a comprehensive pipeline—starting with MHC binding prediction and followed by validation using patient-matched T cells—only approximately 6% of the predicted neoantigens elicit T cell recognition6. These findings illustrate the limitations of current methods in tumor neoantigen prediction and highlight the urgent need for improved algorithms and validation frameworks.

The accuracy of neoantigen prediction depends not only on the sophistication of the computational algorithms but also critically on intrinsic biological factors pertaining to the tumor cells, antigen-presenting cells, and antigen processing preferences. Therefore, complementary approaches enabling direct integration of mass spectrometry immunopeptidomics data might provide better results. For instance, the NeoDis computational framework integrates data from genomic, transcriptomic, and immunopeptidomic sources to accurately identify neoantigens. Among the 66 HLA-I neoantigenic peptides for T cell screening assays, 11 were found to be immunogenic7. Nevertheless, these direct detection methods face their own technical challenges, including immunopeptidome dilution due to non-tumor cells infiltration, the requirement for large quantities of tumor tissue or cells, and frequently low MHC class I expression on tumor cells. These challenges might be potentially overcome by strategies such as cultivating tumor organoids to enrich for homologous peptides or using methods to enhance MHC class I expression8.

Adjuvants for tumor neoantigen vaccines

Because of the inherently low immunogenicity of tumor neoantigens, administration of the antigen alone is generally insufficient to provoke robust immune response, thereby necessitating a combination of adjuvants. Certain vaccine platforms, such as plasmid DNA and mRNA-based vaccines, can partially function as self-adjuvants and boost the immune response, whereas SLP neoantigen vaccines require the addition of adjuvants to achieve effective immunogenicity. An adjuvant is an additive to vaccines that enhances the immunogenicity of antigens, common adjuvants include ligands of pattern recognition receptors, cytokines, and immune checkpoint inhibitors.

In recent years, extensive researches have focused on the roles of pattern recognition receptors, such as Toll-like receptors, and the cGAS-STING pathway. Ligands targeting these receptors are typically small molecules with well-defined mechanisms of action and favorable drug potential, and therefore highly promising for clinical translation. SLP neoantigen vaccines critically require dendritic cell (DC)-mediated antigen cross-presentation to effectively elicit CD8+ T cell immune responses. However, in the absence of adequate adjuvant support, mobilizing DCs to achieve potent T cell activation and expansion remains difficult. Inadequate DC activation can even lead to undesirable immune tolerance, thereby diminishing CD8+ T cell responses.

To circumvent this limitation, neopeptides are frequently co-administered with adjuvants. Nonetheless, the enhanced immunogenicity achieved through adjuvant addition is often modest and falls short of the robust and durable immune responses required for effective tumor control. This shortcoming arises from a predominant focus on the adjuvant’s immunostimulatory function, without adequately addressing the inherent inefficiency of the cross-presentation process itself. Crucially, because the cross-presentation process of exogenous antigens by DCs is inherently intricate and often inefficient, effectively presenting neoantigen epitopes is difficult. Therefore, to stimulate high-level specific CD8+ T cell immune responses to an extent comparable level to those induced by viral vaccines, not only adjuvant activity but also antigen cross-presentation by DCs must be optimized9.

Our recent study has offered new insights to address this important issue. We have developed a new series of STING agonists, collectively termed STING Agonist-Based ER targeting molecules (SABER), to not only potently activate DCs but also critically target antigens directly to the endoplasmic reticulum (ER). These characteristics depend on SABER’s high binding affinity toward the STING protein, which resides within the ER, a critical hub for antigen processing. Our findings have demonstrated that after neoantigens delivered to the ER, they can form a localized “micro-reactor” that effectively promotes antigen cleavage and transport. This mechanism subsequently augments the specific CD8+ T cell immune response. Compared with conventional adjuvanted SLPs, SABER-based neoantigen vaccines induce tenfold greater CD8+ T cell induction. Our findings strongly suggest that the “last mile” of antigen delivery, specifically from the cytoplasm to the ER, is crucial for optimizing antigen cross-presentation10.

