Skip to main content

EDITORIAL article

Front. Immunol., 23 April 2024
Sec. Cancer Immunity and Immunotherapy
This article is part of the Research Topic Targeting Nucleotide Metabolism for Enhancing Antitumor Immunity View all 8 articles

Editorial: Targeting nucleotide metabolism for enhancing antitumor immunity

Jun WuJun Wu1Yu RongYu Rong2Tian LiTian Li3Cornelia M. WilsonCornelia M. Wilson4Yazhou HeYazhou He5Danqian Chen*Danqian Chen6*Jin Han*Jin Han7*Xingmei Zhang*Xingmei Zhang8*
  • 1Division for Research, Shandong College of Traditional Chinese Medicine, Yantai, China
  • 2Department of Thoracic Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
  • 3School of Basic Medicine, Fourth Military Medical University, Xi'an, China
  • 4School of Psychology and Life Sciences, Life Sciences Industry Liaison Lab, Canterbury Christ Church University, Sandwich, United Kingdom
  • 5West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
  • 6School of Life Sciences, Northwest University, Xi’an, China
  • 7Department of Nephrology, Chang’an District Hospital, Xi’an, China
  • 8College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China

Neoplasm remain the main killer worldwide (15).Tumor microenvironment (TME) is a special environment for tumor survival, which has the characteristics of acidity, lack of nutrition and immunosuppression (6). Among them, the competition and cooperation of tumor, fibroblast, endothelial cell, immune cell and other clonal populations play a key role in cell proliferation, tumor metastasis and the development of therapeutic resistance (7). Nucleotides are major components of genetic material and are essential for DNA and RNA biosynthesis, cell signaling, enzyme regulation, and metabolism (8). Malignancies exhibit rapid growth and rely more on de novo synthesis than external sources (9), with very high nucleotide requirements. Immune cells in the TME, especially effector T cells, require rapid expansion when the immune response is activated, as well as lots of nucleotides in order to replicate (10). The regulation of TME and immune cell function is strongly dependent on nucleotide metabolism, according to a growing body of research (6, 11).

LncRNAs (long non-coding RNAs) are a class of RNAs longer than 200 nucleotides that play a variety of roles in cell differentiation, cell cycle regulation, and epigenetic regulation. Ye et al. explored the nucleotide molecular mechanism of compound Fushen injection in the treatment of lung adenocarcinoma and found that mir-15b could enhance the proliferation and migration of lung adenocarcinoma by targeting BCL2, and increase the expression of BIRC5 could improve the radiotherapy efficacy of LUAD cells. Copper plays an important role in various processes related to nucleotide metabolism, especially ribonucleotide reductase is a key enzyme affected. It is an important electron carrier for ribonucleotide reductase and is essential for maintaining appropriate nucleotide levels and ensuring accurate DNA replication and repair. Luo et al. found that IGFBP2, a biomarker associated with copper metabolism, is not only a glioma tumor promoter that promotes disease progression, but also affects immunotherapy response. Zhao et al. used TCGA and XENA-TCGA databases to find that lncRNA SLC25A25-AS1 is highly expressed in many cancers. Further analysis of clinical samples by prostate cancer (PC) showed that high expression of SLC25A25-AS1 was associated with T stage, clinical stage, Gleason score (GS) and poor prognosis. SLC25A25-AS1 is associated with an infiltration of CD8 T cells, IDCs (interstitial dendritic cells), macrophages, and other immune cells in the PC immune microenvironment. Targeting SLC25A25-AS1 in conjunction with PC immune microenvironment provides a potential therapeutic target for the treatment of PC.

Zhang et al. found that lung adenocarcinomatosis with lower types of immune cell infiltration were more responsive to immune checkpoint (LAG3, PD-L1, IDO1) inhibition and targeted drugs (JNK inhibitor VIII, BI-D1870, RO-3306, etc.). Exonuclease 1 (EXO1) genes encodes a protein with 5′ to 3′ exonuclease activity as well as RNase H activity. It has both an endonuclease domain and an exonuclease domain. Capable of acting on nucleotide excision repair (NER) and mismatch repair (MMR) pathways (12). In addition, it was also found that EXO1, one of the key module genes, is related to a variety of immune cells in the immune environment and significantly affects anti-tumor immunity (Zhang et al.). Autophagy is an intracellular lysosomal degradation pathway that reconnects cellular metabolism. Metabolites produced by this process can be reused for core biosynthetic processes or energy production, including nucleotides (13). Another study used bioinformatics methods to identify immune heterogeneity in hepatocellular carcinoma (HCC) from public databases and analyze genes associated with autophagy. In malignant tumors, autophagy-related pathways were more prevalent, and group C1 was associated with a better survival rate, enhanced immune infiltration, and a greater response to immunotherapy (Liu et al.).

