Next Article in Journal
Failure to Achieve 70% of Recommended Protein Intake at One Year Predicts 13-Fold Higher Mortality After Gastrectomy
Previous Article in Journal
Intermittent Fasting and Probiotics for Gut Microbiota Modulation in Type 2 Diabetes Mellitus: A Narrative Review
Previous Article in Special Issue
Novel Scale for Clinical Identification of Adverse Magnesium/Calcium Imbalances: Applications and Perspectives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Opinion

Magnesium and Cancer Immunotherapy: A Narrative Practical Review

1
Medical Oncology Unit, Department of Medicine and Surgery, Kore University of Enna, 94019 Enna, Italy
2
Medical Oncology Unit, Department of Precision Medicine, University of Palermo, 90127 Palermo, Italy
3
Medical Oncology Unit, Casa di Cure Torina, via Spallitta n. 8, 90100 Palermo, Italy
*
Author to whom correspondence should be addressed.
Nutrients 2026, 18(1), 121; https://doi.org/10.3390/nu18010121 (registering DOI)
Submission received: 8 November 2025 / Revised: 20 December 2025 / Accepted: 26 December 2025 / Published: 30 December 2025
(This article belongs to the Special Issue Magnesium in Aging, Health and Diseases)

Abstract

Magnesium (Mg2+) has garnered the attention of oncologists due to its wide range of biological functions and frequent use as a complementary or integrative agent. In this study, a concise narrative review of the complex relationships between Mg2+ and immunotherapy for human malignancies is presented, in addition to a possible future therapeutic scenario. Pertinent full-text articles were thoroughly examined, and the most relevant ones were selected for inclusion in this review. A significant body of preclinical studies highlights the role of Mg2+ in regulating immune function, particularly in cytotoxic effector cells, underscoring the importance of maintaining adequate Mg2+ homeostasis mainly when immune-modulating agents are used in clinical practice. Whether serum Mg2+ levels influence the clinical outcomes of cancer patients treated with immune checkpoint blocker treatment remains to be fully elucidated. However, over the last decade, an increasing amount of data suggests that maintaining normal or slightly elevated serum levels of Mg2+ may enhance the response to immune therapy and even improve survival outcomes. New potential modulators of the tumor microenvironment and response to immunotherapy, such as injectable gels and metal-based biomaterials, are discussed.

1. Introduction

Magnesium (Mg2+) is one of the most important micronutrients in biological systems due to its wide range of biological functions and its pivotal role in regulating inflammation and immune responses to infectious diseases and cancer [1,2,3]. Mg2+ is essential in numerous biochemical processes, including adenosine triphosphate production, cellular signal transduction, nucleic acids and protein synthesis, and bone formation [4].
As shown in Figure 1, the relationships between Mg2+ and cancer are complex and multifaceted [5,6,7]. The scientific evidence on the association between Mg2+ and cancer risk is conflicting, apart from colorectal cancer. In general, Mg2+ deficiency results in immune dysfunctions and enhanced baseline inflammation associated with oxidative stress related to various age-associated morbidities and cancer. Mg2+ has a more evident role in anticancer therapy since Mg2+ deficiency is associated with drug- or chemotherapy-related hypomagnesemia, postoperative pain, cachexia, opioid-induced constipation, normal tissue protection from radiation damage, and prevention of nephrotoxicity [7].
In the last decade, immunotherapy has gained a pivotal role in the treatment of human malignancies, especially melanoma, lung cancer, renal cell carcinoma, triple-negative breast cancer, and uterine neoplasms [8]. Immunotherapy encompasses a range of agents, including immune checkpoint inhibitors, monoclonal antibodies, adoptive cell therapy, oncolytic viruses, and cancer vaccines. The former are routinely employed in patients, while the latter are in various stages of clinical development [8]. Despite immune checkpoint blockade therapy having gained a pivotal role in treating various cancer types, it still exhibits some limitations in terms of objective response rates and duration, as cancer cells can develop immune escape or tolerance [9,10]. In a recent study, 88% of inpatients with advanced cancers did not show any response to immunotherapy, with a median survival shorter than two months [11].
In this study, we review preclinical and clinical data of the relationship between Mg2+ and anticancer response to immune checkpoint blockade therapy. We (DS, GS, and MG) conducted a literature review on MEDLINE, PubMed, EMBASE, and the Web of Science to report the relationship between Mg2+ and cancer immunotherapy, selecting research deemed relevant to the purpose of this narrative review, which was approved by other authors (VG and MRV). The search complied partially with the Prisma guidelines (Figure 2). Selection was carried out according to Cohen’s kappa coefficient calculations to measure inter-rater reliability for qualitative items [12].