Delivery system for tumor neoantigen vaccines

Beyond the essential role of adjuvants, delivery systems are equally important for enhancing the T-cell immune response to neoantigens. In recent years, mRNA has attracted considerable attention, because of its rapid production and broad therapeutic scope across a diverse array of diseases. Despite its advantages, mRNA’s inherent instability and negative charge present major obstacles to efficient cellular uptake and membrane translocation. Consequently, encapsulation and transport via lipid nanoparticles (LNPs) have become imperative. LNPs not only protect mRNA against rapid degradation in vivo but also notably target antigen-presenting cells, which have a propensity for nanoparticle phagocytosis. Moreover, by precisely modulating the lipid composition and physicochemical properties of LNPs, organ and tissue-specific targeting can be optimized11. For instance, our research group has successfully used biomimetic nanoparticles to encapsulate 2′,3′-cyclic guanosine monophosphate–adenosine monophosphate, the natural ligand for the STING protein, thereby eliciting effective antigen-specific CD8+ T cell immunity in the lungs12. Additionally, the use of polymeric materials or inorganic nanoparticles has also been demonstrated to effectively enhance CD8+ T cell induction. However, a key impediment is the safety profiles of these delivery systems, particularly concerns regarding off-target organ accumulation and the potential induction of undesirable nanoparticle-triggered antibody or inflammatory responses. Therefore, we developed an in vivo electroporation system as an alternative delivery system13,14.

Metabolic effects on tumor neoantigen vaccines

Despite substantial progress in tumor neoantigen vaccines, some post-injection challenges must be addressed to achieve meaningful clinical outcomes. These obstacles include tumor immune evasion, immunosuppressive TME, and T cell exhaustion. Recent evidence has highlighted the critical roles of metabolic changes in shaping the immune landscape of neoantigen-based vaccines. These metabolic alterations affect key processes, such as antigen presentation in tumor cells and DCs, as well as the overall state of T cells.

The immunopeptidome is highly plastic and is modulated by cellular metabolic activity. For example, altering cellular metabolism via inhibiting the mammalian target of rapamycin (mTOR) results in dynamic changes in the composition of the immunopeptidome and leads to high abundance of MHC class I-presented peptides15. Furthermore, autophagy, a key metabolic process in the degradation and recycling of various cytoplasmic components, enhances antigen processing in antigen-presenting cells. Notably, active autophagy facilitates the delivery of cytosolic antigens to MHC class II loading compartments, thereby promoting CD4+ T cell responses to intracellular antigens16.

Metabolic shifts in tumors frequently result in a highly immunosuppressive TME. Accumulated lactate, a byproduct of aerobic glycolysis, inhibits DC functions which are essential for effective T cell priming. Moreover, lactate-driven acidosis destabilizes peptide–MHC class I complexes and impairs antigen presentation, particularly via the cross-presentation pathway17. Cross-presentation is an energy-intensive process sensitive to the metabolic fitness of DCs. Recent studies have demonstrated that optimal cross-presentation requires finely tuned metabolic programs, including glycolysis, oxidative phosphorylation, and fatty acid metabolism18. Nevertheless, aberrant lipid accumulation in tumor-infiltrating DCs impairs their ability to present antigens, whereas pharmacological inhibition of fatty acid synthesis has been shown to substantially enhance the efficacy of cancer vaccines19.

In parallel, T cell metabolism, which underpins the effector function of CD8+ T cells responding to neoantigen vaccines, is also governed by nutrient availability within the TME. Notably, recent findings have revealed that lactate, rather than being merely immunosuppressive, also serves as a bioenergetic and biosynthetic fuel that supports CD8+ T cell activity in certain metabolic contexts20.