Zhang et al. also found that immune cells in the tumor microenvironment were closely related to the effect of tumor immunotherapy and chemotherapy. Patients with a high degree of infiltration of antigen presenting B cells, tumor killer natural killer cells, CD8+T cells and other cells in the immune microenvironment have enhanced immune status, which is more suitable for immunotherapy and chemotherapy. Copper plays an important role in various processes related to nucleotide metabolism, especially ribonucleotide reductase is a key enzyme affected. It is an important electron carrier for ribonucleotide reductase and is essential for maintaining appropriate nucleotide levels and ensuring accurate DNA replication and repair. Luo et al. found that IGFBP2, a biomarker associated with copper metabolism, is not only a glioma tumor promoter that promotes disease progression, but also affects immunotherapy response.

Studies have shown that aberrant nucleotide metabolism not only accelerates tumor development, but also suppresses normal immune responses (1417). Studies have demonstrated that targeting nucleotide metabolism can increase anti-tumor immune responses through multiple pathways (8, 9). Such as activating the host immune system by maintaining the concentration of several important metabolites, such as adenosine and ATP; The imbalance of purine/pyrimidine ratio leads to an increase in tumor mutation burden (TMB), which leads to an increase in surface neoantigen expression, thus enhancing immunotherapy. According to Keshet and colleagues, inhibiting purine synthesis increases the ratio of pyrimidines to purines, improves immunoproteasome expression, and improves anti-PD-1 response in autologous primary T cells (18). Thus, targeting nucleotide metabolism in the TME holds great promise for preventing and treating cancer.

Methotrexate was the earliest antimetabolite used to treat cancer. In cancer chemotherapy, more than 20 approved nucleotides and nucleotide analogues are used. These drugs account for nearly 20% of all cancer therapies (8). It is not well understood how other cell types influence metabolic cancer therapy in the context of the tumor microenvironment. Hyrossova et al. and Youssef et al. in the manuscript described the effects of interventions including targeting nucleotide pathways on the cellular components of the tumor microenvironment (9, 10).

Drugs that target the adenosine pathway are shown to transform an immunosuppressive microenvironment into a more immunopermissive one and reduce metastasis and resistance to treatment (14). The review by Sek (19), Xia (20) et al. provides a detailed description of the role of adenosine and its receptor signaling in tumors and immune cells. CD73, CD39 and CD38 expressed on tumor cells and immune cells catabolize extracellular ATP into adenosine (ADO) and accumulate in TME, thus mediating the suppression of anti-tumor immunity through the activation of ADO receptors(A1R、A2AR、A2BR、A3R). The α subunits of G proteins are classified into 4 major families (Gαs, Gαi/o, Gq/11, Gα12/13), which are the main signaling pathways of the A2AR, A2BR and A1R, A3R receptors, and bind to the Gαs and Gαi/o family of Gα proteins, respectively. A2AR and A2BR conjugated to Gαs can promote cyclic adenosine monophosphase (cAMP) accumulation and play a dominant role in suppressing anti-tumor immune cell responses. In contrast, A1R and A3R conjugated to Gαi/o subunits inhibited the production of adenylyl cyclase and cAMP (19). Targeting the CD39/CD73/A2AR signaling pathway shows powerful anti-tumor efficacy, which predicts its wide application in the clinical field (21). In addition, Franco et al. believe that A2AR is not only the main target of immunotherapy methods, A2BR seems to be more promising as a target for chemotherapy intervention (or even radiotherapy) (22). It is well known that many malignant tumors overexpress purinergic receptors, which is associated with tumor cell proliferation, metastasis, therapeutic resistance and poor prognoses. Jia et al. and Aria et al. described in the review that P2X7R inhibition has a powerful effect in reversing therapeutic resistance in various types of cancer therapy (23, 24).

A tumor microenvironment, including immune cells, fibroblasts, endothelial cells, and fibroblasts, is involved in the development of radioresistance, among other factors. A single metabolic target is not enough to overcome tumor cell radioresistance due to metabolic heterogeneity. According to Yu et al., related nucleotide synthesis enzymes (such as TS/PPAT/IMPDH) have been shown to be radiation sensitizing (25).