2. Preclinical Evidence

The role of Mg2+ in the immune response is best summarized by the title of a recent commentary by Dr. L. Bird, “Magnesium: essential for T cells” [13]. As shown in Figure 1, Mg2+ deficiency influences the activation and cytotoxicity of immune cells, the effectors of modern cancer immunotherapy [13,14,15,16,17,18,19]. Suboptimal extracellular Mg2+ levels reduce T cell receptor signal transduction by IL-2-inducible T cell kinase (ITK), necessary for lymphoid cell proliferation. The presence of Mg2+ is, in fact, required in the catalytic pocket of ITK since it is a pivotal element for various serine/threonine and tyrosine kinases’ function. In mice, reduced serum Mg2+ concentration is associated with an impaired CD8+ T cell response to influenza A virus and reduced T cell activation [14].
Figure 3 shows the complex effects of Mg2+ on the immune system, which result in cytokine production, cytotoxicity, and tumor cell apoptosis. Low intracellular free Mg2+ levels cause the defective expression of the natural killer activating receptor NKG2D in natural killer (NK) and CD8+ T cells and impair cytolytic responses against viruses, as observed in individuals deficient in the Mg2+ transporter 1 MAGT1—a critical regulator of basal intracellular free Mg2+ concentrations—who are at high risk of lymphoma and Epstein–Barr virus (EBV) infection [14,15]. In these patients, Mg2+ supplementation restores intracellular free Mg2+ and NKG2D while concurrently reducing EBV-infected cells in vivo, demonstrating a link between NKG2D cytolytic activity and EBV antiviral immunity in humans.
The lymphocyte function-associated antigen-1 (LFA-1) is an integrin present on the surface of immune cells, including lymphocytes, which plays a pivotal role in the emigration and activation of cytotoxicity [17]. This adhesion molecule requires Mg2+ to reach its active conformation on CD8+ T cells, allowing an increase in calcium flux, transduction, metabolic changes, immune synapse formation, and eventually cytotoxicity. LFA-1 transmits signals from the tumor microenvironment to the interior of cells, thereby enhancing the immune response, as demonstrated in bi-specific T cells and CAR-T cells [18]. This Mg2+-LFA-1 axis may represent a potentially targeted cellular pathway for nutrients.
Yuan et al. recently reviewed the immunomodulatory effects of metal-based biomaterials and their potential enhancing effects on anti-tumor immune responses, which are devoid of direct toxicity [19]. Degradable biomaterials/hydrogels/implantable-Mg2+ have excellent biocompatibility and mechanical issues, and combined with specific alloys (gadolinium, silver, and collagen scaffold), they may modulate the immune system, enhance tissue repair, and elicit an antitumor response through the release of Mg2+ ions and hydrogen during degradation. Lin et al. reviewed the antitumor activity of Mg2+ alloys [20]. In preclinical models, Mg2+ with a concentration exceeding 20 mM inhibits ovarian cancer cells, blocking cell cycles in the G0/G1 stage [21]. Moreover, it inhibits the in vitro and in vivo growth of hepatobiliary cancer cells and induces apoptosis [22,23]. Other authors also showed that Mg2+-containing implants prevent the formation of bone metastasis and tumor recurrence [24].
In vitro, non-cytotoxic Mg2+-containing tricalcium phosphates loaded with a hydrothermal extract of a human tubercle bacillus effectively stimulated granulocyte–macrophage colony-stimulating factor secretion via macrophage-like cells [25]. They also inhibited the growth of Lewis lung carcinoma cells [25]. A temperature-sensitive hydrogel containing alginate microspheres with MgCl2 allowed a gradual and sustained release of Mg2+ within the tumor microenvironment, promoting an immune microenvironment favorable to the cytotoxic activity of CD8+ T cells and NK cells and thereby enhancing the efficacy of immunotherapy [26]. Moreover, it converted low-immunogenic “cold” tumors into “hot” tumors with a high PD-L1 response.
Mechanistic studies of immune cell transcriptomics and flow cytometry phenotyping showed that Mg2+ induces a shift in proinflammatory M1 macrophages toward an anti-inflammatory M2 phenotype; activates CD8+ T-cells; and decreases monocyte production of proinflammatory cytokines (TNF-α and IL-6) [27,28]. Mg2+ may also reduce the expression of the CC-chemokine receptor 7 on macrophages, which plays a pivotal role in guiding lymphoid cells within organs and acquiring immunity and tolerance [29].

