Next Article in Journal
Solid-Phase Microextraction as an Antibiotic Resistance Detector in Staphylococcus aureus Strains
Previous Article in Journal
Is There a Relationship between Biofilm Forming-Capacity and Antibiotic Resistance in Staphylococcus spp.? In Vitro Results
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Proceeding Paper

Potential Applications of Vanadium-Based Anticancer Drugs for Intratumoral Injections †

1
Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
2
School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
3
Cellular and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
*
Authors to whom correspondence should be addressed.
Presented at the Biosystems in Toxicology and Pharmacology—Current challenges, 8–9 September 2022; Available online: https://bitap.sciforum.net/.
Med. Sci. Forum 2022, 11(1), 10; https://doi.org/10.3390/BiTaP-12783
Published: 29 June 2022
(This article belongs to the Proceedings of Biosystems in Toxicology and Pharmacology—Current Challenges)

Abstract

:
The administration of highly cytotoxic or immunomodulating drugs directly into a tumor is a method used in the clinic for late stages of cancers and in clinical trials for platinum-based drugs. A hydrophobic non-innocent Schiff base V(V) complex with a sterically hindered catecholate ligand was taken up rapidly into cancer cells and caused cell death as required for potential use as intratumoral agents. The synthesis was non-trivial on large scales and high purities. This class of complexes with sterically hindred catecholates is sufficiently stable to survive briefly under physiological conditions before hydrolysis and/or redox reactions. Degradation reactions occur very rapidly for complexes with less sterically hindered catacholates.

1. Introduction

The first chemotherapeutic metal-complex, cisplatin, was approved by the FDA in 1978 and since that time, other cytotoxic metal-based chemotherapeutics have been approved, are in clinical trials, or are under development [1,2,3,4,5,6], Figure 1. A major objective in the development of effective chemotherapeutics is increasing efficacy while reducing toxicity to healthy tissue to acceptable levels. After the approval of cisplatin, early investigations turned to the development of other metal-based drugs, including ruthenium- and titanium-based drugs. However, these drugs were canceled during clinical trials due to nephrotoxicity, which limited dosage to ineffective levels [2]. One strategy to reduce toxicity relies on designing metal complexes that degrade after intratumoral injection and before encountering healthy tissue.
A major obstacle facing chemotherapeutic metal complexes is effective movement across cell membranes. It is well-established that the cell membrane is distinctly hydrophobic and interacts favorably with hydrophobic drugs. Hydrophobic non-innocent vanadium(V) complexes are one example of such a class of potential drugs that may be suitable for intratumoral injection due to their rapid uptake [2,6]. These vanadium(V) complexes are designed to break down before diffusion or transport outside the tumor and into contact with healthy tissue. The breakdown products of these potential drugs are inoffensive to the human body and may have beneficial properties. In this way, such a compound acts to support healthy tissue during an otherwise toxic event. Currently, one intratumoral pharmaceutical, an oncolytic virus (T-VEC), is in clinical use for melanoma which cannot be surgically removed. Furthermore, seven platinum-based anticancer preparations are in clinical trials.

