Vanadium, a Promising Element for Cancer Treatment
Abstract
1. Introduction
2. Vanadium and Its Antineoplastic Potential
- -
- Inhibition of enzymes, such as tyrosine phosphatase, promoting the activation of tyrosine kinases and oncogene suppressors, which induces apoptosis in cancer cells.
- -
- DNA interaction with vanadium complexes inserting between DNA strands, causing cell cycle breaks or alterations. Some other compounds, such as ‘Metvan’, which has an extensive aromatic system strongly coordinated with V, can also interact with DNA through aromatic π-stacking, which interferes with DNA replication and uncontrolled cell division.
- -
- Generation of reactive oxygen species (ROS) increasing the production of superoxide and hydrogen peroxide, leading to a state of oxidative stress and apoptosis of cancer cells.
- -
- Differentiation between normal and cancer cells, with some vanadium complexes such as VO(maltol)2, which showed selectivity in affecting cancer cells while sparing normal cells.
- -
- Inhibition of specific demethylases, through the vanadium complex bearing tridentate shiff base ligands, which showed potential to inhibit lysine-specific demethylase (LSD1), an important target in cancer treatment.
3. In Vivo and in Vitro Evidence of Vanadium’s Anticancer Activity
3.1. Breast Cancer
3.2. Liver Cancer
3.3. Colorectal Cancer
3.4. Hematological Neoplasms
3.5. Skin Cancer
3.6. Lung Cancer
3.7. Nervous System Cancer
3.8. Bone Cancer
4. Anticancer Effects in Other Types of Cancer
5. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
NH4VO3 | Ammonium metavanadate |
NaVO3 | Sodium metavanadate |
KVO3 | Potassium metavanadate |
VO4SO4 | Vanadyl sulfate |
Na6V10O28 | Sodium decavanadate |
V2O5 | Vanadium pentoxide |
VOCl2 | Vanadyl dichloride |
VCl3 | Vanadium(III) chloride |
(C5H5)2VCl2 | Vanadocene dichloride |
VO1a | Vanadium oxide compound 1a |
VO1b | Vanadium oxide compounds 1b |
VOL | IV- oxidovanadium imidazole complex |
OVMI | 1-methylimidazole oxidovanadium complex |
VnNp | Vanadium nanoparticles |
SOV | Sodium orthovanadate |
VO(ma)2 | Bis(maltolate)- oxidovanadium(IV) |
VO(acac)2 | Bis(acetylacetonate)- oxidovanadium(IV) |
([IV(L)] complex) | 4-bromo-2-(((5-chloro-2-hydroxyphenyl) imino) methyl) phenol ([IV(L)] complex |
VIO nanoparticles | Vanadium-doped iron oxide (VIO) nanoparticles |
VO(NG)2 | Vanadyl N-(2-hydroxyacetophenone) glycinate |
Metvan | 4,7-dimethyl-1,10-phenathroline 7 sulphate oxidovanadium(IV) |
VOsil Na2[VO(silibinin)2]·6H2O | Oxidovanadium complex coupled with silibinin |
VOchrys [VO(chrysin)2EtOH]2 | Oxidovanadium complex coupled with chrysin |
VOlut [VO(lut)(H2O)2]Na3·H2O | Oxidovanadium complex coupled with luteolin |
VCrZnO4 NPs | Zinc-chromium-vanadate nanoparticles |
PTANAP- VO | ((E)-N′-((2-hydroxy naphthalene -1-yl)methylene)-4-methylbenzohydrazide) PTANAP oxidovanadium complex |
[VO(CUAP)SO4] | Vanadyl complex with a tridentate coumarin ligand |
Nano Cu-VO (IV) | Vanadyl complex with a tridentate coumarin ligand functionalized with copper nanoparticles |
VS2 | 1,2-dimethyl-3-hydroxy-4 (1H)-pyridinonate |
VS3 | 1-methyl-3-hydroxy-4 (1H)-pyridinonate |
VS4 | 1-phenyl-2-methyl-3-hydroxy-4 (1H)-pyridinonate |
V2O5 NP | V2O5 nanoparticles |
Ni@V2O5 NP | V2O5 nanoparticles doped with nickel |
VNPs@Salvia officinalis | Vanadium nanoparticles synthesized using Salvia officinalis extract |
VO(CQ)2 | Oxidovanadium(IV)-clioquinol complex |
V-doped HAPs | Hydroxyapatite nanoparticles doped with V |
NPIP | VO-2-hydroxy-1-naphthaldehyde thiosemicarbazone (hntdtsc) 2-(4-nitrophenyl)-,10-phenanthroline |
LpVO | Oxidovanadium complexes coupled with phenolate |
References
- Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
- Chen, S.; Cao, Z.; Prettner, K.; Kuhn, M.; Yang, J.; Jiao, L.; Wang, Z.; Li, W.; Geldsetzer, P.; Bärnighausen, T.; et al. Estimates and Projections of the Global Economic Cost of 29 Cancers in 204 Countries and Territories From 2020 to 2050. JAMA Oncol. 2023, 9, 465–472. [Google Scholar] [CrossRef] [PubMed]
- Lucaciu, R.L.; Hangan, A.C.; Sevastre, B.; Oprean, L.S. Metallo-drugs in cancer therapy: Past, present and future. Molecules 2022, 27, 6485. [Google Scholar] [CrossRef] [PubMed]
- Ghosh, S. Cisplatin: The first metal based anticancer drug. Bioorg. Chem. 2019, 88, 102925. [Google Scholar] [CrossRef]
- Paprocka, R.; Wiese-Szadkowska, M.; Janciauskiene, S.; Kosmalski, T.; Kulik, M.; Helmin-Basa, A. Latest developments in metal complexes as anticancer agents. Coord. Chem. Rev. 2022, 452, 214307. [Google Scholar] [CrossRef]
- Sastry, J.; Kellie, S.J. Severe neurotoxicity, ototoxicity and nephrotoxicity following high-dose cisplatin and amifostine. Pediatr. Hematol. Oncol. 2005, 22, 441–445. [Google Scholar] [CrossRef]
- Peña, Q.; Wang, A.; Zaremba, O.; Shi, Y.; Scheeren, H.W.; Metselaar, J.M.; Kiessling, F.; Pallares, R.M.; Wuttke, S.; Lammers, T. Metallodrugs in cancer nanomedicine. Chem. Soc. Rev. 2022, 51, 2544–2582. [Google Scholar] [CrossRef]
- Sigel, A.; Sigel, H.; Freisinger, E.; Sigel, R.K.O. (Eds.) Preface to Volume 18. In Metallo-Drugs: Development and Action of Anticancer Agents; Metal Ions in Life Sciences; Walter de Gruyter GmbH: Berlin, Germany, 2018. [Google Scholar] [CrossRef]
- Adhikari, S.; Nath, P.; Das, A.; Datta, A.; Baildya, N.; Duttaroy, A.K.; Pathak, S. A review on metal complexes and its anti-cancer activities: Recent updates from in vivo studies. Biomed. Pharmacother. 2024, 171, 116211. [Google Scholar] [CrossRef]
- Ndagi, U.; Mhlongo, N.; Soliman, M.E. Metal complexes in cancer therapy—An update from drug design perspective. Drug Des. Dev. Ther. 2017, 11, 599–616. [Google Scholar] [CrossRef]