3. Clinical Data

Despite hypomagnesemia—i.e., serum Mg2+ levels below the normal range of 0.7 to 1.05 mmol/L—being common in clinical practice, Mg2+ dosages are usually not included in routine laboratory assessments. The total intracellular Mg2+ concentration is between 17 and 20 mM, but the free, or ionized, concentration is substantially lower at 0.5–1 mM. Most of the magnesium is bound to other molecules such as ATP, DNA, and proteins, which explains the large difference between the total and free concentrations. Whether serum Mg2+ levels influence the clinical outcomes of cancer patients treated with immune checkpoint blockers (ICBs) remains unclear. However, over the last decade, an increasing amount of data suggests that maintaining normal or slightly elevated serum levels of Mg2+ may enhance the response to immune therapy and even improve survival outcomes [18,30].
As shown in Figure 4, Feng et al. recently reported a large multi-center retrospective study involving 1441 patients—of whom 1042 had lung cancer, 270 had esophageal cancer, and 129 had Hodgkin lymphoma—treated with ICBs to assess the correlation between serum Mg2+ levels and clinical outcomes [30]. Patients were stratified according to a Mg2+ cut-off level of 0.79 mmol/L. Patients with Mg2+ levels of ≥0.79 mmol/L had longer progression-free survival and overall survival compared to those with Mg2+ levels of <0.79 mmol/L. In patients with lung cancer, high Mg2+ levels were associated with longer progression-free and overall survival, with hazard ratios of 1.486 and 1.78, respectively. Similar data were found for patients with esophageal cancer, with hazard ratios of 1.751 and 2.526 for progression-free and overall survival, respectively. With respect to both univariate and multivariate analyses, Mg2+ levels were found to be an independent prognostic factor.
Similar data were reported by Lötscher et al., who showed that patients with low Mg2+ levels affected by lung cancer treated with durvalumab and those with lymphoma treated with CAR-T had a worse clinical outcome (Figure 5) [18]. As shown in Figure 6, very recently, Liu et al. reported that low pre-treatment serum Mg2+ levels were significantly associated with poorer disease-free survival (HR = 8.730, p < 0.001) and OS (HR = 21.48, p < 0.001) in a large series of patients with advanced lung and esophageal cancer and Hodgkin’s lymphoma [31]. This difference also persisted in subgroup analysis. Multivariate Cox regression analysis confirmed that low serum magnesium was an independent prognostic factor for both OS (HR = 0.232, p = 0.007) and DFS (HR = 0.274, p = 0.004). However, no prospective trial has confirmed data from retrospective studies. Despite these intriguing retrospective data, to date, there is no prospective evidence of the impact of Mg2+ levels on the efficacy of immunotherapy in cancer.
The optimal timing and dosage of Mg2+ supplementation have not yet been established. Cancer itself and its treatments can sometimes cause electrolyte imbalances, including low Mg2+ levels, resulting in side effects such as muscle cramps or fatigue [7]. Maintaining stable Mg2+ levels may mitigate these effects, allowing patients to tolerate treatment more effectively. Table 1 shows the main oncological clinical settings of potential Mg2+ imbalance, symptoms, and suggested interventions. Physiological Mg2+ levels contribute to maintaining a healthy and balanced gut microbiome, which plays a role in immune regulation and can impact the effectiveness of ICBs [32,33]. Emerging evidence suggests that modulating the gut microbiome through interventions such as fecal microbiota transplantation, probiotics, prebiotics, and lifestyle modifications, as well as the wise use of antibiotics, may enhance therapeutic outcomes [33]. These data may be important when ICBs are administered in combination with chemotherapy with platinum salts, which are known to sometimes cause symptomatic hypomagnesemia [7].
A balanced diet usually provides sufficient Mg2+, but supplementation may be necessary in some cases. However, excessive supplementation can have negative impacts on immune function. Therefore, caution should be exercised when assuming it without a precise medical prescription, considering that Mg2+ is frequently used as a complementary or integrative agent and is readily available as an over-the-counter pharmaceutical product [34]. Hypermagnesemia can cause severe side effects, such as hypotension and respiratory depression, which may be particularly harmful in patients with lung cancer treated with immunotherapy.