Conclusion and future prospects

The identification and subsequent utilization of tumor neoantigens have led to paradigm shift in the understanding of malignancies and are highly promising avenue for cancer immunotherapy. Nevertheless, despite these bright prospects, substantial challenges persist. A major obstacle is that the high-level similarity between neoantigens and their wild-type counterparts often renders neoantigens poor immunogenicity and makes them concealed from the immune system. Consequently, stimulating comparably robust immune responses to those elicited by potent foreign substances, such as viruses, is inherently difficult.

Although bioinformatics approaches based on whole-exome sequencing and RNA sequencing have rapidly advanced, their predictive accuracy requires further refinement. Future developments are expected to rely on the seamless integration of machine learning and big data analytics to discern broader spectrum of epitope features, alongside the application of immunopeptidomics to more precisely identify antigen sequences. Moreover, ongoing advancements in mass spectrometry and organoid culture technologies are expected to facilitate the discovery of novel neoantigen epitopes.

Crucially, antigen optimization alone is insufficient, because the inherently low immunogenicity of neoantigens limits the host’s ability to mount robust, antigen-specific CD8+ T cell responses. Consequently, optimization of adjuvants and delivery systems is imperative. Although many adjuvants and delivery systems have been developed to date, their overall efficacy remains constrained. The immunostimulatory bias inherent to various adjuvants requires evaluation; moreover, most delivery systems function primarily at the organ or tissue level, and therefore fall short of achieving precise subcellular antigen delivery. Therefore, future efforts must focus on refining adjuvant structures and identifying more efficacious adjuvant combinations, also improving delivery systems that significantly enhance cross-presentation efficiency.

Furthermore, to overcome the overarching challenges inherent in tumor treatment, post-vaccination bottlenecks must be addressed. Metabolic reprogramming in tumor cells and the TME establishes a hostile immunological niche that adversely affects nearly every step involved in neoantigen vaccine efficacy, from antigen generation to cross-presentation and T cell activation. To fully harness the therapeutic potential of neoantigen-based cancer vaccines, these metabolic barriers must be overcome through rational combinatorial strategies that modulate both tumor-intrinsic and microenvironmental metabolic constraints.

In summary, substantial progress has been made in the identification of tumor neoantigens and the development of novel technologies to induce effective neoantigen-specific CD8+ T cell responses. However, multiple impediments persist, particularly regarding vaccine delivery and anti-tumor efficacy. Addressing these limitations through innovative and integrated strategies will be essential to fully achieve the clinical potential of neoantigen vaccines and deliver meaningful therapeutic benefits to patients with cancer.

Conflict of interest statement

No potential conflicts of interest are disclosed.

Author contributions

Conceived and designed the analysis: Jianfeng Huang, Ji Wang.

Collected the data: Xiafeng Wang, Zhangping Huang, Lin Peng, Shuoxi Xu.

Wrote the paper: Xiafeng Wang.

Reviewed and revised the paper: Jianfeng Huang, Ji Wang.

  • Received August 2, 2025.
  • Accepted September 8, 2025.
  • Copyright: © 2025, The Authors

This work is licensed under the Creative Commons Attribution-NonCommercial 4.0 International License.