- Leon, I.E.; Cadavid-Vargas, J.F.; Di Virgilio, A.L.; Etcheverry, S.B. Vanadium, ruthenium and copper compounds: A new class of nonplatinum metallodrugs with anticancer activity. Curr. Med. Chem. 2017, 24, 112–148. [Google Scholar] [CrossRef]
- Yeo, C.I.; Ooi, K.K.; Tiekink, E.R. Gold-based medicine: A paradigm shifts in anti-cancer therapy? Molecules 2018, 23, 1410. [Google Scholar] [CrossRef] [PubMed]
- González-Ballesteros, M.M.; Mejía, C.; Ruiz-Azuara, L. Metallodrugs: An approach against invasion and metastasis in cancer treatment. FEBS Open Bio 2022, 12, 880–899. [Google Scholar] [CrossRef] [PubMed]
- Lu, Y.; Ma, X.; Chang, X.; Liang, Z.; Lv, L.; Shan, M.; Lu, Q.; Wen, Z.; Gust, R.; Liu, W. Recent development of gold (I) and gold (III) complexes as therapeutic agents for cancer diseases. Chem. Soc. Rev. 2022, 51, 5518–5556. [Google Scholar] [CrossRef]
- Sharma, A.; Sudhindra, P.; Roy, N.; Paira, P. Advances in novel iridium (III) based complexes for anticancer applications: A review. Inorg. Chim. Acta 2020, 513, 119925. [Google Scholar] [CrossRef]
- Bouché, M.; Hognon, C.; Grandemange, S.; Monari, A.; Gros, P.C. Recent advances in iron-complexes as drug candidates for cancer therapy: Reactivity, mechanism of action and metabolites. Dalton Trans. 2020, 49, 11451–11466. [Google Scholar] [CrossRef]
- Konkankit, C.C.; Marker, S.C.; Knopf, K.M.; Wilson, J.J. Anticancer activity of complexes of the third row transition metals, rhenium, osmium, and iridium. Dalton Trans. 2018, 47, 9934–9974. [Google Scholar] [CrossRef]
- Li, X.; Gorle, A.K.; Sundaraneedi, M.K.; Keene, F.R.; Collins, J.G. Kinetically inert polypyridylruthenium(II) complexes as therapeutic agents. Coord. Chem. Rev. 2018, 375, 134–147. [Google Scholar] [CrossRef]
- Zhang, P.; Huang, H. Future potential of osmium complexes as anticancer drug candidates, photosensitizers and organelle-targeted probes. Dalton Trans. 2018, 47, 14841–14854. [Google Scholar] [CrossRef]
- Dasgupta, S.; Karim, S.; Banerjee, S.; Saha, M.; Saha, K.D.; Das, D. Designing of novel zinc (II) Schiff base complexes having acyl hydrazone linkage: Study of phosphatase and anti-cancer activities. Dalton Trans. 2020, 49, 1232–1240. [Google Scholar] [CrossRef]
- Singh, A.K.; Kumar, A.; Singh, H.; Sonawane, P.; Pathak, P.; Grishina, M.; Yadav, J.P.; Verma, A.; Kumar, P. Metal Complexes in cancer treatment: Journey so far. Chem. Biod. 2023, 20, e202300061. [Google Scholar] [CrossRef]
- Ferretti, V.A.; León, I.E. An overview of vanadium and cell signaling in potential cancer treatments. Inorganics 2022, 10, 47. [Google Scholar] [CrossRef]
- Rehder, D. Perspectives for vanadium in health issues. Future Med. Chem. 2016, 8, 325–338. [Google Scholar] [CrossRef]
- Sharfalddin, A.A.; Al-Younis, I.M.; Mohammed, H.A.; Dhahri, M.; Mouffouk, F.; Abu Ali, H.; Emwas, A.H. Therapeutic properties of vanadium complexes. Inorganics 2022, 10, 244. [Google Scholar] [CrossRef]
- Satya, H.K.; Gupta, S.; Siddique, A.; Joshi, S. Vanadium complexes as potential anticancer agents. Eng. Proc. 2023, 56, 91. [Google Scholar] [CrossRef]
- Singh, A.P.; Roy, S.; Maurya, I.C. Vanadium complexes: Potential candidates for therapeutic applications. Transit. Met. Chem. 2024, 49, 101–119. [Google Scholar] [CrossRef]
- De Sousa-Coelho, A.L.; Fraqueza, G.; Aureliano, M. Repurposing Therapeutic Drugs Complexed to Vanadium in Cancer. Pharmaceuticals 2023, 17, 12. [Google Scholar] [CrossRef]
- Kioseoglou, E.; Petanidis, S.; Gabriel, C.; Salifoglou, A. The chemistry and biology of vanadium compounds in cancer therapeutics. Coord. Chem. Rev. 2015, 301, 87–105. [Google Scholar] [CrossRef]
- Treviño, S.; Díaz, A.; Sánchez-Lara, E.; Sanchez-Gaytan, B.L.; Perez-Aguilar, J.M.; González-Vergara, E. Vanadium in biological action: Chemical, pharmacological aspects, and metabolic implications in diabetes mellitus. Biol. Trace Elem. Res. 2019, 188, 68–98. [Google Scholar] [CrossRef] [PubMed]
- Kumar, S.; Kumari, S.; Karan, R.; Kumar, A.; Rawal, R.K.; Gupta, P.K. Anticancer perspectives of vanadium complexes. Inorg. Chem. Commun. 2024, 161, 112014. [Google Scholar] [CrossRef]
- Barceloux, D.G. Vanadium. Clin. Toxicol. 1999, 37, 265–278. [Google Scholar] [CrossRef]
- Imtiaz, M.; Rizwan, M.S.; Xiong, S.; Li, H.; Ashraf, M.; Shahzad, S.M.; Tu, S. Vanadium, recent advancements and research prospects: A review. Environ. Int. 2015, 80, 79–88. [Google Scholar] [CrossRef]
- Hanus-Fajerska, E.; Wiszniewska, A.; Kamińska, I. A dual role of vanadium in environmental systems—Beneficial and detrimental effects on terrestrial plants and humans. Plants 2021, 10, 1110. [Google Scholar] [CrossRef] [PubMed]
- Crans, D.C.; Henry, L.; Cardiff, G.; Posner, B.I. Developing vanadium as an antidiabetic or anticancer drug: A clinical and historical perspective. Met. Ions Life Sci. 2019, 19, 203–230. [Google Scholar]
- Kostenkova, K.; Klugh, K.; Crans, D.C. Recent Advances of Medicinal Properties of Vanadium Compounds: Cancer and Other Diseases in Metal-Containing Molecules and Nanomaterials: From Diagnosis to Therapy, 1st ed.; CRC Press: Oxford, UK, 2024; pp. 19–50. [Google Scholar]
- Crans, D.C.; Yang, L.; Haase, A.; Yang, X. Health benefits of vanadium and its potential as an anticancer agent. Met. Ions Life Sci. 2018, 18, 251–279. [Google Scholar] [CrossRef]
- Kieler, J.; Gromek, A.; Nissen, N.I. Studies on the antineoplastic effect of vanadium salts. Acta Chir. Scand. Suppl. 1965, 343, 154–164. [Google Scholar] [PubMed]
- Bishayee, A.; Waghray, A.; Patel, M.A.; Chatterjee, M. Vanadium in the detection, prevention and treatment of cancer: The in vivo evidence. Cancer Lett. 2010, 294, 1–12. [Google Scholar] [CrossRef]
- Global Cancer Observator; IARC. Cancer TODAY. Globocan. 2025. Available online: https://gco.iarc.fr/today/en/dataviz/bars?mode=cancer&group_populations=1&multiple_populations=1 (accessed on 26 May 2025).
