View Article

Abstract

Cancer remains one of the most significant global health challenges, with high mortality rates and limited treatment success for advanced stages. Since the approval of cisplatin, metal-based drugs have played a central role in chemotherapy. However, the severe toxicity, drug resistance, and poor selectivity of platinum-based agents have driven the exploration of next-generation metal complexes with novel structures, mechanisms, and delivery strategies. This review integrates recent advances in the design of non-platinum metal complexes—including ruthenium, gold, copper, titanium, and emerging metals—linking their structure–activity relationships (SAR) to mechanistic pathways such as redox modulation, enzyme inhibition, photodynamic activation, and epigenetic regulation. We highlight strategies to improve efficacy and selectivity through ligand modification, prodrug activation, nanocarrier systems, and tumor microenvironment targeting. Clinical trial updates are discussed alongside future perspectives for translational research. This comprehensive analysis aims to bridge the gap between laboratory innovation and clinical application, underscoring the potential of mechanism-driven design to redefine metal-based chemotherapy.

Keywords

Non-platinum metallodrugs, Ruthenium, Gold, Titanium, Copper, Palladium, Iridium, Anticancer, Mechanism, Clinical trials

Introduction

1.1 Global cancer burden

Cancer is a leading cause of morbidity and mortality worldwide, responsible for an estimated 10 million deaths annually [1]. Incidence rates continue to rise due to aging populations and lifestyle factors; projections indicate a significant increase in global cancer cases over the next two decades [2]. Although early detection and targeted therapies have improved outcomes for certain tumor types, systemic chemotherapies remain indispensable in the management of many solid and hematological malignancies.

1.2 Evolution of metallodrugs

The discovery of cisplatin in the 1960s marked a watershed in medicinal chemistry, demonstrating that simple coordination complexes can exert profound biological effects [3]. Subsequent development of carboplatin and oxaliplatin sought to retain antitumor efficacy while reducing side effects. Nevertheless, platinum drugs primarily operate via DNA crosslinking and encounter limitations in selectivity and resistance.

Figure 1: Overview of non-platinum metal complexes in anticancer therapy.

1.3 Limitations of platinum-based chemotherapy

Platinum therapeutics incur dose-limiting toxicities such as nephrotoxicity, peripheral neuropathy, and myelosuppression, which restrict optimal dosing [4]. Tumor resistance emerges through multiple mechanisms: enhanced DNA repair (nucleotide excision repair), increased drug efflux (ATP-binding cassette transporters), detoxification via glutathione conjugation, and inhibition of apoptotic signaling [5]. These clinical challenges underpin the ongoing search for alternative metal-based therapeutics with distinct mechanisms and improved therapeutic windows.

1.4 Rationale for next-generation metal complexes

Non-platinum metals (ruthenium, gold, copper, titanium, palladium, iridium, etc.) present rich coordination chemistry, multiple accessible oxidation states, and varied ligand architectures. These properties enable modes of action not limited to DNA crosslinking — e.g., redox modulation, enzyme inhibition, photochemical activation, and epigenetic regulation — which can address resistance and selectivity shortcomings of platinum compounds [6].

1.5 Scope and novelty of this review

This review focuses on mechanism-driven design of next-generation metal complexes and their translation from laboratory discovery to clinical testing. We place particular emphasis on structure–activity relationships (SAR), delivery strategies, and representative clinical advances to give a cohesive view bridging chemistry and medicine.

2. Classification Of Non-Platinum Metal-Based Anticancer Agents

This section groups representative non-platinum complexes by metal and highlights key ligand types, mechanisms, and current translational status. For each compound listed in Table 1.

Table 1 – Representative of Next-Generation Metal Complexes with Mechanism and Clinical status

Sr.no:

Metal

Example Compounds (structure below each name)

Ligands Used / Typical Donor Types

Key Mechanism(s)

Clinical Status

1

Ruthenium

NAMI-A
KP1019
NKP-1339

 

Imidazole, indazole, polypyridyl

Redox activation, metastasis inhibition, DNA interactions

Phase I/II (varies by compound)

2

Gold

Auranofin

Au(III)-porphyrin

 

Phosphine, N-heterocyclic carbenes (NHC), porphyrin cores

Thioredoxin reductase inhibition, protein targeting

Clinical evaluation (Auranofin in various trials)

3

Copper

Casiopeínas

Cu-phenanthroline

 

Phenanthroline, Schiff bases, mixed N,O donors

ROS generation, proteasome inhibition, redox cycling

Preclinical / early translational

4

Titanium

Titanocene dichloride
 

Cyclopentadienyl (Cp), alkyl/halide ligands

DNA interaction, hydrolytic activation

Early-phase clinical studies historically

5

Palladium

Pd-salen


 

Salen, polypyridyl ligands

DNA intercalation, enzyme inhibition

Preclinical

2.1 Ruthenium complexes

Ruthenium complexes have attracted attention because of their kinetic stability, multiple oxidation states (Ru(II)/Ru(III)), and favorable ligand exchange chemistry that can be tuned for activation in biological milieus. Notable examples include NAMI-A (imidazole-containing, metastasis-inhibiting), KP1019 (indazole-based, antitumor), and NKP-1339 (a Na-derivative with improved solubility and pharmacokinetics). Mechanistically, ruthenium complexes can act via DNA binding, inhibition of metastasis pathways, and redox-mediated cytotoxicity; some are designed as prodrugs activated by reduction in the tumor microenvironment [7-9].