References

  1. 1.↵
    1. Katsikis PD,
    2. Ishii KJ,
    3. Schliehe C.
    Challenges in developing personalized neoantigen cancer vaccines. Nat Rev Immunol. 2024; 24: 213–27.
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. Stone JD,
    2. Harris DT,
    3. Kranz DM.
    TCR affinity for p/MHC formed by tumor antigens that are self-proteins: impact on efficacy and toxicity. Curr Opin Immunol. 2015; 33: 16–22.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Huang D,
    2. Zhu X,
    3. Ye S,
    4. Zhang J,
    5. Liao J,
    6. Zhang N, et al.
    Tumour circular RNAs elicit anti-tumour immunity by encoding cryptic peptides. Nature. 2024; 625: 593–602.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Ely ZA,
    2. Kulstad ZJ,
    3. Gunaydin G,
    4. Addepalli S,
    5. Verzani EK,
    6. Casarrubios M, et al.
    Pancreatic cancer-restricted cryptic antigens are targets for T cell recognition. Science. 2025; 388: eadk3487.
  5. 5.↵
    1. Bjerregaard A-M,
    2. Nielsen M,
    3. Jurtz V,
    4. Barra CM,
    5. Hadrup SR,
    6. Szallasi Z, et al.
    An analysis of natural T cell responses to predicted tumor neoepitopes. Front Immunol. 2017; 8: 1566.
    OpenUrlCrossRefPubMed
  6. 6.↵
    1. Wells DK,
    2. van Buuren MM,
    3. Dang KK,
    4. Hubbard-Lucey VM,
    5. Sheehan KCF,
    6. Campbell KM, et al.
    Key parameters of tumor epitope immunogenicity revealed through a consortium approach improve neoantigen prediction. Cell. 2020; 183: 818–34.e13.
    OpenUrlCrossRefPubMed
  7. 7.↵
    1. Huber F,
    2. Arnaud M,
    3. Stevenson BJ,
    4. Michaux J,
    5. Benedetti F,
    6. Thevenet J, et al.
    A comprehensive proteogenomic pipeline for neoantigen discovery to advance personalized cancer immunotherapy. Nat Biotechnol. 2025; 43: 1360–72.
    OpenUrlPubMed
  8. 8.↵
    1. Demmers LC,
    2. Kretzschmar K,
    3. Van Hoeck A,
    4. Bar-Epraïm YE,
    5. van den Toorn HWP,
    6. Koomen M, et al.
    Single-cell derived tumor organoids display diversity in HLA class I peptide presentation. Nat Commun. 2020; 11: 5338.
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. Wang J,
    2. Xie X,
    3. Jiang S,
    4. Lu L.
    Immunoengineered adjuvants for universal vaccines against respiratory viruses. Fundam Res. 2021; 1: 189–92.
    OpenUrlPubMed
  10. 10.↵
    1. Wang X,
    2. Huang Z,
    3. Xing L,
    4. Shang L,
    5. Jiang J,
    6. Deng C, et al.
    STING agonist-based ER-targeting molecules boost antigen cross-presentation. Nature. 2025; 641: 202–10.
    OpenUrlPubMed
  11. 11.↵
    1. Kon E,
    2. Ad-El N,
    3. Hazan-Halevy I,
    4. Stotsky-Oterin L,
    5. Peer D.
    Targeting cancer with mRNA-lipid nanoparticles: key considerations and future prospects. Nat Rev Clin Oncol. 2023; 20: 739–54.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. Wang J,
    2. Li P,
    3. Yu Y,
    4. Fu Y,
    5. Jiang H,
    6. Lu M, et al.
    Pulmonary surfactant-biomimetic nanoparticles potentiate heterosubtypic influenza immunity. Science. 2020; 367: eaau0810.
  13. 13.↵
    1. Li Y,
    2. Mo J,
    3. Liu J,
    4. Liang Y,
    5. Deng C,
    6. Huang Z, et al.
    A micro-electroporation/electrophoresis-based vaccine screening system reveals the impact of vaccination orders on cross-protective immunity. iScience. 2023; 26: 108086.
  14. 14.↵
    1. Liu X,
    2. Jiang J,
    3. Liu J,
    4. Yang H,
    5. Huang Z,
    6. Deng C, et al.
    Nanoneedle array-electroporation facilitates intranuclear ribonucleoprotein delivery and high throughput gene editing. Adv Healthc Mater. 2024; 13: e2400645.
  15. 15.↵
    1. Caron E,
    2. Vincent K,
    3. Fortier M-H,
    4. Laverdure J-P,
    5. Bramoullé A,
    6. Hardy M-P, et al.
    The MHC I immunopeptidome conveys to the cell surface an integrative view of cellular regulation. Mol Syst Biol. 2011; 7: 533.
    OpenUrlAbstract/FREE Full Text
  16. 16.↵
    1. Valečka J,
    2. Almeida CR,
    3. Su B,
    4. Pierre P,
    5. Gatti E.
    Autophagy and MHC-restricted antigen presentation. Mol Immunol. 2018; 99: 163–70.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. Llibre A,
    2. Kucuk S,
    3. Gope A,
    4. Certo M,
    5. Mauro C.
    Lactate: a key regulator of the immune response. Immunity. 2025; 58: 535–54.
    OpenUrlCrossRefPubMed
  18. 18.↵
    1. Giovanelli P,
    2. Sandoval TA,
    3. Cubillos-Ruiz JR.
    Dendritic cell metabolism and function in tumors. Trends Immunol. 2019; 40: 699–718.
    OpenUrlCrossRefPubMed
  19. 19.↵
    1. Herber DL,
    2. Cao W,
    3. Nefedova Y,
    4. Novitskiy SV,
    5. Nagaraj S,
    6. Tyurin VA, et al.
    Lipid accumulation and dendritic cell dysfunction in cancer. Nat Med. 2010; 16: 880–6.
    OpenUrlCrossRefPubMed
  20. 20.↵
    1. Kaymak I,
    2. Luda KM,
    3. Duimstra LR,
    4. Ma EH,
    5. Longo J,
    6. Dahabieh MS, et al.
    Carbon source availability drives nutrient utilization in CD8+ T cells. Cell Metab. 2022; 34: 1298–311.e6.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Cancer Biology & Medicine: 22 (10)
Cancer Biology & Medicine
Vol. 22, Issue 10
15 Oct 2025
  • Table of Contents
  • Index by author
Print
Download PDF
Email Article