- Roy, S.; Banerjee, S.; Chakraborty, T. Vanadium quercetin complex attenuates mammary cancer by regulating the P53, Akt/mTOR pathway and downregulates cellular proliferation correlated with increased apoptotic events. Biometals 2018, 31, 647–671. [Google Scholar] [CrossRef]
- Tian, Y.; Qi, H.; Wang, G.; Li, L.; Zhou, D. Anticancer effect of sodium metavanadate on murine breast cancer both in vitro and in vivo. Biometals 2021, 34, 557–571. [Google Scholar] [CrossRef]
- Kalındemirtaş, F.D.; Kaya, B.; Sert, E.; Şahin, O.; Kuruca, S.E.; Ülküseven, B. New oxovanadium (IV) complexes overcame drug resistance and increased in vitro cytotoxicity by an apoptotic pathway in breast cancer cells. Chem. Biol. Interact. 2022, 363, 109997. [Google Scholar] [CrossRef]
- Ghosh, N.; Chatterjee, S.; Biswal, D.; Pramanik, N.R.; Chakrabarti, S.; Sil, P.C. Oxidative stress imposed in vivo anticancer therapeutic efficacy of novel imidazole-based oxidovanadium(IV) complex in solid tumor. Life Sci. 2022, 301, 120606. [Google Scholar] [CrossRef]
- Ghosh, N.; De, S.; Pramanik, N.R.; Sil, P.C. Multifaceted antineoplastic curative potency of novel water-soluble methylimidazole-based oxidovanadium(IV) complex against triple negative mammary carcinoma. Cell Signal. 2024, 117, 111089. [Google Scholar] [CrossRef] [PubMed]
- Chmur, K.; Tesmar, A.; Zdrowowicz, M.; Rosiak, D.; Chojnacki, J.; Wyrzykowski, D. Exploring the antitumor efficacy of N-heterocyclic nitrilotriacetate oxidovanadium(IV) salts on prostate and breast cancer cells. Molecules 2024, 29, 2924. [Google Scholar] [CrossRef] [PubMed]
- Suma, P.R.; Padmanabhan, R.A.; Telukutla, S.R.; Ravindran, R.; Velikkakath, A.K.G.; Dekiwadia, C.D.; Paul, W.; Laloraya, M.; Srinivasula, S.M.; Bhosale, S.V.; et al. Vanadium pentoxide nanoparticle mediated perturbations in cellular redox balance and the paradigm of autophagy to apoptosis. Free Radic. Biol. Med. 2020, 161, 198–211. [Google Scholar] [CrossRef] [PubMed]
- Kalra, A.; Meltzer, S.J. The Role of DNA Methylation in Gastrointestinal Disease: An Expanded Review of Malignant and Nonmalignant Gastrointestinal Diseases. Gastroenterology 2024, 168, 245–266. [Google Scholar] [CrossRef]
- Wu, Y.; Ma, Y.; Xu, Z.; Wang, D.; Zhao, B.; Pan, H.; Wang, J.; Xu, D.; Zhao, X.; Pan, S.; et al. Sodium orthovanadate inhibits growth of human hepatocellular carcinoma cells in vitro and in an orthotopic model in vivo. Cancer Lett. 2014, 351, 108–116. [Google Scholar] [CrossRef]
- Wang, Q.; Liu, T.T.; Fu, Y.; Wang, K.; Yang, X.G. Vanadium compounds discriminate hepatoma and normal hepatic cells by differential regulation of reactive oxygen species. J. Biol. Inorg. Chem. 2010, 15, 1087–1097. [Google Scholar] [CrossRef]
- Aliabad, H.B.; Falahati-pour, S.K.; Ahmadirad, H.; Mohamadi, M.; Hajizadeh, M.R.; Mahmoodi, M. Vanadium complex: An appropriate candidate for killing hepatocellular carcinoma cancerous cells. BioMetals 2018, 31, 981–990. [Google Scholar] [CrossRef]
- Yang, X.; Xiao, J.; Jiang, L.; Ran, L.; Fan, Y.; Zhang, M.; Xu, Y.; Yao, C.; An, B.; Yang, Y.; et al. A Multifunctional Vanadium-Iron-Oxide Nanoparticle Eradicates Hepatocellular Carcinoma via Targeting Tumor and Endothelial Cells. ACS Appl. Mater. Interfaces 2022, 14, 28514–28526. [Google Scholar] [CrossRef]
- Sarwar, Z.; Abbas, M.K.; Shad, N.A.; Akhtar, K.; Mobeen, A.; Abbas, W.; Abd El-Aziz, K.; Tabassum, M.R.; Zulqarnain, M.; Ali, H.T.; et al. Anticancer and acute toxicity studies of cellulose-coated Vanadium oxide nanomaterials. J. Mol. Struct. 2025, 1322, 140633. [Google Scholar] [CrossRef]
- Sinha, A.; Banerjee, K.; Banerjee, A.; Sarkar, A.; Ahir, M.; Adhikary, A.; Chatterjee, M.; Choudhuri, S.K. Induction of apoptosis in human colorectal cancer cell line, HCT-116 by a vanadium-Schiff base complex. Biomed. Pharmacother. 2017, 92, 509–518. [Google Scholar] [CrossRef]
- León, I.E.; Ruiz, M.C.; Franca, C.A.; Parajón-Costa, B.S.; Baran, E.J. Metvan, bis (4,7-Dimethyl-1,10-phenanthroline) sulfatooxidovanadium(IV): DFT and spectroscopic study—Antitumor action on human bone and colorectal cancer cell lines. Biol. Trace Elem. Res. 2019, 191, 81–87. [Google Scholar] [CrossRef]
- Leon, I.E.; Cadavid-Vargas, J.F.; Tiscornia, I.; Porro, V.; Castelli, S.; Katkar, P.; Desideri, A.; Bollati-Fogolin, M.; Etcheverry, S.B. Oxidovanadium(IV) complexes with chrysin and silibinin: Anticancer activity and mechanisms of action in a human colon adenocarcinoma model. J. Biol. Inorg. Chem. 2015, 20, 1175–1191. [Google Scholar] [CrossRef] [PubMed]
- Naso, L.G.; Badiola, I.; Clavijo, J.M.; Valcarcel, M.; Salado, C.; Ferrer, E.G.; Williams, P.A. Inhibition of the metastatic progression of breast and colorectal cancer in vitro and in vivo in murine model by the oxidovanadium(IV) complex with luteolin. Bioorg. Med. Chem. 2016, 24, 6004–6011. [Google Scholar] [CrossRef] [PubMed]
- Roy, S.; Chakraborty, T. Deciphering the molecular mechanism and apoptosis underlying the in vitro and in vivo chemotherapeutic efficacy of vanadium luteolin complex in colon cancer. Cell Biochem. Funct. 2018, 36, 116–128. [Google Scholar] [CrossRef] [PubMed]
- Xu, R. Salvia officinalis-based green-mediated vanadium nanoparticles: Describing a modern chemotherapeutic drug for the treatment of colorectal carcinoma. Authorea Prepr. 2022. [Google Scholar] [CrossRef]
- Rehman, S.; Alahmari, F.; Aldossary, L.; Alhout, M.; Aljameel, S.S.; Ali, S.M.; Sabir, J.S.M.; Khan, F.A.; Rather, I.A. Nano-sized warriors: Zinc chromium vanadate nanoparticles as a dual solution for eradicating waterborne enterobacteriaceae and fighting cancer. Front. Pharmacol. 2023, 14, 1213824. [Google Scholar] [CrossRef]
- Cheng, Q.; Chen, Y.; Zou, D.; Li, Q.; Shi, X.; Qin, Q.; Liu, M.; Wang, L.; Wang, Z. Targeting Metabolic Adaptation of Colorectal Cancer with Vanadium-Doped Nanosystem to Enhance Chemotherapy and Immunotherapy. Adv. Sci. 2025, 12, 2409329. [Google Scholar] [CrossRef]
- Sochacka-Ćwikła, A.; Mączyński, M.; Regiec, A. FDA-Approved Drugs for Hematological Malignancies—The Last Decade Review. Cancers 2022, 14, 87. [Google Scholar] [CrossRef]
- Milaeva, E.R. Application of Metal Compounds in Medicine. Russ. J. Coord. Chem. 2024, 50, 1043–1123. [Google Scholar] [CrossRef]
- Šebestová, L.; Havelek, R.; Řezáčová, M.; Honzíček, J.; Kročová, Z.; Vinklárek, J. Study of antitumor effect of selected vanadium and molybdenum organometallic complexes in human leukemic T-cells. Chem. Biol. Interact. 2015, 242, 61–70. [Google Scholar] [CrossRef]
- Ta, S.; Ghosh, M.; Ghosh, K.; Brandão, P.; Félix, V.; Hira, S.K.; Manna, P.P.; Das, D. Exploring Anticancer and (Bio)catalytic Activities of New Oxovanadium(V), Dioxomolybdenum(VI), and Copper(II) Complexes of Amide-Imine Conjugates. ACS Appl. Bio Mater. 2019, 2, 2802–2811. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Wei, N.; Guan, G.; Song, T.; Xu, Y.; Wang, S.; Zhou, J. Sodium orthovanadate inhibits growth of acute leukemia HL60 cells and HL60/A cells in vitro. Biosci. Rep. 2020, 40, BSR20201918. [Google Scholar] [CrossRef] [PubMed]
- Mirjalili, S.; Khaleghian, A.; Kalalinia, F. Effects of co-administration of arsenic trioxide and Schiff base oxovanadium complex on the induction of apoptosis in acute promyelocytic leukemia cells. Biometals 2021, 34, 1067–1080. [Google Scholar] [CrossRef] [PubMed]