2. Synthesis and Properties of Hydrophobic Non-Innocent Vanadium(V) Complexes

Metal complexes containing redox-active ligands are named non-innocent metal complexes and are widely used in catalytic reactions, sensors in biological systems, drug delivery, and water remediation [7,8,9]. The term “non-innocent metal complex” refers to a complex where, when a redox reaction has occurred, it is not certain if the resulting charge is localized mainly on the metal ion, or on the coordinated ligand, or is substantially delocalized over both [7]. Our groups first became interested in these compounds after observing their unusual electronic [9], and later cytotoxic, properties. With that in mind, we continued our investigations into the biological activities of these compounds, and have since synthesized, and tested, a number of these derivatives, Figure 1 and Figure 2.
Among the vanadium(V) complexes we have synthesized are [VO(HSHED)Cat], and the more sterically hindered derivative [VO(HSHED)dtb]. Regardless of the exact steric, electronic, or structural nature of a [VO(HSHED)Cat] derivative, the synthetic scheme of these vanadium(V) complexes begins with the condensation product of the Schiff base ligand from 2-((aminomethyl)amino)ethan-1-ol and salicaldehyde under an inert atmosphere, Figure 2. Then, an aqueous solution of vanadyl sulfate was added to the Schiff base scaffold, followed by aqueous sodium hydroxide to give solid [VO2(HSHED)]. To an acetone solution of this solid, catechol was added to give a purple crystalline powder, Figure 2 [8,9].
Due to the profound effect of the catechol ligands upon the electronic properties of these compounds, these reactions and their corresponding products are quite distinctive and can be monitored by both 51V NMR spectroscopy and UV-vis spectrophotometry. Both methods are significantly influenced by the electronic properties of the vanadium(V) as they reflect the size of the HOMO-LUMO gap, Figure 3. For instance, when the catechol substituent is decorated with electron-donating functional groups, the HOMO-LUMO gap is lowered and the 51V NMR is pushed downfield [9].
When functionalized with electron-withdrawing functional groups, the HOMO-LUMO gap is widened and the 51V NMR signal moves upfield. Similarly, the color of this class of compounds varies from the bright yellow from the vanadium scaffold to a non-innocent vanadium complex that ranges from dark purple to green. Furthermore, the HOMO-LUMO gap impacts the redox reactions of these complexes [9]. While [VO(HSHED)cat] is a cytotoxic complex, it also rapidly hydrolyzes. With the substitution of two t-butyl (dtb) functional groups onto the catechol ligand, [VO(HSHED)(dtb)] (previously and in Figure 3 abbreviated [VO(HSHED)dtbCat]) demonstrated a significantly prolonged lifetime in both water and cell culture medium compared to [VO(HSHED)Cat]. The functionalization of catechol with electron-donating di-tert-butyl groups raises both the HOMO and LUMO, with a net decrease in the HOMO-LUMO gap. In contrast, the tetra-brominated analogue, [VO(HSHED)tbCat], decreases both HOMO and LUMO, with a net increase in the HOMO-LUMO gap, Figure 3.

3. Biological Studies

To investigate if there is a relationship between hydrophobicity and V(V) complex cytotoxicity, we conducted experiments with [VO(HSHED)Cat] and [VO(HSHED)dtb] in a simple monolayer model membrane system. Using microemulsions (reverse micelles), we generated a self-assembled system containing H2O/dioctyl sulfosuccinate sodium salt/organic solvent, where the organic solvent was either iso-octane or cyclohexane [10]. Using 51V NMR and UV-vis spectroscopies to monitor the complex stability, we demonstrated that the more hydrophobic and sterically hindered [VO(HSHED)dtb] was stabilized by this monolayer and was slower to hydrolyze than the parent [VO(HSHED)Cat] complex. The greater cellular penetration of [VO(HSHED)dtb] would favor more potent anticancer properties and this was observed for human bone cancer cells (SW1353) [10].
Cellular experiments were designed to cross-test several cancer cell lines against the intact and hydrolyzed complex. Fresh [VO(HSHED)dtb] was tested alongside its breakdown products (aged [VO(HSHED)dtb]). Also tested were fresh and aged cisplatin, as reference compounds. These substances were all tested against cancerous brain (T98g cells), breast (MDA-MB-231), pancreatic (PANC-1), lung (A549), and normal connective (HFF-1) tissues. In all cell lines, fresh [VO(HSHED)dtb] had the lowest IC50 (highest activity) of the substances tested. Furthermore, in T98g cells [VO(HSHED)dtb] was an order of magnitude more cytotoxic than its breakdown products or cisplatin, Figure 4 [6].
The IC50 values of [VO(HSHED)dtb], fresh (intact) and aged (decomposed), showed promise as a candidate for intratumoral injection into brain cancers. Fresh [VO(HSHED)dtb] may be more effective at killing cancerous cells than cisplatin, however, the decomposition products were comparatively safe and harmless. In fact, some of these products have demonstrated neuroprotective and neurostimulating properties and may serve to support healthy tissue through both cancer and chemotherapy, however, further studies are needed to understand how complex reactivity, degradation, and speciation within a biological system affect activity [11,12,13].