A salvage pathway has nevertheless been demonstrated to be associated with cancer development, even though immune cells and cancer cells prefer de novo nucleotide synthesis (26). Walter et al. proposed targeting the pyrimidine rescue pathway in DNA replication alone or in combination with inhibitors of de novo pyrimidine synthesis to overcome the limitations of commonly used antimetabolites in various preclinical cancer models and clinical trials (20). For the purpose of overcoming the limitations of commonly used antimetabolites in various preclinical cancer models clinical trials. Walter et al. proposed targeting the pyrimidine rescue pathway in DNA replication alone or in combination with inhibitors of de novo pyrimidine synthesis.

Overall, given the particularly high proliferation rate and active nucleotide synthesis of cancer cells, interfering with nucleotide metabolism has been proposed as a promising strategy for antitumor therapy. The complexity and diversity of interactions between tumor cells and immune cells in relation to their intracellular and extracellular nucleotide metabolisms necessitates more personalized approaches.

Author contributions

JW: Writing – original draft. YR: Writing – original draft. TL: Writing – original draft. CW: Writing – original draft. YH: Writing – review & editing. DC: Writing – review & editing. JH: Writing – review & editing. XZ: Writing – review & editing.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Li T, Yang Z, Jiang S, Di W, Ma Z, Hu W, et al. Melatonin: does it have utility in the treatment of haematological neoplasms? Br J Pharmacol. (2018) 175:3251–62. doi: 10.1111/bph.13966

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Ma Z, Fan C, Yang Y, Di S, Hu W, Li T, et al. Thapsigargin sensitizes human esophageal cancer to TRAIL-induced apoptosis via AMPK activation. Sci Rep. (2016) 6:35196. doi: 10.1038/srep35196

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Sun M, Liu X, Xia L, Chen Y, Kuang L, Gu X, et al. A nine-lncRNA signature predicts distant relapse-free survival of HER2-negative breast cancer patients receiving taxane and anthracycline-based neoadjuvant chemotherapy. Biochem Pharmacol. (2021) 189:114285. doi: 10.1016/j.bcp.2020.114285

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Yang Z, Jiang S, Lu C, Ji T, Yang W, Li T, et al. SOX11: friend or foe in tumor prevention and carcinogenesis? Ther Adv Med Oncol. (2019) 11:1758835919853449. doi: 10.1177/1758835919853449

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Li T, Qiao T. Unraveling tumor microenvironment of small-cell lung cancer: Implications for immunotherapy. Semin Cancer Biol. (2022) 86:117–25. doi: 10.1016/j.semcancer.2022.09.005

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Elia I, Haigis MC. Metabolites and the tumour microenvironment: from cellular mechanisms to systemic metabolism. Nat Metab. (2021) 3:21–32. doi: 10.1038/s42255-020-00317-z

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Bhattacharya S, Mohanty A, Achuthan S, Kotnala S, Jolly MK, Kulkarni P, et al. Group behavior and emergence of cancer drug resistance. Trends Cancer. (2021) 7:323–34. doi: 10.1016/j.trecan.2021.01.009

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Wu H-L, Gong Y, Ji P, Xie Y-F, Jiang Y-Z, Liu G-Y. Targeting nucleotide metabolism: a promising approach to enhance cancer immunotherapy. J Hematol Oncol. (2022) 15:45. doi: 10.1186/s13045-022-01263-x

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Youssef R, Maniar R, Khan J, Mesa H. Metabolic interplay in the tumor microenvironment: implications for immune function and anticancer response. Curr Issues Mol Biol. (2023) 45:9753–67. doi: 10.3390/cimb45120609

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Hyroššová P, Milošević M, Škoda J, Vachtenheim J, Rohlena J, Rohlenová K. Effects of metabolic cancer therapy on tumor microenvironment. Front Oncol. (2022) 12:1046630. doi: 10.3389/fonc.2022.1046630

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Shi R, Tang YQ, Miao H. Metabolism in tumor microenvironment: Implications for cancer immunotherapy. MedComm. (2020) 1:47–68. doi: 10.1002/mco2.6

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Ferreira AMC, Altemani JMC, Macedo LT, Lourenço GJ, Lima CSP. Genetic variability in cisplatin metabolic pathways and outcome of locally advanced head and neck squamous cell carcinoma patients. Sci Rep. (2023) 13:16762. doi: 10.1038/s41598-023-44040-7

PubMed Abstract | CrossRef Full Text | Google Scholar

13. He C. Balancing nutrient and energy demand and supply via autophagy. Curr Biol. (2022) 32:R684–96. doi: 10.1016/j.cub.2022.04.071