- Sunitha, N.; Isac Sobana, R.C.; Sindhu, K.B. Development of nanofunctionalized oxovanadium(IV) complex and its anticancer, antidiabetic, DNA cleavage and cell imaging studies. Int. J. Pharm. 2023, 644, 123339. [Google Scholar] [CrossRef]
- Roky, A.H.; Islam, M.M.; Ahasan, A.M.F.; Mostaq, M.S.; Mahmud, M.Z.; Amin, M.N.; Mahmud, M.A. Overview of skin cancer types and prevalence rates across continents. Cancer Pathog. Ther. 2025, 3, 89–100. [Google Scholar] [CrossRef]
- Rozzo, C.; Sanna, D.; Garribba, E.; Serra, M.; Cantara, A.; Palmieri, G.; Pisano, M. Antitumoral effect of vanadium compounds in malignant melanoma cell lines. J. Inorg. Biochem. 2017, 174, 14–24. [Google Scholar] [CrossRef]
- De Sousa-Coelho, A.L.; Aureliano, M.; Fraqueza, G.; Serrão, G.; Gonçalves, J.; Sánchez-Lombardo, I.; Link, W.; Ferreira, B. Decavanadate and metformin-decavanadate effects in human melanoma cells. J. Inorg. Biochem. 2022, 235, 111915. [Google Scholar] [CrossRef]
- Pisano, M.; Arru, C.; Serra, M.; Galleri, G.; Sanna, D.; Garribba, E.; Palmieri, G.; Rozzo, C. Antiproliferative activity of vanadium compounds: Effects on the major malignant melanoma molecular pathways. Metallomics 2019, 11, 1687–1699. [Google Scholar] [CrossRef]
- Das, S.; Roy, A.; Barui, A.K.; Alabbasi, M.M.A.; Kuncha, M.; Sistla, R.; Patra, C.R. Anti-angiogenic vanadium pentoxide nanoparticles for the treatment of melanoma and their in vivo toxicity study. Nanoscale 2020, 12, 7604–7621. [Google Scholar] [CrossRef]
- Nivetha, S.; Srivalli, T.; Sathya, P.M.; Mohan, H.; Karthi, N.; Muralidharan, K.; Ramalingam, V. Nickel-doped vanadium pentoxide (Ni@ V2O5) nanocomposite induces apoptosis targeting PI3K/AKT/mTOR signaling pathway in skin cancer: An in vitro and in vivo study. Colloids Surf. B Biointerfaces 2024, 234, 113763. [Google Scholar] [CrossRef]
- Amante, C.; De Sousa-Coelho, A.L.; Aureliano, M. Vanadium and melanoma: A systematic review. Metals 2021, 11, 828. [Google Scholar] [CrossRef]
- Biswas, B.K.; Biswas, N.; Saha, S.; Rahaman, A.; Mandal, D.P.; Bhattacharjee, S.; Sepay, N.; Zangrando, E.; Garribba, E.; Roy-Choudhury, C. Interaction with bioligands and in vitro cytotoxicity of a new dinuclear dioxido vanadium(V) complex. J. Inorg. Biochem. 2022, 237, 111980. [Google Scholar] [CrossRef]
- Mato-López, L.; Sar-Rañó, A.; Fernández, M.R.; Díaz-Prado, M.L.; Gil, A.; Sánchez-González, Á.; Fernández-Bertólez, N.; Méndez, J.; Valdiglesias, V.; Avecilla, F. Relationship between structure and cytotoxicity of vanadium and molybdenum complexes with pyridoxal derived ligands. J. Inorg. Biochem. 2022, 235, 111937. [Google Scholar] [CrossRef]
- Mukhamedov, N.; Wubulikasimu, A.; Rustamova, N.; Nuerxiati, R.; Mirzaakhmedov, S.; Ishimov, U.; Ziyavitdinov, J.; Yili, A.; Aisa, H.A. Synthesis and Characterization of Novel Chickpea Protein Hydrolysate-Vanadium Complexes Having Cell Inhibitory Effects on Lung Cancer A549 Cells Lines. Protein J. 2021, 40, 721–730. [Google Scholar] [CrossRef] [PubMed]
- Melounková, L.; Machálková, A.; Havelek, R.; Honzíček, J.; Řezáčová, M.; Císařová, I.; Peterová, E.; Vinklárek, J. Vanadocene complexes bearing N,N’-chelating ligands: Synthesis, structures and in vitro cytotoxic studies on the A549 lung adenocarcinoma cell line. J. Inorg. Biochem. 2019, 195, 182–193. [Google Scholar] [CrossRef] [PubMed]
- Ribeiro, N.; Bulut, I.; Pósa, V.; Sergi, B.; Sciortino, G.; Pessoa, J.C.; Maia, L.B.; Ugone, V.; Garribba, E.; Enyedy, É.A.; et al. Solution chemical properties and anticancer potential of 8-hydroxyquinoline hydrazones and their oxidovanadium(IV) complexes. J. Inorg. Biochem. 2022, 235, 111932. [Google Scholar] [CrossRef] [PubMed]
- Guerrero-Palomo, G.; Rendón-Huerta, E.P.; Montaño, L.F.; Fortoul, T.I. Vanadium compounds and cellular death mechanisms in the A549 cell line: The relevance of the compound valence. J. Appl. Toxicol. 2019, 39, 540–552. [Google Scholar] [CrossRef]
- Liu, Z.; Zhang, Z.; Du, X.; Liu, Y.; Zhang, Z. Formulation of a novel anti-lung cancer drug: Vanadium nanoparticles containing Salvia officinalis. Inorg. Chem. Commun. 2023, 150, 110520. [Google Scholar] [CrossRef]
- Lopez-Valdez, N.; Rojas-Lemus, M.; Fortoul, T.I. The Effect of Vanadium Inhalation on the Tumor Progression of Urethane-Induced Lung Adenomas in a Mice Model. Inorganics 2021, 9, 78. [Google Scholar] [CrossRef]
- Suwalsky, M.; Fierro, P.; Villena, F.; Gallardo, M.J.; Jemiola-Rzeminska, M.; Strzalka, K.; Gul-Hinc, S.; Ronowska, A.; Zysk, M.; Szutowicz, A. Effects of sodium metavanadate on in vitro neuroblastoma and red blood cells. Arch. Biochem. Biophys. 2013, 535, 248–256. [Google Scholar] [CrossRef]
- Tian, X.; Fan, J.; Hou, W.; Bai, S.; Ao, Q.; Tong, H. Sodium orthovanadate induces the apoptosis of SH-SY5Y cells by inhibiting PIWIL2. Mol. Med. Rep. 2016, 13, 874–880. [Google Scholar] [CrossRef]
- Zhang, Y.; Wang, L.; Zeng, K.; Wang, K.; Yang, X. Vanadyl complexes discriminate between neuroblastoma cells and primary neurons by inducing cell-specific apoptotic pathways. J. Inorg. Biochem. 2018, 188, 76–87. [Google Scholar] [CrossRef] [PubMed]
- Bates, A.C.; Klugh, K.L.; Galaeva, A.O.; Patch, R.A.; Manganaro, J.F.; Markham, S.A.; Scurek, E.; Levina, A.; Lay, P.A.; Crans, D.C. Optimizing Therapeutics for Intratumoral Cancer Treatments: Antiproliferative Vanadium Complexes in Glioblastoma. Int. J. Mol. Sci. 2025, 26, 994. [Google Scholar] [CrossRef] [PubMed]
- Xu, S.; Liu, H.; Li, X.; Zhao, J.; Wang, J.; Crans, D.C.; Yang, X. Approaches to selective and potent inhibition of glioblastoma by vanadyl complexes: Inducing mitotic catastrophe and methuosis. J. Inorg. Biochem. 2024, 257, 112610. [Google Scholar] [CrossRef] [PubMed]
- Ferguson, J.L.; Turner, S.P. Bone Cancer: Diagnosis and Treatment Principles. Am. Fam. Physician AFP 2018, 98, 205–213. [Google Scholar]
- Ścibior, A.; Pietrzyk, L.; Plewac, Z.; Skibad, P. Vanadium: Risks and possible benefits in the light of a comprehensive overview of its pharmacotoxicological mechanisms and multi-applications with a summary of further research trends. J. Trace Elem. Med. Biol. 2020, 61, 126508. [Google Scholar] [CrossRef]
- León, I.; Cadavid-Vargas, J.F.; Resasco, A.; Maschi, F.; Ayala, M.A.; Carbone, C.; Etcheverry, S.B. In vitro and in vivo antitumor effects of the VO-chrysin complex on a new three-dimensional osteosarcoma spheroids model and a xenograft tumor in mice. J. Biol. Inorg. Chem. 2016, 21, 1009–1020. [Google Scholar] [CrossRef]
- Balsa, L.M.; Quispe, P.; Baran, E.J.; Lavecchia, M.J.; LeoÅLn, I.E. In silico and in vitro analysis of FAK/MMP signaling axis inhibition by VO-clioquinol in 2D and 3D human osteosarcoma cancer cells. Metallomics 2020, 12, 1931–1940. [Google Scholar] [CrossRef]
- Kalniņa, D.; Levina, A.; Pei, A.; Gross, K.A.; Lay, P.A. Synthesis, characterization and in vitro anti-cancer activity of vanadium-doped nanocrystalline hydroxyapatite. New J. Chem. NJC 2019, 43, 17891–17901. [Google Scholar] [CrossRef]
- Cacicedo, M.L.; Ruiz, M.C.; Scioli-Montoto, S.; Ruiz, M.E.; Fernandez, M.A.; Torres-Sanchez, R.M.; Baran, E.J.; Castro, G.R.; Leon, I.E. Lipid nanoparticles–Metvan: Revealing a novel way to deliver a vanadium compound to bone cancer cells. New J. Chem. NJC 2019, 43, 17726–17734. [Google Scholar] [CrossRef]