4. Conclusions and Future Directions

We describe [VO(HSHED)dtb] as a potential intratumoral drug, which has the required rapid cellular uptake, enhanced reactivity, and non-toxic decomposition products. While some hydrophobic, sterically hindered vanadium(V) complexes perform quite well in in vitro experiments against cancer cell lines, it remains unclear if complexes with even greater degrees of hydrophobicity and steric hindrance will perform proportionally better. We are currently testing other complexes which we have designed with these properties. We have suggested that not only these non-innocent vanadium complexes, but also other complexes that have been investigated in clinical trials but abandoned because they did not exhibit sufficient stability to be administered by conventional methods could be excellent candidates for intratumoral administration [2].

Author Contributions

Conceptualization, D.C.C. and P.A.L.; writing—original draft preparation, J.M. and D.C.C.; writing—review and editing, J.M., A.L., P.A.L. and D.C.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Australian Research Council (ARC) grants to P.A.L. for a Senior Research Associate position for A.L. (DP160104172, DP180102741 and DP210101632) and from the Office of the Vice President for Research at Colorado State University to D.C.C.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original experimental data for cytotoxicity are available on request from aviva.levina@sydney.edu.au and other data from debbie.crans@colostate.edu. Some of this data have already been reported and is available in supplemental materials in previous publications.

Acknowledgments

The authors acknowledge the facilities and the scientific and technical assistance of Sydney Microscopy & Microanalysis at Microscopy Australia, the Molecular Biology Facility of the Bosch Institute, and Sydney Analytical all at the University of Sydney as well as the ARC at Colorado State University.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cohen, S.M.; Lippard, S.J. Cisplatin: From DNA damage to cancer chemotherapy. Prog. Nucleic Acid Res. Mol. Biol. 2001, 67, 93–130. [Google Scholar] [CrossRef]
  2. Levina, A.; Crans, D.C.; Lay, P.A. Advantageous Reactivity of Unstable Metal Complexes: Potential Applications of Metal-Based Anticancer Drugs for Intratumoral Injections. Pharmaceutics 2022, 14, 790. [Google Scholar] [CrossRef] [PubMed]
  3. Gasser, G.; Ott, I.; Metzler-Nolte, N. Organometallic Anticancer Compounds. J. Med. Chem. 2011, 54, 3–25. [Google Scholar] [CrossRef] [PubMed]
  4. Ang, W.H.; Casini, A.; Sava, G.; Dyson, P.J. Organometallic ruthenium-based antitumor compounds with novel modes of action. J. Organomet. Chem. 2011, 696, 989–998. [Google Scholar] [CrossRef]
  5. Pessoa, J.C.; Etcheverry, S.; Gambinog, D. Vanadium compounds in medicine. Coord. Chem. Rev. 2015, 301, 24–48. [Google Scholar] [CrossRef] [PubMed]
  6. Levina, A.; Vieira, A.P.; Wijetunga, A.; Kaur, R.; Koehn, J.T.; Crans, D.C.; Lay, P.A. A Short-Lived but Highly Cytotoxic Vanadium(V) Complex as a Potential Drug Lead for Brain Cancer Treatment by Intratumoral Injections. Angew. Chem. Int. Ed. Engl. 2020, 59, 15834–15838. [Google Scholar] [CrossRef] [PubMed]
  7. Lyaskovskyy, V.; de Bruin, B. Redox Non-Innocent Ligands: Versatile New Tools to Control Catalytic Reactions. ACS Catal. 2012, 2, 270–279. [Google Scholar] [CrossRef]
  8. Cornman, C.R.; Colpas, G.J.; Hoeschele, J.D.; Kampf, J.; Pecoraro, V.L. Implications for the Spectroscopic Assignment of Vanadium Biomolecules: Structural and Spectroscopic Characterization of Monooxovanadium(V) Complexes Containing Catecholate and Hydroximate Based Noninnocent Ligands. J. Am. Chem. Soc. 1992, 114, 9925–9933. [Google Scholar] [CrossRef]
  9. Chatterjee, P.B.; Goncharov-Zapata, O.; Quinn, L.L.; Hou, G.; Hamaed, H.; Schurko, R.W.; Polenova, T.; Crans, D.C. Characterization of noninnocent metal complexes using solid-state NMR spectroscopy: O-dioxolene vanadium complexes. Inorg. Chem. 2011, 50, 9794–9803. [Google Scholar] [CrossRef] [PubMed]
  10. Crans, D.C.; Koehn, J.T.; Petry, S.M.; Glover, C.M.; Wijetunga, A.; Kaur, R.; Levina, A.; Lay, P.A. Hydrophobicity may enhance membrane affinity and anti-cancer effects of Schiff base vanadium(v) catecholate complexes. Dalton Trans. 2019, 48, 6383–6395. [Google Scholar] [CrossRef] [PubMed]
  11. He, Z.; Han, S.; Wu, C.; Liu, L.; Zhu, H.; Liu, A.; Lu, Q.; Huang, J.; Du, X.; Li, N.; et al. Bis(ethylmaltolato)oxidovanadium(IV) inhibited the pathogenesis of Alzheimer’s disease in triple transgenic model mice. Metallomics 2020, 12, 474–490. [Google Scholar] [CrossRef] [PubMed]
  12. 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, 325, 473–498. [Google Scholar] [CrossRef]
  13. 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]
Figure 1. Anticancer metal complexes used in the clinic, or under investigation [1,2,3,4].
Figure 1. Anticancer metal complexes used in the clinic, or under investigation [1,2,3,4].
Msf 11 00010 g001
Figure 2. The reaction scheme for the synthesis of [VO(HSHED)Cat] and derivatives.
Figure 2. The reaction scheme for the synthesis of [VO(HSHED)Cat] and derivatives.
Msf 11 00010 g002
Figure 3. A qualitative molecular orbital diagram demonstrating the impact of different ligands in free and vanadium(V) coordinated catechol and their effects on 51V NMR chemical shifts. Note in this figure [VO(HSHED)dtb] was abbreviated [VO(HSHED)dtbCat]. Adopted with permission from Ref. [9]. 2011. American Chemical Society.
Figure 3. A qualitative molecular orbital diagram demonstrating the impact of different ligands in free and vanadium(V) coordinated catechol and their effects on 51V NMR chemical shifts. Note in this figure [VO(HSHED)dtb] was abbreviated [VO(HSHED)dtbCat]. Adopted with permission from Ref. [9]. 2011. American Chemical Society.
Msf 11 00010 g003
Figure 4. Dose-response curves of fresh [VO(HSHED)dtb], aged [VO(HSHED)dtb], fresh cisplatin, and aged cisplatin in T98g cells (72 h treatments). “Fresh” solutions were added to the cell culture medium less than 60 s prior to cell treatment. “Aged” solutions were incubated in a cell culture medium under a 5% CO2 atmosphere for 24 h at 310 K before addition to cells. Adopted with permission from Ref. [6]. 2020. Wiley-VCH GmbH.
Figure 4. Dose-response curves of fresh [VO(HSHED)dtb], aged [VO(HSHED)dtb], fresh cisplatin, and aged cisplatin in T98g cells (72 h treatments). “Fresh” solutions were added to the cell culture medium less than 60 s prior to cell treatment. “Aged” solutions were incubated in a cell culture medium under a 5% CO2 atmosphere for 24 h at 310 K before addition to cells. Adopted with permission from Ref. [6]. 2020. Wiley-VCH GmbH.
Msf 11 00010 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Manganaro, J.; Levina, A.; Lay, P.A.; Crans, D.C. Potential Applications of Vanadium-Based Anticancer Drugs for Intratumoral Injections. Med. Sci. Forum 2022, 11, 10. https://doi.org/10.3390/BiTaP-12783

AMA Style

Manganaro J, Levina A, Lay PA, Crans DC. Potential Applications of Vanadium-Based Anticancer Drugs for Intratumoral Injections. Medical Sciences Forum. 2022; 11(1):10. https://doi.org/10.3390/BiTaP-12783

Chicago/Turabian Style

Manganaro, John, Aviva Levina, Peter A. Lay, and Debbie C. Crans. 2022. "Potential Applications of Vanadium-Based Anticancer Drugs for Intratumoral Injections" Medical Sciences Forum 11, no. 1: 10. https://doi.org/10.3390/BiTaP-12783

Article Metrics

Back to TopTop