4. Discussion

Mg2+ ions play a multidimensional role in tumor biology and therapy since they significantly impact tumor cell proliferation, migration, apoptosis, drug resistance, and oxidative stress. Mg2+ ions precisely regulate the cell cycle, ranging from modulating the activity of cyclin-dependent kinases and cyclins in the G1 phase to serving as cofactors for DNA polymerases to ensure DNA replication in the S phase and playing critical roles in spindle formation.
The tumor microenvironment comprises a stroma consisting of an extracellular matrix and various cells, including stromal and immune cells. In many malignancies, this microenvironment presents immunosuppressive factors that inhibit the function of immune cells and promote tumor growth [35]. A characteristic feature of the “cold” tumor phenotype is scarce immune cell infiltration, which responds poorly to immunotherapy [35]. Cancer cells employ camouflage mechanisms and suppress immunological reactions, thereby blocking the priming and infiltration of immune cells, which allows them to escape and elicit a poor response to immunotherapy [36].
Numerous strategies and adjuvants have been developed to extend immune responses in the tumor microenvironment, aiming to counteract cancer immune escape and improve the efficacy of immunotherapy in lung and breast cancer. These strategies include Toll-like receptor agonists, nanocarriers, oncolytic viruses, and phytochemicals [35]. These data have resulted in the evaluation of biomaterials in remodeling the TME to increase the effectiveness of immunotherapy [35]. Progress in nanotechnology has revealed the potential of Mg2+-based platforms (Mg-NMs), which allow the controlled release of Mg2+. This technology represents an innovative approach for delivering cancer therapies, leveraging its biodegradability, biocompatibility, and multifunctional antitumor mechanisms [37].
Injectable hydrogels are versatile delivery matrices. Hydrogels enable the encapsulation and controlled release of small molecules and cells, including NK, T cells, antigen-presenting cells, dendritic cells, and those involved in chemoimmunotherapy, radioimmunotherapy, and photothermal immunotherapy [38]. Injectable hydrogels bypass the extracellular matrix and gradually release small molecules or cells into the tumor microenvironment, being effective carriers for immunotherapy delivery [39]. Mg2+ enhances the hydrogel’s mechanical properties and releases ions that promote cell activity, including the proliferation and differentiation of stem cells, and it modulates the immune response by encouraging the polarization of anti-inflammatory macrophages [38]. In fact, Mg2+ and its byproducts, such as hydrogen and magnesium hydroxide, can regulate lactic acid levels, modify pH, and decrease tumor microenvironment acidity and oxidative stress, thereby promoting immune cell function and ultimately inhibiting tumor growth and metastasis formation [40]. The clinical use of Mg2+-based materials in combination with various therapeutic approaches may potentially enhance cancer treatments. Degradable Mg2+ implants can induce P53-mediated tumor cell apoptosis through a controlled release of hydrogen and inhibit gallbladder cancer [23,41].
Signaling cascades that control the immunological state and cell fate in the tumor microenvironment are profoundly influenced by slight variations in the extracellular/intracellular concentrations of metal ions [42]. Nanomedicine metal ions ensure precise delivery and selectivity to the tumor microenvironment and, therefore, may be applied as immune modulators in anticancer immunotherapy. Metal ions modulate immune status by activating the cGAS-STING pathway and inducing immunogenic cell death, enabling a multidimensional activation of immunotherapy [43].
Our review outlines the current knowledge on the relationships between Mg2+ and cancer immunotherapy. Despite the data published to date, this review highlights the limitations and knowledge gaps with respect to the role of Mg2+. The lack of prospective evidence for supplementation efficacy, potential confounding in retrospective analyses, the inability to distinguish serum magnesium as a causal vs. correlative factor, and the fact that the 0.79 mmol/L cutoff was statistically derived rather than biologically validated are important limitations. Therefore, there is a need for prospective randomized trials in cancer patients treated with ICB, with preplanned Mg2+ supplementation dosage, schedule, pharmaceutical formulation, timing of administration, and efficacy assessments.

5. Conclusions

Immunotherapy is undoubtedly a major advance in medical oncology. However, the results of immunotherapy are not always favorable, probably due to the characteristics of patients and tumors [44]. Mg2+ can influence the immune response and intestinal microbiome, which is known to affect the response to ICBs. Retrospective data suggest a role for Mg2+ in the outcome of cancer immunotherapy [45]. This observation is a matter of scientific debate due to the lack of prospective data [11,46]. Monitoring Mg2+ levels before and during immunotherapy may be valuable for predicting a patient’s response to treatment. Although Mg2+ is effective in counteracting hypomagnesemia induced by anti-EGFR antibodies, the optimal timing and dosing of magnesium supplementation in chemotherapy, if proven beneficial, need to be determined [47]. Supplementation may potentially be considered for patients with low Mg2+ levels to enhance treatment efficacy; however, this requires further clinical testing. Clinical trials are crucial for establishing clear guidelines for Mg2+ supplementation in cancer patients undergoing immunotherapy. For patients considering magnesium supplements, it is essential to consult healthcare providers to ensure safety and appropriate dosing, especially since excessive Mg2+ can cause adverse effects. In the future, prospects include metal-based biomaterials with immunomodulatory functions in cancer immunotherapy, designed to enhance the efficacy of immunotherapy.

Author Contributions

Conceptualization, D.S. and G.S.; methodology, D.S. and G.S.; validation, G.S.; formal analysis, V.G.; investigation, G.S. and D.S.; data curation, A.C., G.S. and D.S.; writing—original draft preparation, M.G. and M.R.V.; writing—review and editing, V.G. and M.R.V.; supervision, V.G. and M.R.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study did not require ethical approval.

Data Availability Statement

The data presented are available in the published medical literature.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
Mg2+Magnesium;
NKsNatural killer cells;
EBVEpstein–Barr virus;
ICBsImmune checkpoint blockers.