- Liu, X.; Zhang, P.; Xu, M.; Zhao, Z.; Yin, X.; Pu, X.; Wang, J.; Liao, X.; Huang, Z.; Cao, S.; et al. Mixed-valence vanadium-doped mesoporous bioactive glass for treatment of tumor-associated bone defects. J. Mater. Chem. B 2025, 13, 3138–3160. [Google Scholar] [CrossRef] [PubMed]
- Lu, L.P.; Suo, F.Z.; Feng, Y.L.; Song, L.L.; Li, Y.; Li, Y.J.; Wang, K.T. Synthesis and biological evaluation of vanadium complexes as novel anti-tumor agents. Eur. J. Med. Chem. 2019, 176, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Mirjalili, S.; Dejamfekr, M.; Moshtaghian, A.; Salehi, M.; Behzad, M.; Khaleghian, A. Induction of cell cycle arrest in MKN45 cells after schiff base oxovanadium complex treatment using changes in gene expression of CdC25 and P53. J. Drug Res. 2020, 70, 545–551. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Jiang, X.G.; Bai, Y.P.; Li, H.; Gao, L.X.; Zhang, T.; Dong, F.; Ding, D.; Zhang, Y. SOV sensitizes gastric cancer cells to radiation by suppressing radiation-induced autophagy in vitro and in vivo. Tissue Cell 2023, 82, 102109. [Google Scholar] [CrossRef]
- Wu, J.X.; Hong, Y.H.; Yang, X.G. Bis (acetylacetonato)-oxidovanadium(IV) and sodium metavanadate inhibit cell proliferation via ROS-induced sustained MAPK/ERK activation but with elevated AKT activity in human pancreatic cancer AsPC-1 cells. JBIC J. Biol. Inorg. Chem. 2016, 21, 919–929. [Google Scholar] [CrossRef]
- Kowalski, S.; Hać, S.; Wyrzykowski, D.; Zauszkiewicz-Pawlak, A.; Inkielewicz-Stępniak, I. Selective cytotoxicity of vanadium complexes on human pancreatic ductal adenocarcinoma cell line by inducing necroptosis, apoptosis and mitotic catastrophe process. Oncotarget 2017, 8, 60324. [Google Scholar] [CrossRef]
- Kowalski, S.; Tesmar, A.; Sikorski, A.; Inkielewicz-Stepniak, I. Oxidovanadium(IV) complex disrupts mitochondrial membrane potential and induces apoptosis in pancreatic cancer cells. Anti-Cancer Agents Med. Chem.-Anti-Cancer Agents 2021, 21, 71–83. [Google Scholar] [CrossRef]
- Griffin, E.; Levina, A.; Lay, P.A. Vanadium (V) tris-3,5-di-tert-butylcatecholato complex: Links between speciation and anti-proliferative activity in human pancreatic cancer cells. J. Inorg. Biochem. 2019, 201, 110815. [Google Scholar] [CrossRef]
- Nair, R.S.; Kuriakose, M.; Somasundaram, V.; Shenoi, V.; Kurup, M.P.; Srinivas, P. The molecular response of vanadium complexes of nicotinoyl hydrazone in cervical cancers—A possible interference with HPV oncogenic markers. Life Sci. 2014, 116, 90–97. [Google Scholar] [CrossRef]
- Inamdar, P.R.; Sheela, A. Exploration of DNA binding mode, chemical nuclease, cytotoxic and apoptotic potentials of diketone based oxovanadium (IV) complexes. Int. J. Biol. Macromol. 2015, 76, 269–278. [Google Scholar] [CrossRef]
- Dankhoff, K.; Ahmad, A.; Weber, B.; Biersack, B.; Schobert, R. Anticancer properties of a new non-oxido vanadium (IV) complex with a catechol-modified 3,3′-diindolylmethane ligand. J. Inorg. Biochem. 2019, 194, 1–6. [Google Scholar] [CrossRef]
- Bai, Y.; Zhang, H.; Wang, Y.; Zhu, L.; Shi, T.; Wei, H.; Xiao, J.; Zhang, Y.; Wang, Z. Novel oxovanadium complex VO (hntdtsc)(NPIP): Anticancer activity and mechanism of action on HeLa cells. Front. Pharmacol. 2021, 11, 608218. [Google Scholar] [CrossRef]
- Reytman, L.; Braitbard, O.; Hochman, J.; Tshuva, E.Y. Highly effective and hydrolytically stable vanadium (V) amino phenolato antitumor agents. Inorg. Chem. 2016, 55, 610–618. [Google Scholar] [CrossRef] [PubMed]
- Lujerdean, C.; Zăhan, M.; Dezmirean, D.S.; Ștefan, R.; Simedru, D.; Damian, G.; Vedeanu, N.S. In Vitro Studies Demonstrate Antitumor Activity of Vanadium Ions from a CaO-P2O5-CaF2: V2O5 Glass System in Human Cancer Cell Lines A375, A2780, and Caco-2. Int. J. Mol. Sci. 2023, 24, 1149. [Google Scholar] [CrossRef] [PubMed]
- Levina, A.; Crans, D.C.; Lay, P.A. Speciation of metal drugs, supplements and toxins in media and bodily fluids controls in vitro activities. Coord. Chem. Rev. 2017, 352, 473–498. [Google Scholar] [CrossRef]
- Nunes, P.; Correia, I.; Cavaco, I.; Marques, F.; Pinheiro, T.; Avecilla, F.; Pessoa, J.C. Therapeutic potential of vanadium complexes with 1,10-phenanthroline ligands, quo vadis? Fate of complexes in cell media and cancer cells. J. Inorg. Biochem. 2021, 217, 111350. [Google Scholar] [CrossRef]
- Pessoa, J.C.; Correia, I. Misinterpretations in Evaluating Interactions of Vanadium Complexes with Proteins and Other Biological Targets. Inorganics 2021, 9, 17. [Google Scholar] [CrossRef]
- Scior, T.; Guevara-Garcia, A.J.; Do, Q.-T.; Bernard, P.; Laufer, S. Why Antidiabetic Vanadium Complexes are Not in the Pipeline of “Big Pharma” Drug Research? A Critical Review. Curr. Med. Chem. 2016, 23, 2874–2891. [Google Scholar] [CrossRef]
- Dinda, R.; Garribba, E.; Sanna, D.; Crans, D.C.; Costa Pessoa, J. Hydrolysis, ligand exchange, and redox properties of vanadium compounds: Implications of solution transformation on biological, therapeutic, and environmental applications. Chem. Rev. 2025, 125, 1468–1603. [Google Scholar] [CrossRef]
- Fugono, J.; Yasui, H.; Sakurai, H. Enteric-coating capsulation of insulinomimetic vanadyl sulfate enhances bioavailability of vanadyl species in rats. J. Pharm. Pharmacol. 2002, 54, 611–615. [Google Scholar] [CrossRef]
- Sakurai, H.; Fugono, J.; Yasui, H. Pharmacokinetic Study and Trial for Preparation of Enteric-Coated Capsule Containing Insulinomimetic Vanadyl Compounds: Implications for Clinical Use. Mini-Rev. Med. Chem. 2004, 4, 41–48. [Google Scholar] [CrossRef]
- Sakurai, H.; Katoh, A.; Yoshikawa, Y. Chemistry and Biochemistry of Insulin-Mimetic Vanadium and Zinc Complexes. Trial for Treatment of Diabetes Mellitus. Bull. Chem. Soc. Jpn. 2006, 79, 1645–1664. [Google Scholar] [CrossRef]
Metal Compounds | Biologic Effects | Advantages | Limitations |
---|---|---|---|
Copper (Cu) complexes | Cytotoxic activity, mitochondrial dysfunction, ROS production, apoptosis activation and cell migration inhibition [5,9]. | More easily absorbed by tumor cells than by normal cells, tolerable toxicity and good selectivity towards tumor cells instead of non-tumor cells [5,9]. | Angiogenesis and metastasis are greatly aided by copper [9]. |
Gold (Au) complexes | DNA damage, mitochondrial disfunction, autophagy, potential antimetastatic effect, tubulin polymerization inhibition, ROS production, cell cycle arrest, apoptosis, topoisomerase inhibition and specific kinase inhibition [9,12,13]. | Gold is a biocompatible and inert metal. In vitro and in vivo antitumor properties towards several human cancer cells [12,14]. | Instability of ligands in Au complexes, low solubility, toxicity against normal cells, drug resistance [9,12]. |
Iridium (Ir) complexes | Cytotoxicity, ROS production, mitochondrial dysfunction and apoptosis [5,9]. | Water tolerance, fewer adverse effects, complexes can form wide range of versatile structures, higher kinetic degree of ligand substitution, efficacy against cisplatin resistant cells [5,9,15]. | The mechanisms by which Ir compounds exert their effects are not yet fully elucidated, as studies on their antitumor activity are still in the preliminary stages [9]. |
Iron (Fe) complexes | ROS production, cytotoxicity, DNA damage, disturbance of iron homeostasis, [5,16]. | As an essential element, this metal has specific physiological processes for uptake and excretion. Variety in structures of its complexes, elevated anticancer activities against a broad range of cancer cell lines, negligible effects on noncancerous cells [16]. | Interactions with endogenous iron and disruptions of iron homeostasis occur in the case of labile complexes [16]. |
Rhenium (Re) complexes | DNA damage, cytotoxic activity, ROS production and apoptosis [17]. | Various oxidation states enables the formation of diverse ligand types and coordination structures [17]. | The use of Re in cancer therapy is still in its early stages, and the mechanisms of action of these compounds are not yet fully understood. A limited number of studies have examined the in vivo applicability of Re compounds [17]. |
Ruthenium (Ru) complexes | Tumor cell cycle arrest, apoptosis, ROS production, protein kinases inhibition, angiogenesis inhibition [9,12,18]. | Slow ligand exchange kinetics, lower toxicity, selective cytotoxic activity in vitro against cancer cells. Several examples of Ru complexes exhibiting in vivo anticancer activity; real clinical potential. Efficacy against drug-resistant malignancies [9,18]. | Not all mechanisms of action have been elucidated. The effect on metastasis has been poorly studied [9,18]. |
Osmium (Os) complexes | Protein inhibition (such es Hypoxia inducible factor—HIF), DNA interactions, antiproliferative activities, ROS production and cell death [5,9,19]. | Osmium’s higher oxidation states are stable, antineoplastic potential both in vitro and in vivo, no cross-resistance with cisplatin in cancer cells [9,19]. | The mechanisms of action of Os compounds are poorly understood, as their antitumor effectiveness has been studied less extensively than that of other metallic complexes [19]. |
Zinc (Zn) complexes | ROS production, DNA fragmentation and cytotoxicity [20,21]. | As an essential metal, there are biological mechanisms for its absorption and elimination, and it is less toxic at higher doses compared to other metals such as Fe and Cu [12,20,21]. | Interactions with endogenous zinc and disruptions of zinc homeostasis occur in the case of labile complexes [20,21]. |
Cancer Model | Vanadium Compound | Dose/Duration | Biological Effects | Mechanisms | References |
---|---|---|---|---|---|
MCF-7 Human breast cancer cell line | Vanadium-quercetin complex | 125, 200 and 275 μM of V-quercetin complex for 12, 24 and 48 h of treatment | Pro-apoptotic effects | Upregulating p53, caspase 3 and 9, downregulating Akt, mTOR and VEGF expressions and DNA fragmentation | [40] |
Imidazole-based oxidovanadium(IV) complex (VOL). | 10–40 µg/mL of VOL for 72 h of treatment | Cytotoxicity | Apoptosis; ROS production, imbalance of catalase and SOD activity; lipid peroxidation. | [43] | |
Four N-heterocyclic nitrilotriacetate oxidovanadium(IV) salts | 0.1–200 µM for the salts for 48 h of treatment | Cytotoxicity, Genotoxicity, cell cycle disruption | DNA cleavage and cell cycle block in the G0/G1 and G2/M phase. | [45] | |
4T1 Murine breast cancer cell line | NaVO3 | 1, 10, 50 and 100 μM of NaVO3 for 12, 24 and 48 h of treatment | Proliferation inhibition | Increasing reactive oxygen species (ROS) levels in a concentration-dependent way, arrest cells at G2/M phase, diminish the mitochondrial membrane potential (MMP), promote the progress of apoptosis | [41] |
MDA-MB-231 Human breast cancer cell line | Vanadium pentoxide nanoparticles (VnNp) | 50 µM of VnNp for 24–48 h of treatment | Ultrastructural alterations, cell death, alterations in cell proliferation and migration. | Increased ROS production and decreased superoxide dismutase (SOD) levels. Cell cycle arrest in the G2/M phase. Apoptosis. Autophagosomes and VnNp are observed in the cytoplasm and mitochondria. | [46] |
Methylimidazole-based oxidovanadium(IV) complex (OVMI) | 0, 10, 201 and 30 μg/mL of OVMI for 24 h of treatment | Cytotoxicity, retarded cell proliferation. | ROS-mediated mitochondrial apoptotic death | [44] | |
MDA-MB-231 and MCF-7. Human breast cancer cell lines | Oxidovanadium compounds (VO1a and VO1b). | MDA-MB-231 cells treated (72 h) with 7.2 µM VO1a or 4.7 µM VO1b; and MCF-7 cells treated (72 h) with 10.7 µM VO1a or 7.3 µM VO1b. | Cytotoxicity Inhibition of multidrug resistance. | Apoptosis cell death. Decreasing ABCG2 transporter activity leads to multidrug resistance and apoptosis. | [42] |
HepG2, SK-Hep-1 and Hep3B Human hepatocellular carcinoma cell line | SOV | 7.5–30 µM of SOV for 48 h of treatment | Suppression of cell proliferation and cell death | Decreasing Ki67-positive cells, cell cycle arrest at G2/M phase, down-regulation of pro-caspase-9 and 3, increasing cleaved poly(ADP-Ribose) polymerase (PARP) expression, increased apoptosis and inhibition of autophagy | [48] |
HepG2 Human hepatocellular carcinoma cell line | NaVO3, Bis(maltolato)-oxidovanadium(IV) (VO(ma)2) and Bis(acetylacetonato)-oxidovanadium(IV) (VO(acac)2) | 50, 100 and 400 µM of NaVO3, VO(ma)2 and VO(acac)2 for 4, 8, 12, 16, 24 and 72 h of treatment | Growth inhibition Selectivity of V compounds by tumor cells. | Cell cycle arrest in G1/S phase. Activation of MAPK (ERK) pathway and increase in ROS levels. | [49] |
4-bromo-2-(((5-chloro-2-hydroxyphenyl) imino) methyl) phenol ([IV(L)] complex) | 79 and 100 µg/m L of [IV(L)] complex for 24 and 48 h of treatment | Apoptosis and necrosis | Early apoptosis; late apoptosis/necrosis | [50] | |
Hepa 1-6 Murine hepatoma cell line | Vanadium (V)-iron-oxide (ION) nanoparticle (VIO) | 5, 10, 20, 40, 80, 160 and 200 µg/m Lof (VIO) for 24 h of treatment | Cell viability inhibition | Increased levels of ROS, apoptosis and ferroptosis. | [51] |
HuH-7.0 Human hepatocellular carcinoma cell line | V2O5 nanoparticles (NPs) and cellulose-coated vanadium oxide NPs. | 25, 50, 75 and 100 µg/mL of NPs for 72 h of treatment. | Alteration of cell morphology and decreased viability. | The collapse of the cytoskeleton is proposed. | [52] |
HCT-116 Human colorectal carcinoma cell line | Vanadyl N-(2-hydroxyacetophenone) glycinate [VO(NG)2] | 20 μg/mL of VO(NG)2 for 24, 48 and 72 h of treatment. | Cell cycle arrest in G2/M and cytotoxicity | Depletion of GSH levels, increasing ROS production, oxidative stress and apoptosis. | [53] |
Zinc Chromium Vanadate nanoparticles (VCrZnO4 NPs) | 2.0–75 μg/mL of VCrZnO4 NPs for 48 h of treatment | Cytotoxicity | Apoptosis | [59] | |
HT-29 Human colorectal carcinoma cell line | Bis(4,7-dimethyl-1,10-phenantroline)sulfatooxidovanadium(IV), Metvan | 0.25–5.0 μM of Metvan for 24 h of treatment | Cell viability inhibition | Inhibition of mitochondrial metabolism | [54] |
Na2[VO(silibinin)2 ]· 6H2 O (VOsil) and chrysin [VO(chrysin)2EtOH]2 (VOchrys) | 2.5–100 μM of the complexes for 24 and 48 h of treatment. | Cytotoxicity and cell death | Exposure to VOcrhrys caused cell cycle arrest in G2/M. Exposure to VOsil activated caspase 3 and cell apoptosis. | [55] | |
Vanadium-luteolin complex | 5, 15 and 25 μM of the V-luteolin complex for 24, 36 and 48 h of treatment | Proliferation reduction and cell death. y apoptosis | Apoptosis, activation of p53, Bax, caspase-3 and downregulation of Bcl2 and mTOR/Akt | [57] | |
CT26 Murine colorectal carcinoma cell line | Flavonoid luteolin and oxidovanadium(IV) complex [VO(lut)(H2O)2]Na3H2O (VOlut) | 8.5 μM of VOlut for 5 and 25 h of treatment | Cytotoxicity | The mechanism of action was not explored in the study. | [56] |
Caco-2, COLO 320, DLD-1, HCT-15, HCT-116 and HT-29 Human colorectal carcinoma cell lines | Salvia officinalis-based, green-mediated vanadium nanoparticles (VNPs@Salvia officinalis) | 100–300 μg/mL of VNPs@Salvia officinalis for 48 h of treatment | Cell viability reduction, cytotoxicity | Antioxidant activity of nanoparticles | [58] |
CT26 and MC38 Murine colorectal carcinoma cell line | Vanadium chloride (VCl3) | 40 μg/mL of VCl3 for 24 h of treatment | Suppression of cell proliferation | Inhibition of the activity of several enzymes (G6PD, LDH, HK, and PK), blocking glucose metabolism. | [60] |
Vanadium and glutaminase inhibitors (bis 2-(5-phenylacetamido- 1,2,4-thiadiazol-2-yl) ethyl sulfide, BPTES) VSi-BP | 40 μg/mL of for 24 h of treatment | Suppression of cell proliferation, reduction in viability and cell death | Inhibition of the activity of several enzymes (G6PD, LDH, HK, and PK), blocking glucose metabolism, apoptosis, decreasing intracellular GSH levels and increasing ROS | ||
MOLT-4 Human acute T-lymphoblastic leukemia cells Jurkat Human T-cell leukemia cells | [(η5-C5H5)2V(5-NH2-phen)]OTf (V1) | 1, 2, 3, 5 and 7 μM/L of V1 in MOLT-4 cells for 24, 48 and 72 h of treatment 1, 4, 8, 12 and 20 μM/L V1 in Jurkat cells for 24, 48 and 72 h of treatment | Cytotoxicity, inhibition of proliferation and cell death | Activation of intrinsic and extrinsic apoptosis pathways, with increased levels of caspases 8 and 9. Induction and activation of p53 | [63] |
K-562 Human leukemia cells 2PK3 Mouse lymphoma cells | (E)-N′-((2-hydroxynaphthalen-1-yl) methylene)-4-methylbenzohydrazide (PTANAP) conjugated with oxidovanadium [V(PTANAP)(μ2-MeO)]2 (V1) | 10, 25, 50 and 100 μg/mL of V1 for 24 and 48 h of treatment | Cytotoxicity Inhibition of proliferation, cell death | ROS generation, DNA damage and apoptosis | [64] |
HL-60 Acute promyelocytic leukemia cells | SOV | 5, 10 and 20 μM of SOV for 72 h of treatment | Cytotoxicity Inhibition of proliferation Cell death | Caspases 3 and 9 activation (mitochondrial apoptosis), activation of poly-ADP-ribose polymerase (PARP). G2/M cell cycle arrest. Inhibition of autophagy, which promotes apoptosis. | [65] |
Schiff base oxidovanadium complex individually or simultaneously administered with arsenic trioxide | 1, 10 and 40 μg/mL of Schiff base oxidovanadium complex | Simultaneous exposure to arsenic trioxide: Increased cytotoxicity, cell death | p21 and p53 induction and apoptosis | [66] | |
K-562 Human leukemia cells | Vanadyl complex [VO(CUAP)SO4] Nano Cu-VO(IV) complex | 10, 20, 40 and 80 μg/mL | Cytotoxicity Inhibition of proliferation. The vanadyl complex showed greater anticarcinogenic effect. | ROS generation and DNA damage | [67] |
A375 and CN-mel Malignant melanoma cell lines | NaVO3 and three oxidovanadium complexes (VS2, VS3 and VS4). | 10 and 20 μM of VN and the three complexes for 24, 48 h and 72 h of treatment | Antiproliferative activity and cytotoxicity | Apoptosis induction and cell cycle arrest in G2/M phase | [69] |
NaVO3 and oxidovanadium complex [VO(dhp)2] VS2 | 10 and 20 μM of VN and VS2 complex for 24, 48 h and 72 h of treatment | Cell death | Increasing ROS levels, inactivation of the MAPK pathway, increased p21Cip1 inhibitor and apoptosis | [71] | |
B16F10 Murine melanoma cell line | Vanadium pentoxide nanoparticles (V2O5 NPs 30–60 nm) | 1–50 μg mL of V2O5 NPs | Antiproliferative activity | Generation of intracellular ROS, increased p53, and decreased survivin in cancer cells | [72] |
Vanadium pentoxide nanoparticles (V2O5 NPs) and nickel doped vanadium pentoxide nanoparticles (Ni@V2O5 NPs) | V2O5 (0–40 μg/mL) and Ni@V2O5 (0–20 μg/mL) for 24 h of treatment | Proliferation inhibition and cytotoxicity | Mitochondrial ROS generation, mitochondrial and nuclear damage, apoptosis | [73] | |
UACC-62 Human melanoma cell line | Decavanadate (V10) and metformin-decavanadate (Metf-V10) | 0 and 20 μM of V10 and Metf-V10 for 24, 48 and 72 h of treatment | Decrease in cellular viability and antiproliferative effects. | Cell cycle arrest in G2/M phase, ERK and AKT signaling pathways hyperactivation | [70] |
A549 Human lung adenocarcinoma cell line | Organometallic complexes of vanadocene with N,N′-chelating ligands | 1–60 μM complexes for 24, 48 and 72 h of treatment | Cytotoxicity and antiproliferative activity in a dose dependent manner | Apoptosis mediated by activation of caspase-8,-3 and-7. G2 and S phase cell cycle arrest, modification in the expression of proteins that regulate the cell cycle such as: Chk1, Chk2, ERK1/2 and p21. | [78] |
VOSO4 and NaVO3 | 0–100 µM of VOSO4 and NaVO3 for 24 and 48 h of treatment | Cell death and inhibition of proliferation | Increasing in ROS production. | [80] | |
Vanadyl sulfate complexes with Cicer arietium L. (CPH-V Complexes) | 1–30 µg/mL of CPH-V Complexes for 48 h of treatment | Antiproliferative activity in a concentration dependent manner | Cell growth inhibition | [77] | |
Bis(μ-oxido)-bridged vanadium(V) dimer with a Schiff base ligand (pedf) [(VVO2)2(pedf)2] | 2–80 µg/m L of [(VVO2)2(pedf)2] for 24 h of treatment | Antiproliferative activity in a concentration dependent manner, decreased cell viability and DNA damage. | Loss of membrane integrity, DNA cleavage and apoptosis regulated by modification in p53, PPAR and PCNA expression | [75] | |
Vanadium complexes with pyridoxal-derived ligands | 0.1–500 µM for 24 h of treatment | Antiproliferative activity in a concentration dependent manner | The mechanism of action was not explored in the study. | [76] | |
Oxidovanadium(IV) complexes | 0.78–50 µM for 24 h of treatment | Decreased cell viability and concentration-dependent antiproliferative activity | Apoptosis mediated by activation of caspase-3 and -7. Increased ROS. | [79] | |
SN56 Murine neuroblastoma cell line | NaVO3 | 50–200 µM for 1 h of treatment | Concentration-dependent decrease in viability and morphological changes | Loss of membrane integrity. Loss of cellular arborization, resulting in a rounded structure with vacuoles inside. | [83] |
SH-SY5Y Human neuroblastoma cell line | SOV | 0.05–50 µM of SOV for 24, 48 and 72 h of treatment. | Concentration- and time-dependent cell death. Reduction in cell proliferation. | Apoptosis associated with reduced Bcl-2 protein expression. Cell cycle arrest in G2/M and S phase and inhibition of PIWIL2. | [84] |
Bis(acetylacetonato) oxidovanadium(IV) ([VO(acac)2]) | 12.5–400 µM of [VO(acac)2] for 24 and 48 h of treatment. | Concentration- and time-dependent cell death. Cell cycle dysregulation | Increased ROS production and cell apoptosis. Arrest in different cell cycle phases associated with Cyclin D1 levels, expression in GSK-3 β, Akt/PKB and Rb phosphorylation levels. | [85] | |
SH-SY5Y Human neuroblastoma cell line U251 Human glioblastoma cell line GL261 Murine glioblastoma cell line | 2–16 µM of (VO(acac)2) for 24 h of treatment | Inhibitory effect on concentration-dependent cell viability, cell cycle suppression, micropinocytosis-derived cytoplasmic vacuole formation, energy deficiency and cell death. | Decreased cell viability, cell cycle arrest in the G2/M phase, low intracellular ATP levels, a decrease in mTOR and P70S6K and an increase in AMPK. Death by the process of Metuosis upon initiation of Rac-MKK-JNK pathway activation. | [87] | |
T98G Human glioblastoma cell line | Oxidovanadium(IV) Schiff base–catecholate complex [VO(3-tBuHSHED)(TIPCAT)] | 0.1–100 µM of [VO(3-tBuHSHED)(TIPCAT)] for 24 and 72 h of treatment | Decreased cell viability. Concentration-dependent antiproliferative effect. | The mechanism of action was not explored in the study. | [86] |
MG-63 Human Osteosarcoma cell line | Oxidovanadium with flavonoid chrysin [VO(chrysin)2 EtOH]2 (VOchrys) | Multicellular spheroids treated with 100 µM of VOchrys for 72 h of treatment. | Cytotoxicity affecting the shape and volume of the spheroids | The mechanism of action was not explored in the study. | [90] |
4,7-dimethyl-1,10-phenathroline 7 sulphatooxidovanadium(IV) nanostructured lipid carriers (NLCs Mv) | 25 μM and 50 μM of NLCs Mv for 24 h of treatment | Reduction in cell viability and cytotoxicity. | Apoptosis | [93] | |
Oxidovanadium(IV) -clioquinol complex VO(CQ)2 | 2.5–10 µM of VO(CQ) for 24 h of treatment | Inhibition of cell migration | Reduced activity of MMP-2 and MMP-9. | [91] | |
SW1353 Human osteosarcoma | Vanadium-doped nanocrystalline hydroxyapatite V-doped HAP nanoparticles | 5.0 mg/mL of V-doped HAP nanoparticles for 72 h of treatment | Cytotoxicity | The mechanism of action was not explored in the study. | [92] |
Cancer Model | Vanadium Compounds | Route/Dose/Duration | Biological Effects | Mechanisms | References |
---|---|---|---|---|---|
DMBA breast cancer model in female Sprague-Dawley rats | Vanadium-quercetin complex | Oral administration, 20 mg/kg of body mass with V-quercetin complex (1 dose per day for 24 weeks). | Inhibition of the development of mammary lesions in rats treated with the complex. | Increased apoptotic index, p53 and Bax; decreased proliferation. | [40] |
Breast cancer model 4T1-induced mammary tumors in female Balb/c mice | Sodium metavanadate (NaVO3) | Intraperitoneal administration of 10, 15 and 20 mg/Kg of body mass with NaVO3 (1 dose per day for three weeks). | Decrease in tumor volume, less development of microvasculature. | Inhibition of tumor growth and increased death by apoptosis. Increase in ROS. | [41]. |
Methylimidazole-based oxidovanadium(IV) complex (OVMI) | Intravenously administration of 10, 20 and 30 mg/kg of body mass with OVMI (Once 72 h after the previous injection for a range of 16 days. | Size and volume tumor size reduction. Antimetastatic activity. | Decreasing cell proliferation and Ki67 expression, apoptosis and decreased metastasis markers (E-cadherin and N-cadherin). | [44]. | |
Orthotopic Hepatocellular carcinoma model in male nude BALB/C mice | Sodium orthovanadate (SOV) | Intraperitoneal administration of 10, 15 and 20 mg/Kg of body mass with SOV (1 dose per day for three weeks). | Decrease in tumor volume, and inhibition of tumor growth, less development of microvasculature. | Suppression of cell proliferation, decreasing Ki67-positive cells, inhibition of autophagy. | [48] |
CT26 Liver metastasis model in BALB/C mice | Flavonoid luteolin and oxidovanadium(IV) complex [VO(lut)(H2O)2]Na3H2O (VOlut) | Intravenously administration of 20 mg/kg of body mass with VOlut. | No evidence of metastatic nodules in the liver | The mechanism of action was not explored in the study. | [56] |
DMH colorectal cancer model in male Wistar rats | Vanadium-luteolin complex | Via oral gavage commencing the day after DMH injection and continued for 18 weeks. | Reduction in the formation of aberrant crypt foci and cell death. | Increasing in p53 and Bax expressions, Bcl2, apoptosis. Elevation of SOD, CAT, and glutathione levels | [57] |
Mouse melanoma in female C57BL/6J mice implanted with the B16F10 cell cancer line | Vanadium pentoxide nanoparticles (V2O5 NPs 30–60 nm) | Intraperitoneal administration of 10 and 20 mg of V2O5 NPs (7 consecutive doses during 28 days). | Improved survival of melanoma tumor-bearing C57BL6/J mice and showed no toxic effects in treated animals. | Tumoral angiogenesis inhibition. | [72] |
Vanadium pentoxide nanoparticles (V2O5 NPs) and Ni doped vanadium pentoxide nanoparticles (Ni@V2O5 NPs) | Intraperitoneal administration of 100 mg/kg of V2O5 NPs and 50 mg/kg of Ni@V2O5 NPs (1 dose per day for 28 days). | Reduction in the volume of melanomas generated | Increased expression of Akt and PI3K interfering with this signaling pathway. | [73] | |
Urethane lung cancer model in male CD-1 mice | Vanadium pentoxide (V2O5) | Inhalation administration of 1.56 mg V/m3 of V2O5 (twice a week, for 8 weeks). | Decreased tumor size, increased cell death. | Increase in the apoptotic index in terminal deoxynucleotidyl transferase-labeled tumor cells, which reduced the number and size of tumors | [82] |
Murine glioblastoma GL261 induced tumors in male C57BL/6J mice | Bis(acetylacetonato)oxidovanadium(IV) (VO(acac)2) | Intratumoral injection of 10 and 25 μmol/kg VO(acac)2 (1 dose daily for 14 days). | Inhibition of tumor growth, inhibition of angiogenesis and tumor ischemia. | Suppression of HIF-1α levels, formation of vacuoles in tumor tissues leading to death via Metuosis. | [87] |
Osteosarcoma cell xenograft in females and males N:NIH (S) Fox1nu mice | Oxidovanadium with flavonoid chrysin [VO(chrysin)2 EtOH]2 (VOchrys) | Intraperitoneal administration of 27.5 mg/kg of body mass with VOchrys. | Decreased tumor volume in a concentration dependent manner. | The mechanism of action was not explored in the study. | [90] |
Osteosarcoma cell xenograft in male SD rats injected with UMR-106 cancer cells. | Vanadium-doped mesoporous bioactive glass (V(IV/V)-MBG) | The V(IV/V)-MBG scaffold was implanted in the tumor excision area and euthanized at 4 and 8 weeks. | Inhibition of tumor cell growth and promotion of normal bone in the tumor excision site. | Increased ROS generation. The continued release of V(IV) and V(V) significantly promoted osteogenesis. | [94] |
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
López-Valdez, N.; Gonzalez-Villalva, A.; Rojas-Lemus, M.; Bizarro-Nevares, P.; Casarrubias-Tabarez, B.; Cervantes-Valencia, M.E.; Ustarroz-Cano, M.; Guerrero-Palomo, G.; Morales-Ricardes, G.; Salgado-Hernández, J.Á.; et al. Vanadium, a Promising Element for Cancer Treatment. Inorganics 2025, 13, 298. https://doi.org/10.3390/inorganics13090298
López-Valdez N, Gonzalez-Villalva A, Rojas-Lemus M, Bizarro-Nevares P, Casarrubias-Tabarez B, Cervantes-Valencia ME, Ustarroz-Cano M, Guerrero-Palomo G, Morales-Ricardes G, Salgado-Hernández JÁ, et al. Vanadium, a Promising Element for Cancer Treatment. Inorganics. 2025; 13(9):298. https://doi.org/10.3390/inorganics13090298
Chicago/Turabian StyleLópez-Valdez, Nelly, Adriana Gonzalez-Villalva, Marcela Rojas-Lemus, Patricia Bizarro-Nevares, Brenda Casarrubias-Tabarez, María Eugenia Cervantes-Valencia, Martha Ustarroz-Cano, Gabriela Guerrero-Palomo, Guadalupe Morales-Ricardes, José Ángel Salgado-Hernández, and et al. 2025. "Vanadium, a Promising Element for Cancer Treatment" Inorganics 13, no. 9: 298. https://doi.org/10.3390/inorganics13090298
APA StyleLópez-Valdez, N., Gonzalez-Villalva, A., Rojas-Lemus, M., Bizarro-Nevares, P., Casarrubias-Tabarez, B., Cervantes-Valencia, M. E., Ustarroz-Cano, M., Guerrero-Palomo, G., Morales-Ricardes, G., Salgado-Hernández, J. Á., & Fortoul, T. I. (2025). Vanadium, a Promising Element for Cancer Treatment. Inorganics, 13(9), 298. https://doi.org/10.3390/inorganics13090298