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Ariav Y, Ch’ng JH, Christofk HR, Ron-Harel N, Erez A. Targeting nucleotide metabolism as the nexus of viral infections, cancer, and the immune response. Sci Adv. (2021) 7:eabg6165. doi: 10.1126/sciadv.abg6165

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Ma J, Zhong M, Xiong Y, Gao Z, Wu Z, Liu Y, et al. Emerging roles of nucleotide metabolism in cancer development: progress and prospect. Aging (Albany NY). (2021) 13:13349–58. doi: 10.18632/aging.v13i9

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Shi DD, Savani MR, Abdullah KG, McBrayer SK. Emerging roles of nucleotide metabolism in cancer. Trends In Cancer. (2023) 9:624–35. doi: 10.1016/j.trecan.2023.04.008

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Liu J, Zhong L, Deng D, Zhang Y, Yuan Q, Shang D. The combined signatures of the tumour microenvironment and nucleotide metabolism-related genes provide a prognostic and therapeutic biomarker for gastric cancer. Sci Rep. (2023) 13:6622. doi: 10.1038/s41598-023-33213-z

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Keshet R, Lee JS, Adler L, Iraqi M, Ariav Y, Lim LQJ, et al. Targeting purine synthesis in ASS1-expressing tumors enhances the response to immune checkpoint inhibitors. Nat Cancer. (2020) 1:894–908. doi: 10.1038/s43018-020-0106-7

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Sek K, Mølck C, Stewart GD, Kats L, Darcy PK, Beavis PA. Targeting adenosine receptor signaling in cancer immunotherapy. Int J Mol Sci. (2018) 19:3837. doi: 10.20944/preprints201810.0707.v1

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Walter M, Herr P. Re-discovery of pyrimidine salvage as target in cancer therapy. Cells. (2022) 11:739. doi: 10.3390/cells11040739

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Xia C, Yin S, To KKW, Fu L. CD39/CD73/A2AR pathway and cancer immunotherapy. Mol Cancer. (2023) 22:44. doi: 10.1186/s12943-023-01733-x

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Franco R, Rivas-Santisteban R, Navarro G, Reyes-Resina I. Adenosine receptor antagonists to combat cancer and to boost anti-cancer chemotherapy and immunotherapy. Cells. (2021) 10:2831. doi: 10.3390/cells10112831

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Jia W, Huang Z, Zhou L, Liou Y-C, Di Virgilio F, Ulrich H, et al. Purinergic signalling in cancer therapeutic resistance: From mechanisms to targeting strategies. Drug Resist Update. (2023) 70:100988. doi: 10.1016/j.drup.2023.100988

CrossRef Full Text | Google Scholar

24. Aria H, Rezaei M, Nazem S, Daraei A, Nikfar G, Mansoori B, et al. Purinergic receptors are a key bottleneck in tumor metabolic reprogramming: The prime suspect in cancer therapeutic resistance. Front In Immunol. (2022) 13:947885. doi: 10.3389/fimmu.2022.947885

CrossRef Full Text | Google Scholar

25. Yu Y, Yu J, Ge S, Su Y, Fan X. Novel insight into metabolic reprogrammming in cancer radioresistance: A promising therapeutic target in radiotherapy. Int J Biol Sci. (2023) 19:811. doi: 10.7150/ijbs.79928

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Madsen HB, Peeters MJ, Straten PT, Desler C. Nucleotide metabolism in the regulation of tumor microenvironment and immune cell function. Curr Opin In Biotechnol. (2023) 84:103008. doi: 10.1016/j.copbio.2023.103008

CrossRef Full Text | Google Scholar

Keywords: nucleotide metabolism, nucleotide, oncoimmunity, cancer, neoplasms

Citation: Wu J, Rong Y, Li T, Wilson CM, He Y, Chen D, Han J and Zhang X (2024) Editorial: Targeting nucleotide metabolism for enhancing antitumor immunity. Front. Immunol. 15:1412057. doi: 10.3389/fimmu.2024.1412057

Received: 04 April 2024; Accepted: 12 April 2024;
Published: 23 April 2024.

Edited and Reviewed by:

Peter Brossart, University of Bonn, Germany

Copyright © 2024 Wu, Rong, Li, Wilson, He, Chen, Han and Zhang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Xingmei Zhang, ZhangXingMei2022@outlook.com; Jin Han, hanjin_06@163.com; Danqian Chen, 20193019@nwu.edu.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.