References

  1. Dominguez, L.J.; Veronese, N.; Barbagallo, M. Magnesium and the Hallmarks of Aging. Nutrients 2024, 16, 496. [Google Scholar] [CrossRef] [PubMed]
  2. Trapani, V.; Wolf, F.I. Dysregulation of Mg2+ homeostasis contributes to the acquisition of cancer hallmarks. Cell Calcium 2019, 83, 102078. [Google Scholar] [CrossRef] [PubMed]
  3. Ashique, S.; Kumar, S.; Hussain, A.; Mishra, N.; Garg, A.; Gowda, B.H.J.; Farid, A.; Gupta, G.; Dua, K.; Taghizadeh-Hesary, F. A narrative review on the role of magnesium in immune regulation, inflammation, infectious diseases, and cancer. J. Health Popul. Nutr. 2023, 42, 74, Erratum in J. Health Popul. Nutr. 2023, 42, 117. [Google Scholar] [CrossRef] [PubMed]
  4. Schwalfenberg, G.K.; Genuis, S.J. The Importance of Magnesium in Clinical Healthcare. Scientifica 2017, 2017, 4179326. [Google Scholar] [CrossRef]
  5. Durlach, J.; Bara, M.; Guiet-Bara, A.; Collery, P. Relationship between magnesium, cancer, and carcinogenic or anti-cancer metals. Anti-Cancer Res. 1986, 6, 1353–1361. [Google Scholar]
  6. Castiglioni, S.; Maier, J.A. Magnesium and cancer: A dangerous liaison. Magnes. Res. 2011, 24, S92–S100. [Google Scholar] [CrossRef]
  7. Sambataro, D.; Scandurra, G.; Scarpello, L.; Gebbia, V.; Dominguez, L.J.; Valerio, M.R. A Practical Narrative Review on the Role of Magnesium in Cancer Therapy. Nutrients 2025, 17, 2272. [Google Scholar] [CrossRef]
  8. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef]
  9. Aktar, N.; Yueqing, C.; Abbas, M.; Zafar, H.; Paiva-Santos, A.C.; Zhang, Q.; Chen, T.; Ahmed, M.; Raza, F.; Zhou, X. Understanding of Immune Escape Mechanisms and Advances in Cancer Immunotherapy. J. Oncol. 2022, 2022, 8901326. [Google Scholar] [CrossRef]
  10. Tufail, M.; Jiang, C.H.; Li, N. Immune evasion in cancer: Mechanisms and cutting-edge therapeutic approaches. Signal Transduct. Target. Ther. 2025, 10, 227. [Google Scholar] [CrossRef]
  11. Riaz, F.; Vaughn, J.L.; Zhu, H.; Dickerson, J.C.; Sayegh, H.E.; Brongiel, S.; Baldwin, E.; Kier, M.W.; Zaemes, J.; Hearn, C.; et al. Inpatient Immunotherapy Outcomes Study: A Multicenter Retrospective Analysis. JCO Oncol. Pract. 2025, 21, 1165–1173. [Google Scholar] [CrossRef] [PubMed]
  12. McHugh, M.L. Interrater reliability: The kappa statistic. Biochem. Med. 2012, 22, 276–282. [Google Scholar] [CrossRef]
  13. Bird, L. Magnesium: Essential for T cells. Nat. Rev. Immunol. 2022, 22, 144–145. [Google Scholar] [CrossRef]
  14. Kanellopoulou, C.; George, A.B.; Masutani, E.; Cannons, J.L.; Ravell, J.C.; Yamamoto, T.N.; Smelkinson, M.G.; Jiang, P.D.; Matsuda-Lennikov, M.; Reilley, J.; et al. Mg2+ regulation of kinase signaling and immune function. J. Exp. Med. 2019, 216, 1828–1842. [Google Scholar] [CrossRef]
  15. Chaigne-Delalande, B.; Lenardo, M.J. Divalent cation signaling in immune cells. Trends Immunol. 2014, 35, 332–344. [Google Scholar] [CrossRef]
  16. Chaigne-Delalande, B.; Li, F.Y.; O’Connor, G.M.; Lukacs, M.J.; Jiang, P.; Zheng, L.; Shatzer, A.; Biancalana, M.; Pittaluga, S.; Matthews, H.F.; et al. Mg2+ regulates cytotoxic functions of NK and CD8 T cells in chronic EBV infection through NKG2D. Science 2013, 341, 186–191. [Google Scholar] [CrossRef] [PubMed]
  17. Verma, N.K.; Kelleher, D. Not Just an Adhesion Molecule: LFA-1 Contact Tunes the T Lymphocyte Program. J. Immunol. 2017, 199, 1213–1221. [Google Scholar] [CrossRef]
  18. Lötscher, J.; Martí, I.; Líndez, A.A.; Kirchhammer, N.; Cribioli, E.; Giordano Attianese, G.M.P.; Trefny, M.P.; Lenz, M.; Rotschild, S.I.; Strati, P.; et al. Magnesium sensing via LFA-1 regulates CD8+ T cell effector function. Cell 2022, 185, 585–602.e29. [Google Scholar] [CrossRef]
  19. Yuan, K.; Zhang, C.; Pan, X.; Hu, B.; Zhang, J.; Yang, G. Immunomodulatory metal-based biomaterials for cancer immunotherapy. J. Control Release 2024, 375, 249–268. [Google Scholar] [CrossRef]
  20. Lin, Z.; Wei, Y.; Yang, H. Mg alloys with antitumor and anticorrosion properties for orthopedic oncology: A review from mechanisms to application strategies. APL Bioeng. 2024, 8, 021504. [Google Scholar] [CrossRef]