Thank you for your interest in spreading the word on Cancer Biology & Medicine.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Immunological and metabolic optimization of tumor neoantigen vaccines
(Your Name) has sent you a message from Cancer Biology & Medicine
(Your Name) thought you would like to see the Cancer Biology & Medicine web site.
Citation Tools
Immunological and metabolic optimization of tumor neoantigen vaccines
Xiafeng Wang, Zhangping Huang, Lin Peng, Shuoxi Xu, Jianfeng Huang, Ji Wang
Cancer Biology & Medicine Sep 2025, 20250445; DOI: 10.20892/j.issn.2095-3941.2025.0445

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Immunological and metabolic optimization of tumor neoantigen vaccines
Xiafeng Wang, Zhangping Huang, Lin Peng, Shuoxi Xu, Jianfeng Huang, Ji Wang
Cancer Biology & Medicine Sep 2025, 20250445; DOI: 10.20892/j.issn.2095-3941.2025.0445
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Sources of neoantigens
    • Neoantigen identification and design
    • Adjuvants for tumor neoantigen vaccines
    • Delivery system for tumor neoantigen vaccines
    • Metabolic effects on tumor neoantigen vaccines
    • Conclusion and future prospects
    • Conflict of interest statement
    • Author contributions
    • References
  • Info & Metrics
  • References
  • PDF

Related Articles

  • No related articles found.
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • Chromatin relaxation dynamics and histone PTMs in the early DNA damage response
  • Modeling cervical cancer elimination: a pathway to inform policy decisions
  • Strengthening human papillomavirus vaccination programs through multi-country peer learning: lessons from the CHIC initiative
Show more Perspective

Similar Articles

Navigate

  • Home
  • Current Issue

More Information

  • About CBM
  • About CACA
  • About TMUCIH
  • Editorial Board
  • Subscription

For Authors

  • Instructions for authors
  • Journal Policies
  • Submit a Manuscript

Journal Services

  • Email Alerts
  • Facebook
  • RSS Feeds
  • Twitter

 

© 2025 Cancer Biology & Medicine

Powered by HighWire