NAMI-A (imidazolium trans-[tetrachloro(dimethyl sulfoxide)(imidazole)ruthenate(III)])

  • Primarily anti-metastatic rather than cytotoxic to primary tumors.
  • Prefers binding to extracellular matrix components and inhibits tumor cell invasion [9].
  • Activity is enhanced in hypoxic tumor regions due to its redox-activated Ru(III)/Ru(II) conversion.

Figure 1.2: Chemical structure of NAMI-A.  imidazolium trans-[tetrachloro (dimethyl sulfoxide) (imidazole) ruthenate (III)] complex.

KP1019 (indazolium trans-[tetrachlorobis(indazole)ruthenate(III)])

  • Targets mitochondria, causing rapid depolarization and apoptosis induction [10].
  • Exhibits selective uptake by cancer cells due to elevated transferrin receptor expression.

Figure 1.3: Chemical structure of KP1019, an indazolium trans-[tetrachlorobis(indazole)ruthenate(III)] complex.

NKP-1339 (sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)])

  • A sodium salt analogue of KP1019 with improved solubility and pharmacokinetics [11].
  • Induces oxidative stress and ER stress, triggering unfolded proteinactivated by reduction in the tumor microenvironment [7-9].

Figure 1.4: Chemical structure of NKP-1339, a sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] complex.

SAR Note – Ligand Electronics & Geometry Effects in Ruthenium Complexes

  1. Electronic Effects

Electron-donating ligands (e.g., alkyl-substituted amines, methoxy-aryl) raise the Ru(III)/Ru(II) redox potential, facilitating easier reduction under hypoxic tumor conditions → ↑ activation in cancer cells[39]. Electron-withdrawing ligands (e.g., halogens, nitro-aryl) lower the redox potential, stabilizing the Ru(III) oxidation state → ↓ premature activation, ↑ stability in plasma.

  1. Geometry / Coordination Environment
  • Octahedral low-spin Ru(III) with bidentate N,N-ligands (as in Complex 1) offers high kinetic stability but slower ligand exchange → often reduced uptake without transport mediation[44].
  • Distorted octahedral / mixed N,O coordination (Complex 2) increases ligand lability → ↑ reactivity with biomolecules, faster activation.
  • Planarized chelates (Complex 3, with extended aromatic systems) enhance π–π stacking with DNA and improve passive membrane diffusion.
  1. Cellular Uptake Influence
  • More lipophilic ligands → ↑ passive diffusion across membranes, but risk of nonspecific binding[35].
  • Hydrophilic / charged ligands rely on active transporters (e.g., transferrin receptor), increasing selectivity for cancer cells over normal tissue.
  • Balanced polarity (logP ~1–3) gives optimal uptake without rapid clearance.

2.2 Gold complexes

Gold(I/III) complexes (e.g., Auranofin) show potent inhibition of thioredoxin reductase (TrxR) and related selenoenzymes, disrupting redox homeostasis and inducing apoptosis particularly in drug-resistant and hematological malignancies. Auranofin’s repurposing potential has been explored in several clinical settings, and Au(III)-porphyrin complexes add photophysical handles for combined photodynamic/chemical cytotoxicity [11-12].

Au(III)-Porphyrin

  • Structurally rigid complexes that mimic platinum’s square planar geometry but are redox-active.
  • Potently inhibit thioredoxin reductase (TrxR), a key enzyme in redox homeostasis, leading to ROS accumulation [12].
  • Show high cytotoxicity in cisplatin-resistant tumor lines.

Figure 2: Chemical structure of a gold(III) complex with a tetradentate N-heterocyclic macrocyclic ligand coordinated to a chloride ligand.

2.3 Copper complexes

Copper-based agents (Casiopeínas, Cu-phenanthroline derivatives) leverage copper’s redox cycling ability to catalyze Fenton-like reactions in cells, raising ROS to cytotoxic levels selectively in cancer cells with already elevated oxidative stress. Additionally, copper complexes have been reported to inhibit proteasome activity and modulate signaling pathways [13-15].

Casiopeina III-ia (Cu(4,7-dimethyl-1,10-phenanthroline) (acetylacetonate))

  • Capable of Fenton-like reactions, producing hydroxyl radicals that damage DNA and proteins [13].
  • Chelates target nucleic acids with high affinity, causing double-strand breaks.

Figure 3: Chemical structure of a copper(II) complex containing 2,9-dimethyl-1,10-phenanthroline and an amino acid-derived ligand, coordinated with a water molecule and nitrate counterion.

Figure 3.1: Chemical structures of copper(II) complexes with various substituted phenanthroline ligands, showing different methyl, phenyl, chloro, and nitro substitutions that influence electronic properties and biological activity.