  21. Qiao, S.; Wang, Y.; Zan, R.; Wu, H.; Sun, Y.; Peng, H.; Zhang, R.; Song, Y.; Ni, J.; Zhang, S.; et al. Biodegradable Mg Implants Suppress the Growth of Ovarian Tumor. ACS Biomater. Sci. Eng. 2020, 6, 1755–1763. [Google Scholar] [CrossRef]
  22. Li, T.; Xu, W.; Liu, C.; He, J.; Wang, Q.; Zhang, D.; Sui, K.; Zhang, Z.; Sun, H.; Yang, K.; et al. Anticancer Effect of Biodegradable Magnesium on Hepatobiliary Carcinoma: An In Vitro and In Vivo Study. ACS Biomater. Sci. Eng. 2021, 7, 2774–2782. [Google Scholar] [CrossRef] [PubMed]
  23. Peng, H.; Fan, K.; Zan, R.; Gong, Z.J.; Sun, W.; Sun, Y.; Wang, W.; Jiang, H.; Lou, J.; Ni, J.; et al. Degradable magnesium implants inhibit gallbladder cancer. Acta Biomateralia 2021, 128, 514–522. [Google Scholar] [CrossRef]
  24. Yang, G.; Yang, H.; Shi, L.; Wang, T.; Zhou, W.; Zhou, T.; Han, W.; Zhang, Z.; Lu, W.; Hu, J. Enhancing Corrosion Resistance, Osteoinduction, and Antibacterial Properties by Zn/Sr Additional Surface Modification of Magnesium Alloy. ACS Biomater. Sci Eng. 2018, 4, 4289–4298. [Google Scholar] [CrossRef]
  25. Wang, X.; Li, X.; Onuma, K.; Sogo, Y.; Ohno, T.; Ito, A. Zn- and Mg- containing tricalcium phosphates-based adjuvants for cancer immunotherapy. Sci. Rep. 2013, 3, 2203. [Google Scholar] [CrossRef]
  26. Ma, L.; Wang, X.; Wu, Y.; Zhang, Y.; Yuan, X.; Mao, J.; Li, Q.; Gong, S. Controlled release of manganese and magnesium ions by microsphere-encapsulated hydrogel enhances cancer immunotherapy. J. Control Release 2024, 372, 682–698. [Google Scholar] [CrossRef]
  27. Hu, T.; Xu, H.; Wang, C.; Qin, H.; An, Z. Magnesium enhances the chondrogenic differentiation of mesenchymal stem cells by inhibiting activated macrophage-induced inflammation. Sci. Rep. 2018, 8, 3406. [Google Scholar] [CrossRef] [PubMed]
  28. Sun, L.; Li, X.; Xu, M.; Yang, F.; Wang, W.; Niu, X. In vitro immunomodulation of magnesium on monocytic cell toward anti-inflammatory macrophages. Regen. Biomater. 2020, 7, 391–401. [Google Scholar] [CrossRef]
  29. Alrumaihi, F. The Multi-Functional Roles of CCR7 in Human Immunology and as a Promising Therapeutic Target for Cancer Therapeutics. Front. Mol. Biosci. 2022, 9, 834149. [Google Scholar] [CrossRef]
  30. Feng, Y.; Gao, M.; Xu, X.; Liu, H.; Lu, K.; Song, Z.; Yu, J.; Liu, X.; Han, X.; Li, L.; et al. Elevated serum magnesium levels prompt favorable outcomes in cancer patients treated with immune checkpoint blockers. Eur. J. Cancer 2024, 213, 115069. [Google Scholar] [CrossRef] [PubMed]
  31. Liu, X.; Sun, X.; Lin, Y.; Li, C.; Shen, Q.; Ding, J.; Li, T.; Lu, D.; Li, X.; Zhao, S.; et al. Preoperative serum magnesium level as a prognostic and predictive biomarker in locally advanced gastric cancer patients receiving neoadjuvant immunochemotherapy. World J. Surg. Oncol. 2025, 23, 432. [Google Scholar] [CrossRef]
  32. Laragione, T.; Harris, C.; Azizgolshani, N.; Beeton, C.; Bongers, G.; Gulko, P.S. Magnesium increases numbers of Foxp3+ Treg cells and reduces arthritis severity and joint damage in an IL-10-dependent manner mediated by the intestinal microbiome. EBioMedicine 2023, 92, 104603. [Google Scholar] [CrossRef]
  33. Elkrief, A.; Pidgeon, R.; Maleki Vareki, S.; Messaoudene, M.; Castagner, B.; Routy, B. The gut microbiome as a target incancer immunotherapy: Opportunities and challenges for drug development. Nat. Rev. Drug Discov. 2025, 24, 685–704. [Google Scholar] [CrossRef]
  34. Valerio, M.R.; Scandurra, G.; Greco, M.; Gebbia, V.; Piazza, D.; Sambataro, D. A Survey on the Prescribing Orientation Towards Complementary Therapies Among Oncologists in Italy: Symptoms and Unmet Patient Needs. In Vivo 2025, 39, 1000–1008. [Google Scholar] [CrossRef]
  35. Gao, F.; You, X.; Yang, L.; Zou, X.; Sui, B. Boosting immune responses in lung tumor immune microenvironment: A comprehensive review of strategies and adjuvants. Int. Rev. Immunol. 2024, 43, 280–308. [Google Scholar] [CrossRef] [PubMed]
  36. Liu, Y.T.; Wang, Y.L.; Wang, S.; Li, J.J.; He, W.; Fan, X.J.; Wan, X.B. Turning cold tumors into hot tumors to ignite immunotherapy. Mol. Cancer 2025, 24, 254. [Google Scholar] [CrossRef]
  37. Li, J.; Zhang, G.; Yu, C.; Hu, Z.; Feng, L.; Yang, P. Magnesium-based nanomaterials for cancer therapy: Unraveling mechanisms and current developments. Coord. Chem. Rev. 2026, 547, 217119. [Google Scholar] [CrossRef]
  38. Huang, L.; Lin, R.; Chen, J.; Qi, Y.; Lin, L. Magnesium Ion: A New Switch in Tumor Treatment. Biomedicines 2024, 12, 1717. [Google Scholar] [CrossRef]
  39. Mohaghegh, N.; Ahari, A.; Zehtabi, F.; Buttles, C.; Davani, S.; Hoang, H.; Tseng, K.; Zamanian, B.; Khosravi, S.; Daniali, A.; et al. Injectable hydrogels for personalized cancer immunotherapies. Acta Biomater. 2023, 172, 67–91. [Google Scholar] [CrossRef]
  40. Ma, Y.; Wang, Y.; Song, S.; Yu, X.; Xu, C.; Wan, L.; Yao, F.; Witte, F.; Yang, S. Mechanism and application prospect of magnesium-based materials in cancer treatment. J. Magnes. Alloys 2025, 13, 982–1011. [Google Scholar] [CrossRef]
  41. Zan, R.; Wang, H.; Cai, W.; Ni, J.; Luthringer-Feyerabend, B.J.C.; Wang, W.; Peng, H.; Ji, W.; Yan, J.; Xia, J.; et al. Controlled release of hydrogen by implantation of magnesium induces P53-mediated tumor cells apoptosis. Bioact. Mater. 2021, 9, 385–396. [Google Scholar] [CrossRef] [PubMed]
  42. Shen, F.; Fang, Y.; Wu, Y.; Zhou, M.; Shen, J.; Fan, X. Metal ions and nanometallic materials in antitumor immunity: Function, application, and perspective. J. Nanobiotechnol. 2023, 21, 20. [Google Scholar] [CrossRef]
  43. Wang, L.; Lin, D.; Li, M.; Jiang, Y.; Yang, Y.; Wang, H.; Chu, H.; Ye, J.; Liu, Y. Bioactive metallic nanoparticles for synergistic cancer immunotherapy. Acta Pharm. Sin. B 2025, 15, 1869–1911. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, Z.; Wang, Z. Letter Re: Elevated serum magnesium levels prompt favorable outcomes in cancer patients treated with immune checkpoint blockers. Eur. J. Cancer 2025, 216, 115187. [Google Scholar] [CrossRef]
  45. Lima, F.D.S.; Santos, M.Q.D.; Makiyama, E.N.; Hoffmann, C.; Fock, R.A. The essential role of magnesium in immunity and gut health: Impacts of dietary magnesium restriction on peritoneal cells and intestinal microbiome. J. Trace Elem. Med. Biol. 2025, 88, 127604. [Google Scholar] [CrossRef]
  46. Feng, Y.; Zhang, H.; Wang, X. Response to letter re: “Elevated serum magnesium levels prompt favorable outcomes in cancer patients treated with immune checkpoint blockers”. Eur. J. Cancer 2025, 216, 115249. [Google Scholar] [CrossRef]
  47. Kato, T.; Kawaguchi, T.; Funakoshi, T.; Fujiwara, Y.; Yasuda, Y.; Ando, Y. Efficacy of Magnesium Supplementation in Cancer Patients Developing Hypomagnesemia Due to Anti-EGFR Antibody: A Systematic Review. Cancer Diagn. Progn. 2024, 4, 390–395. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The relationships between magnesium and cancer in various settings. The figure depicts the pathophysiology and causative factors of hypomagnesemia, as well as biochemical consequences leading to an increase in inflammatory mediators. The red rectangle outlines the effects of hypomagnesemia on the immune system, which may cause a reduced response to ICBs or CAR-T therapy in cancer patients (modified with permission from Sambataro et al., Nutrients, 2025 [7]).
Figure 1. The relationships between magnesium and cancer in various settings. The figure depicts the pathophysiology and causative factors of hypomagnesemia, as well as biochemical consequences leading to an increase in inflammatory mediators. The red rectangle outlines the effects of hypomagnesemia on the immune system, which may cause a reduced response to ICBs or CAR-T therapy in cancer patients (modified with permission from Sambataro et al., Nutrients, 2025 [7]).
Nutrients 18 00121 g001
Figure 2. Flow diagram of manuscript selection process for this narrative review.
Figure 2. Flow diagram of manuscript selection process for this narrative review.
Nutrients 18 00121 g002
Figure 3. The multiple relationships between magnesium and immune system activation. The figure depicts the effect of Mg2+ on the natural killer activating receptor NKG2D, ITK activation, etc.
Figure 3. The multiple relationships between magnesium and immune system activation. The figure depicts the effect of Mg2+ on the natural killer activating receptor NKG2D, ITK activation, etc.