2.4 Titanium complexes

Organotitanium complexes such as titanocene derivatives exhibit unique DNA binding modes and lower nephrotoxicity relative to cisplatin. Their clinical evaluation faced formulation and stability challenges, but they remain of mechanistic interest due to different cellular processing and activation pathways [14].

Titanocene Dichloride (Cp?TiCl?)

  • The first non-platinum metallodrug to enter clinical trials [14].
  • Hydrolyzes under physiological conditions, releasing Ti species that bind DNA phosphate groups.
  • Demonstrates immunomodulatory effects alongside direct cytotoxicity.

Figure 4: Chemical structure of titanocene dichloride, a metallocene titanium(IV) complex featuring two cyclopentadienyl ligands and two chloride ligands.

2.5 Palladium Complexes

Palladium complexes, especially with extended aromatic ligands (dppz), can intercalate DNA and disrupt enzyme function. Iridium and osmium complexes are emerging for photodynamic/photocatalytic therapies owing to their strong luminescence and ability to produce singlet oxygen [17-19].

Pd(II)-Salen Complex

  • Forms bidentate Schiff-base ligands with palladium, enabling DNA crosslinking similar to cisplatin but with faster ligand exchange rates [15].
  • Modifications on the salen ligand allow fine-tuning of lipophilicity and cytotoxicity.

Figure 5: ORTEP diagram of a palladium(II) complex showing the coordination of bidentate N,O-donor ligands to the central Pd atom, with thermal ellipsoids depicted at the 50% probability level.

2.6 Iridium Complexes

Ir(III)-Cyclometalated Complexes

  • Exhibit strong phosphorescence, enabling theranostic applications [16].
  • Activate under visible or near-infrared light for photodynamic therapy, generating singlet oxygen to kill cancer cells.

Figure 6: Chemical structures of a series of iridium(III) polypyridyl complexes (1–5) featuring varied ancillary ligands, illustrating structural modifications aimed at tuning their photophysical and biological properties.

2.7 Osmium Complexes

Os(II)-Polypyridyl Complexes

  • Display DNA intercalation and oxidative cleavage abilities [17].
  • More inert ligand exchange kinetics than Ru, potentially leading to prolonged circulation times and controlled activity.

Figure 7: ORTEP representation of an osmium (II) polypyridyl complex with coordinated aromatic ligands and hexafluorophosphate counterions, showing the atomic labeling scheme and thermal ellipsoids.

3. Mechanisms Of Action

Non-platinum metal complexes exert anticancer effects through multiple, often overlapping, pathways. The diversity in metal coordination chemistry allows targeting of DNA, enzymes, redox systems, and specific signaling pathways.

3.1 DNA Damage and Crosslinking

  • Ruthenium (II/III) complexes (e.g., KP1019, NKP-1339) can bind to guanine N7 positions, causing mono- and bifunctional adducts that distort DNA geometry [18].
  • Palladium (II) and titanium (IV) complexes also form DNA adducts, inhibiting replication and transcription.
  • Some complexes intercalate between DNA base pairs, stabilizing the double helix in a distorted conformation.

3.2 Enzyme Inhibition

  • Gold(I/III) complexes strongly inhibit thioredoxin reductase (TrxR), disrupting redox balance and promoting oxidative damage [19].
  • Copper(II) complexes target topoisomerase I/II, hindering DNA relaxation needed for replication.
  • Inhibition is often irreversible due to covalent binding with enzyme cysteine/selenocysteine residues.

3.3 Reactive Oxygen Species (ROS) Generation

  • Copper and iron-mimicking Ru complexes catalyze Fenton-like reactions, generating hydroxyl radicals [20].
  • ROS accumulation leads to mitochondrial membrane depolarization, cytochrome c release, and apoptosis.

3.4 Photodynamic Effects

  • Iridium(III) and some osmium(II) complexes act as photosensitizers [21].
  • Upon light activation, they transfer energy to oxygen molecules, generating singlet oxygen (^1O?) and triggering localized oxidative damage.

3.5 Apoptotic Pathway Activation

  • Metal complexes activate intrinsic apoptosis by modulating Bcl-2 family proteins (↑ Bax, ↓ Bcl-2) [22].
  • Mitochondrial outer membrane permeabilization (MOMP) leads to caspase-9 and -3 activation.
  • Some complexes also engage extrinsic death receptor pathways via Fas–FasL signaling.

4. Advantages And Limitations

The design of non-platinum complexes aims to overcome platinum-based drug drawbacks such as nephrotoxicity, ototoxicity, and acquired resistance. However, limitations remain.

5. Clinical Progress and Trials

Several non-platinum complexes have entered various stages of clinical evaluation

Table 2: Advantages and Limitations of Non-Platinum Metal Complexes

Sr no:

Metal Complex Type

Advantages

Limitations

 

1

Ruthenium (III/II) complexes

Lower toxicity than cisplatin, activity in hypoxic tumors, redox activation potential

Limited oral bioavailability, possible off-target protein binding

2

Gold(I/III) complexes

Strong TrxR inhibition, potent against resistant lines

Risk of systemic oxidative stress, stability issues in vivo

3

Copper (II) complexes

ROS-mediated DNA/protein damage, inexpensive and abundant

Non-selective ROS generation may harm healthy tissue

4

Titanium (IV) complexes

Different binding target (DNA phosphates), potential immunomodulation

Hydrolysis instability, limited clinical efficacy so far

5

Palladium (II) complexes

Rapid ligand exchange, tunable activity via ligand modification

Instability in biological media, non-specific DNA damage

6

Iridium (III) complexes

Theranostic potential, photodynamic therapy capability

Requires light activation, tissue penetration limits

Table 3: Clinical Trials Overview

Sr no:

Compound

Metal

Indication

Trial Phase

Key Findings

1

NAMI-A

Ru(III)

Non-small cell lung cancer, melanoma

Phase II

Anti-metastatic effect with minimal tumor shrinkage

2

KP1019

Ru(III)

Gastrointestinal cancers

Phase I

Well tolerated, disease stabilization in some patients

3

KP1339

Ru(III)

Advanced solid tumors

Phase I/II

Improved pharmacokinetics, early signs of efficacy

4

Auranofin

Au(I)

Ovarian cancer, leukemia

Phase II

Targets TrxR, manageable toxicity

5

Titanocene dichloride

Ti(IV)

Renal cell carcinoma

Phase II

Limited efficacy, short plasma half-life

 

Figure 8: Timeline of clinical progress for selected non-platinum metal-based anticancer agents

6. Strategies To Improve Efficacy and Selectivity

  • Ligand Modification: Altering lipophilicity, charge, and steric properties to improve tumor targeting.
  • Prodrug Approach: Using Ru(III) prodrugs activated in hypoxic tumors.
  • Nanocarrier-Based Delivery: Encapsulation in liposomes, polymeric nanoparticles to improve biodistribution.
  • Tumor Microenvironment Targeting: Designing complexes responsive to pH, glutathione levels.
  • Combination Therapies: Pairing with kinase inhibitors, immunotherapy to enhance effects.

Figure 9: Schematic representation of strategies to improve the selectivity of metal-based anticancer drugs.

7. CONCLUSION AND FUTURE PERSPECTIVES

Non-platinum complexes represent a promising expansion of the metallodrug field, with tunable structures for multi-targeted approaches. Combining therapeutic and diagnostic capabilities (theranostics) may revolutionize personalized cancer treatment. Advances in nanotechnology, ligand chemistry, and bioinorganic design will likely lead to next-generation metal complexes with improved selectivity and safety profiles. Non-platinum metal complexes offer mechanistic diversity, tumor selectivity, and activity against resistant cancers. Yet, clinical adoption has been hindered by stability, pharmacokinetic, and efficacy challenges. Future research should prioritize:

  1. Targeted delivery systems (nanocarriers, ligand conjugates) [39].
  2. Biomarker-driven therapy to improve patient selection [40].
  3. Combination strategies with immunotherapy or targeted drugs [41].
  4. Comprehensive ADME studies to balance efficacy and long-term safety [42].

REFERENCES

  1. Alessio E. Thirty years of the drug candidate NAMI-A and the myths in the field of ruthenium anticancer compounds. Met Ions Life Sci. 2018;18:233-254.
  2. Hartinger CG, Dyson PJ. Bioorganometallic chemistry—from teaching paradigms to medicinal applications. Chem Soc Rev. 2021;50(8):5301-5365.
  3. Casini A, Reedijk J. Challenges and promises for ruthenium anticancer drugs. Curr Top Med Chem. 2020;20(4):270-282.
  4. Heffeter P, et al. NAMI-A and KP1019—ruthenium anticancer drugs in different phases of clinical trials. Bioinorg Chem Appl. 2018;2018:1-15.
  5. Sava G, Bergamo A. Ruthenium-based drugs for cancer therapy: An update. Curr Med Chem. 2020;27(9):1466-1482.
  6. Bindoli A, Rigobello MP. Principles in redox signaling: From chemistry to functional significance. Antioxid Redox Signal. 2019;31(10):625-649.
  7. Gandin V, et al. Gold compounds as anticancer agents: A systematic review. Cancer Chemother Pharmacol. 2021;88(3):447-468.
  8. Moraes GC, et al. Anticancer activity of copper complexes. J Inorg Biochem. 2022;233:111839.
  9. Ang WH, Dyson PJ. Classical and non-classical ruthenium-based anticancer drugs: Towards targeted chemotherapy. Eur J Inorg Chem. 2020;2020(14):1541-1556.
  10. Pichler V, et al. KP1019 and NKP-1339: Ruthenium anticancer drug candidates. Inorganics. 2021;9(5):36.
  11. Zou T, et al. Recent advances in platinum-free metallodrugs for cancer therapy. Coord Chem Rev. 2022;469:214652.
  12. Bertrand B, Casini A. A golden future in medicinal inorganic chemistry: The promise of gold-based therapeutics. Dalton Trans. 2021;50(25):8709-8725.
  13. Fikselová M, et al. Biological activities of copper-based complexes in oncology. Molecules. 2023;28(3):1025.
  14. Melchart M, et al. Titanium-based anticancer drugs: Past, present, and future. J Med Chem. 2020;63(22):12531-12544.
  15. Savi? N, et al. Palladium complexes in anticancer therapy. Coord Chem Rev. 2021;436:213854.
  16. Liu Z, et al. Cyclometalated iridium(III) complexes for photodynamic therapy. Chem Rev. 2020;120(9):5150-5190.
  17. Streciwilk W, et al. Osmium complexes in medicinal chemistry. Dalton Trans. 2019;48(34):12899-12914.
  18. Zhou Q, et al. Ruthenium complexes targeting DNA in cancer therapy. ChemMedChem. 2020;15(19):1778-1790.
  19. Wang X, et al. Thioredoxin reductase inhibition by gold complexes. J Inorg Biochem. 2021;225:111580.
  20. Kalinowski DS, et al. ROS generation and metal complexes in cancer therapy. Free Radic Biol Med. 2022;170:1-21.
  21. Zhao J, et al. Photodynamic applications of metal complexes. Coord Chem Rev. 2021;438:213885.
  22. Wang J, et al. Apoptotic pathways induced by non-platinum metal complexes. Biochim Biophys Acta Mol Cell Res. 2020;1867(10):118789.
  23. Monro S, Colón KL, Yin H, et al. Transition metal complexes and photodynamic therapy… Chem Rev. 2019;119(2):797–828.
  24. Graf N, Lippard SJ. Redox activation of metal-based prodrugs. Adv Drug Deliv Rev. 2012;64(11):993–1004.
  25. Messori L, Merlino A. Gold and silver complexes with antimicrobial and anticancer activity… Chem Commun. 2016;52(18):3186–3197.
  26. Wheate NJ, Walker S, Craig GE, Oun R. The status of platinum anticancer drugs… Dalton Trans. 2010;39:8113–8127.
  27. Thota S, Rodrigues DA, Crans DC, Barreiro EJ. Ru(II) compounds… J Med Chem. 2018;61(14):5805–5821.
  28. De Almeida A, Oliveira C, Correia I. Copper complexes as anticancer agents. Anticancer Agents Med Chem. 2020;20(11):1276–1300.
  29. Ndagi U, Mhlongo N, Soliman ME. Metal complexes in cancer therapy – update… Drug Des Devel Ther. 2017;11:599–616.
  30. Clavel C, Iengo E, Alessio E. BOLD-100 inhibits glycolysis and targets GRP78. Pharmaceutics. 2022;14(1):1.
  31. Smith N, et al. Imaging BOLD-100 with radiolabelled ruthenium isotopes. Dalton Trans. 2024;53:234–242.
  32. Mukherjee S, Chowdhury R. Nanocarrier-mediated delivery of non-platinum metal complexes… Pharm Res. 2024;41(3):221–236.
  33. Kaluderovi? GN, Rilak A, Bugar?i? ŽD. Polymeric nanocarriers for the delivery of non-platinum metallodrugs. Pharmaceutics. 2021;13(7):1096. doi:10.3390/pharmaceutics13071096.
  34. van der Laan T, van den Heuvel E, Moons KGM, et al. Biomarker-guided trials: challenges and practical considerations. Contemp Clin Trials Commun. 2019;16:100493. doi:10.1016/j.conctc.2019.100493.
  35. Liu J, Chen Y, Gasser G, Kepp KP, Liu Z. Immunogenic cell-death-inducing metal complexes for cancer therapy. Angew Chem Int Ed. 2023;62(34):e202300662. doi:10.1002/anie.202300662.
  36. Stathopoulos GP, Antoniou D, Dimitroulis J, et al. Liposomal cisplatin plus paclitaxel vs cisplatin plus paclitaxel in non-small cell lung cancer: a randomized phase III trial. Ann Oncol. 2010;21(11):2227–2232. doi:10.1093/annonc/mdq109.
  37. Dong L, Zhang H, Chen J, et al. The outcast of medicine: metals in medicine—from traditional mineral medicine to modern metallodrugs. Front Pharmacol. 2025;16:1542560. doi:10.3389/fphar.2025.1542560.
  38. Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action and drug resistance. J Exp Pharmacol. 2014;6:113–122. doi:10.2147/JEP.S93414.
  39. Liu Z, Sadler PJ. Metals in cancer research: beyond platinum metallodrugs. ACS Cent Sci. 2024;10(2):242–250. doi:10.1021/acscentsci.3c01340.
  40. Simon R, Roychowdhury S. Implementing personalized cancer therapy. Clin Cancer Res. 2013;19(6):1311–1316. doi:10.1158/1078-0432.CCR-12-2767.
  41. Schilsky RL. Implementing personalized cancer care. Clin Cancer Res. 2014;20(9):2268–2274. doi:10.1158/1078-0432.CCR-13-2658.
  42. Fricker SP. Metal-based drugs: from serendipity to design. Metallomics. 2024;16(1):mfad013. doi:10.1093/mtomcs/mfad013.
  43. Bertrand B, Santini C. A golden era for gold-based anticancer agents? Future Med Chem. 2017;9(17):1889–1900. Doi:10.4155/fmc-2017-0126.
  44. Bruijnincx PCA, Sadler PJ. Metallodrugs are unique: opportunities and challenges of discovery and development. Chem Rev. 2019;119(2):775–808. doi:10.1021/acs.chemrev.8b00271.
  45. ?ilerdži? J, Vu?kovi? I, Nikoli? M, et al. Anticancer activity of metallodrugs and metallizing host-defense peptides—recent advances. Int J Mol Sci. 2024;25(13):7314. doi:10.3390/ijms25137314.
  46. Zeng S, Zhou J, Liu Z, et al. Metallodrugs in the battle against non-small-cell lung cancer. Front Pharmacol. 2023;14:1242488. doi:10.3389/fphar.2023.1242488.
  47. Arranja A, Pathak V, Lacerda M. Liposomal formulations of metallodrugs for cancer therapy. Int J Mol Sci. 2024;25(17):9337. doi:10.3390/ijms25179337.
  48. Wang X, Liu G, Zhang Z, et al. Smart nanoparticles in cancer therapy. Nat Rev Mater. 2023;8(5):358–378. doi:10.1038/s41578-023-00505-0.
  49. Angajala G, Pavan P, Subashini R. Metal nanoparticles as drug delivery carriers in cancer therapy. Emerg Mater. 2022;5:1593–1615. doi:10.1007/s42247-021-00335-x.
  50. Kalu?erovi? GN, Komazec T, Bogojevi? D, et al. Multifunctional nanomaterials functionalized with metal complexes for cancer therapy. Beilstein J Nanotechnol. 2025;16:1037–1068. doi:10.3762/bjnano.16.85.
  51. Nguyen TT, Pathak S, Regmi S, et al. Sensors for cancer detection and diagnosis: recent advances. Sensors (Basel). 2023;23(22):9152. doi:10.3390/s23229152.
  52. Marquart J, Chen EY, Prasad V. Estimation of the percentage of US patients with cancer who benefit from genome-driven oncology. JAMA Oncol. 2018;4(8):1093–1098. doi:10.1001/jamaoncol.2018.1484.
  53. Fricker SP, Slade R. The pharmacology of metallodrugs: absorption, distribution, metabolism, and excretion. Drug Metab Dispos. 2025;53(4):xx–xx. doi:10.1124/dmd.125.090189.
  54. Kelland L. The resurgence of platinum-based cancer chemotherapy. Endocr Relat Cancer. 2007;14(2):R25–R41. doi:10.1677/ERC-06-0245.
  55. Kostova I, Momekov G, Stoytcheva M, et al. Lanthanide complexes as anticancer agents: an emerging class. Curr Med Chem Anticancer Agents. 2005;5(6):591–602. doi:10.2174/156801105774574694.

Reference

  1. Alessio E. Thirty years of the drug candidate NAMI-A and the myths in the field of ruthenium anticancer compounds. Met Ions Life Sci. 2018;18:233-254.
  2. Hartinger CG, Dyson PJ. Bioorganometallic chemistry—from teaching paradigms to medicinal applications. Chem Soc Rev. 2021;50(8):5301-5365.
  3. Casini A, Reedijk J. Challenges and promises for ruthenium anticancer drugs. Curr Top Med Chem. 2020;20(4):270-282.
  4. Heffeter P, et al. NAMI-A and KP1019—ruthenium anticancer drugs in different phases of clinical trials. Bioinorg Chem Appl. 2018;2018:1-15.
  5. Sava G, Bergamo A. Ruthenium-based drugs for cancer therapy: An update. Curr Med Chem. 2020;27(9):1466-1482.
  6. Bindoli A, Rigobello MP. Principles in redox signaling: From chemistry to functional significance. Antioxid Redox Signal. 2019;31(10):625-649.
  7. Gandin V, et al. Gold compounds as anticancer agents: A systematic review. Cancer Chemother Pharmacol. 2021;88(3):447-468.
  8. Moraes GC, et al. Anticancer activity of copper complexes. J Inorg Biochem. 2022;233:111839.
  9. Ang WH, Dyson PJ. Classical and non-classical ruthenium-based anticancer drugs: Towards targeted chemotherapy. Eur J Inorg Chem. 2020;2020(14):1541-1556.
  10. Pichler V, et al. KP1019 and NKP-1339: Ruthenium anticancer drug candidates. Inorganics. 2021;9(5):36.
  11. Zou T, et al. Recent advances in platinum-free metallodrugs for cancer therapy. Coord Chem Rev. 2022;469:214652.
  12. Bertrand B, Casini A. A golden future in medicinal inorganic chemistry: The promise of gold-based therapeutics. Dalton Trans. 2021;50(25):8709-8725.
  13. Fikselová M, et al. Biological activities of copper-based complexes in oncology. Molecules. 2023;28(3):1025.
  14. Melchart M, et al. Titanium-based anticancer drugs: Past, present, and future. J Med Chem. 2020;63(22):12531-12544.
  15. Savi? N, et al. Palladium complexes in anticancer therapy. Coord Chem Rev. 2021;436:213854.
  16. Liu Z, et al. Cyclometalated iridium(III) complexes for photodynamic therapy. Chem Rev. 2020;120(9):5150-5190.
  17. Streciwilk W, et al. Osmium complexes in medicinal chemistry. Dalton Trans. 2019;48(34):12899-12914.
  18. Zhou Q, et al. Ruthenium complexes targeting DNA in cancer therapy. ChemMedChem. 2020;15(19):1778-1790.
  19. Wang X, et al. Thioredoxin reductase inhibition by gold complexes. J Inorg Biochem. 2021;225:111580.
  20. Kalinowski DS, et al. ROS generation and metal complexes in cancer therapy. Free Radic Biol Med. 2022;170:1-21.
  21. Zhao J, et al. Photodynamic applications of metal complexes. Coord Chem Rev. 2021;438:213885.
  22. Wang J, et al. Apoptotic pathways induced by non-platinum metal complexes. Biochim Biophys Acta Mol Cell Res. 2020;1867(10):118789.
  23. Monro S, Colón KL, Yin H, et al. Transition metal complexes and photodynamic therapy… Chem Rev. 2019;119(2):797–828.
  24. Graf N, Lippard SJ. Redox activation of metal-based prodrugs. Adv Drug Deliv Rev. 2012;64(11):993–1004.
  25. Messori L, Merlino A. Gold and silver complexes with antimicrobial and anticancer activity… Chem Commun. 2016;52(18):3186–3197.
  26. Wheate NJ, Walker S, Craig GE, Oun R. The status of platinum anticancer drugs… Dalton Trans. 2010;39:8113–8127.
  27. Thota S, Rodrigues DA, Crans DC, Barreiro EJ. Ru(II) compounds… J Med Chem. 2018;61(14):5805–5821.
  28. De Almeida A, Oliveira C, Correia I. Copper complexes as anticancer agents. Anticancer Agents Med Chem. 2020;20(11):1276–1300.
  29. Ndagi U, Mhlongo N, Soliman ME. Metal complexes in cancer therapy – update… Drug Des Devel Ther. 2017;11:599–616.
  30. Clavel C, Iengo E, Alessio E. BOLD-100 inhibits glycolysis and targets GRP78. Pharmaceutics. 2022;14(1):1.
  31. Smith N, et al. Imaging BOLD-100 with radiolabelled ruthenium isotopes. Dalton Trans. 2024;53:234–242.
  32. Mukherjee S, Chowdhury R. Nanocarrier-mediated delivery of non-platinum metal complexes… Pharm Res. 2024;41(3):221–236.
  33. Kaluderovi? GN, Rilak A, Bugar?i? ŽD. Polymeric nanocarriers for the delivery of non-platinum metallodrugs. Pharmaceutics. 2021;13(7):1096. doi:10.3390/pharmaceutics13071096.
  34. van der Laan T, van den Heuvel E, Moons KGM, et al. Biomarker-guided trials: challenges and practical considerations. Contemp Clin Trials Commun. 2019;16:100493. doi:10.1016/j.conctc.2019.100493.
  35. Liu J, Chen Y, Gasser G, Kepp KP, Liu Z. Immunogenic cell-death-inducing metal complexes for cancer therapy. Angew Chem Int Ed. 2023;62(34):e202300662. doi:10.1002/anie.202300662.
  36. Stathopoulos GP, Antoniou D, Dimitroulis J, et al. Liposomal cisplatin plus paclitaxel vs cisplatin plus paclitaxel in non-small cell lung cancer: a randomized phase III trial. Ann Oncol. 2010;21(11):2227–2232. doi:10.1093/annonc/mdq109.
  37. Dong L, Zhang H, Chen J, et al. The outcast of medicine: metals in medicine—from traditional mineral medicine to modern metallodrugs. Front Pharmacol. 2025;16:1542560. doi:10.3389/fphar.2025.1542560.
  38. Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action and drug resistance. J Exp Pharmacol. 2014;6:113–122. doi:10.2147/JEP.S93414.
  39. Liu Z, Sadler PJ. Metals in cancer research: beyond platinum metallodrugs. ACS Cent Sci. 2024;10(2):242–250. doi:10.1021/acscentsci.3c01340.
  40. Simon R, Roychowdhury S. Implementing personalized cancer therapy. Clin Cancer Res. 2013;19(6):1311–1316. doi:10.1158/1078-0432.CCR-12-2767.
  41. Schilsky RL. Implementing personalized cancer care. Clin Cancer Res. 2014;20(9):2268–2274. doi:10.1158/1078-0432.CCR-13-2658.
  42. Fricker SP. Metal-based drugs: from serendipity to design. Metallomics. 2024;16(1):mfad013. doi:10.1093/mtomcs/mfad013.
  43. Bertrand B, Santini C. A golden era for gold-based anticancer agents? Future Med Chem. 2017;9(17):1889–1900. Doi:10.4155/fmc-2017-0126.
  44. Bruijnincx PCA, Sadler PJ. Metallodrugs are unique: opportunities and challenges of discovery and development. Chem Rev. 2019;119(2):775–808. doi:10.1021/acs.chemrev.8b00271.
  45. ?ilerdži? J, Vu?kovi? I, Nikoli? M, et al. Anticancer activity of metallodrugs and metallizing host-defense peptides—recent advances. Int J Mol Sci. 2024;25(13):7314. doi:10.3390/ijms25137314.
  46. Zeng S, Zhou J, Liu Z, et al. Metallodrugs in the battle against non-small-cell lung cancer. Front Pharmacol. 2023;14:1242488. doi:10.3389/fphar.2023.1242488.
  47. Arranja A, Pathak V, Lacerda M. Liposomal formulations of metallodrugs for cancer therapy. Int J Mol Sci. 2024;25(17):9337. doi:10.3390/ijms25179337.
  48. Wang X, Liu G, Zhang Z, et al. Smart nanoparticles in cancer therapy. Nat Rev Mater. 2023;8(5):358–378. doi:10.1038/s41578-023-00505-0.
  49. Angajala G, Pavan P, Subashini R. Metal nanoparticles as drug delivery carriers in cancer therapy. Emerg Mater. 2022;5:1593–1615. doi:10.1007/s42247-021-00335-x.
  50. Kalu?erovi? GN, Komazec T, Bogojevi? D, et al. Multifunctional nanomaterials functionalized with metal complexes for cancer therapy. Beilstein J Nanotechnol. 2025;16:1037–1068. doi:10.3762/bjnano.16.85.
  51. Nguyen TT, Pathak S, Regmi S, et al. Sensors for cancer detection and diagnosis: recent advances. Sensors (Basel). 2023;23(22):9152. doi:10.3390/s23229152.
  52. Marquart J, Chen EY, Prasad V. Estimation of the percentage of US patients with cancer who benefit from genome-driven oncology. JAMA Oncol. 2018;4(8):1093–1098. doi:10.1001/jamaoncol.2018.1484.
  53. Fricker SP, Slade R. The pharmacology of metallodrugs: absorption, distribution, metabolism, and excretion. Drug Metab Dispos. 2025;53(4):xx–xx. doi:10.1124/dmd.125.090189.
  54. Kelland L. The resurgence of platinum-based cancer chemotherapy. Endocr Relat Cancer. 2007;14(2):R25–R41. doi:10.1677/ERC-06-0245.
  55. Kostova I, Momekov G, Stoytcheva M, et al. Lanthanide complexes as anticancer agents: an emerging class. Curr Med Chem Anticancer Agents. 2005;5(6):591–602. doi:10.2174/156801105774574694.

Photo
V. Srivarshan
Corresponding author

Department of pharmaceutical chemistry, Kamalakshi Pandurangan college of pharmacy, Ayyampalayam, Tiruvannamalai, Tamilnadu, India 606603.

Photo
A. Arunmozhivarman
Co-author

Department of pharmaceutical chemistry, Kamalakshi Pandurangan college of pharmacy, Ayyampalayam, Tiruvannamalai, Tamilnadu, India 606603.

Photo
M. Chanduru
Co-author

Department of pharmaceutical chemistry, Kamalakshi Pandurangan college of pharmacy, Ayyampalayam, Tiruvannamalai, Tamilnadu, India 606603.

Photo
K. Bakkiyaraj
Co-author

Department of pharmaceutical chemistry, Kamalakshi Pandurangan college of pharmacy, Ayyampalayam, Tiruvannamalai, Tamilnadu, India 606603.

Photo
Dr. R. Rajalingam
Co-author

Department of pharmaceutical chemistry, Kamalakshi Pandurangan college of pharmacy, Ayyampalayam, Tiruvannamalai, Tamilnadu, India 606603.

V. Srivarshan*, A. Arun Mozhi Varman, M. Chanduru, K. Bakkiyaraj, Dr. D. Rajalingam, Next-Generation Metal Complexes in Anticancer Therapy: From Mechanism-Driven Design to Clinical Translation, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 8, 2436-2451 https://doi.org/10.5281/zenodo.16931001

More related articles
In Silico Molecular Docking Investigation to Devel...
Soumallya Chakraborty, Somenath Bhattacharya, Santu Mandal, ...
Nutritional Support In Critical Care: The Role Of ...
Nishigandha Sandip Kharde, Ajit B. Tuwar, Megha Salve, ...
Related Articles
Pharmacovigilance and Over Counter Drugs Ensuring Safety ...
Vrushali Gaikwad , Sonal Kumbhar , Dr.Nilesh B.chougule , ...
Review On Various Analytical Approaches For The Estimation Of Rilpivirine And It...
Chaganti soujanya, Srinija Naluka, Karuna Devi Barla, Durga Panikumar Anumolu, ...
A Novel Validated Stability Indicating Qbd Based Assay Method For The Quantifica...
Dr. Rashid Azeez, Vinod Bairagi, Ziyaurrahman Azeez, ...