Nutrients 18 00121 g003
Figure 4. Main retrospective studies showing the effects of Mg2+ in cancer patients treated with immunotherapy (modified from Liu et al. [31] and Feng et al. [30]).
Figure 4. Main retrospective studies showing the effects of Mg2+ in cancer patients treated with immunotherapy (modified from Liu et al. [31] and Feng et al. [30]).
Nutrients 18 00121 g004
Figure 5. Results of CAR-T in lymphoma and immunotherapy with durvalumab in non-small cell carcinoma according to the presence of hypomagnesemia, showing mean serum Mg2+ level differences and reduced overall survival in patients with reduced Mg2+ levels (reproduced with permission from Lötscher et al., Cell, 2022 [18]). ** p < 0.01, **** p < 0.0001.
Figure 5. Results of CAR-T in lymphoma and immunotherapy with durvalumab in non-small cell carcinoma according to the presence of hypomagnesemia, showing mean serum Mg2+ level differences and reduced overall survival in patients with reduced Mg2+ levels (reproduced with permission from Lötscher et al., Cell, 2022 [18]). ** p < 0.01, **** p < 0.0001.
Nutrients 18 00121 g005
Figure 6. Schematic representation of the tumor microenvironment and involved cells in direct and indirect antitumor activity. CAR-T cells and effector gamma lymphocytes are involved in direct cancer cell killing; T cells, gamma interferon stimulated dendritic cells, B cell class switching, and granulocyte/macrophage stimulating factors (GM-CSF) activated granulocytes participate in indirect antineoplastic activity.
Figure 6. Schematic representation of the tumor microenvironment and involved cells in direct and indirect antitumor activity. CAR-T cells and effector gamma lymphocytes are involved in direct cancer cell killing; T cells, gamma interferon stimulated dendritic cells, B cell class switching, and granulocyte/macrophage stimulating factors (GM-CSF) activated granulocytes participate in indirect antineoplastic activity.
Nutrients 18 00121 g006
Table 1. Main oncological clinical settings of potential Mg2+ imbalance, symptoms, and suggested interventions.
Table 1. Main oncological clinical settings of potential Mg2+ imbalance, symptoms, and suggested interventions.
SettingEffect on Mg2+ LevelsActionsPharmaceutical Form
Platinum salts
chemotherapy
Renal reduction
(hypomagnesemia);
tremor, spams, arrythmia
Oral or i.v.
supplementation; regular serum Mg2+
evaluation
Mg2+ Citrate or chloride; dose
200–400 mg/day i.v.
Anti-EGFR antibodies
(cetuximab; panitumumab)
Increased Mg2+
renal excretion;
asthenia, tremors
Monitoring every 2–4 weeks;
supplement if Mg2+ <1.5 mg/dL
Mg2+ citrate or pidolate; dose 200–400 mg/day
Data
Proton pump inhibitorsReduced Mg2+
intestinal absorption
Consider withdrawal or long- term
supplementation
Mg2+ glycinate or malate; dose 150–200 mg/day
ImmunotherapyNormal or slightly elevated
Mg2+ levels correlated to
better outcomes
Keep Mg2+ within the physiological range of 1.7–2.3 mg/dLData are missing, possibly the same as for anti-EGFR antibodies
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sambataro, D.; Scandurra, G.; Gebbia, V.; Greco, M.; Ciminna, A.; Valerio, M.R. Magnesium and Cancer Immunotherapy: A Narrative Practical Review. Nutrients 2026, 18, 121. https://doi.org/10.3390/nu18010121

AMA Style

Sambataro D, Scandurra G, Gebbia V, Greco M, Ciminna A, Valerio MR. Magnesium and Cancer Immunotherapy: A Narrative Practical Review. Nutrients. 2026; 18(1):121. https://doi.org/10.3390/nu18010121

Chicago/Turabian Style

Sambataro, Daniela, Giuseppa Scandurra, Vittorio Gebbia, Martina Greco, Alessio Ciminna, and Maria Rosaria Valerio. 2026. "Magnesium and Cancer Immunotherapy: A Narrative Practical Review" Nutrients 18, no. 1: 121. https://doi.org/10.3390/nu18010121

APA Style

Sambataro, D., Scandurra, G., Gebbia, V., Greco, M., Ciminna, A., & Valerio, M. R. (2026). Magnesium and Cancer Immunotherapy: A Narrative Practical Review. Nutrients, 18(1), 121. https://doi.org/10.3390/nu18010121

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop