Alejandro
Llamedo
ab,
Pablo
Rodríguez
a,
Yaiza
Gabasa
cd,
Raquel G.
Soengas
*b,
Humberto
Rodríguez-Solla
b,
David
Elorriaga
b,
Francisco J.
García-Alonso
*b and
Sara M.
Soto
*cd
aNanovex Biotechnologies S.L., Parque Tecnológico de Asturias Edificio CEEI, 33428 Llanera, Spain
bDepartamento de Química Orgánica e Inorgánica, Instituto Universitario de Química Organometálica “Enrique Moles”, Universidad de Oviedo, Julián Clavería 8, 33006 Oviedo, Spain. E-mail: rsoengas@uniovi.es
cISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
dCIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
First published on 28th August 2024
A novel lipoformulation was developed by encapsulating cationic (S^C)-cyclometallated gold(III) complex [Au(dppta)(N2Py-PZ-dtc)]+ (AuPyPZ) in liposomes. The liposomal form of compound AuPyPZ has a bactericidal action similar to that of the free drug without any appreciable effect on the viability of mammalian cells. Furthermore, the nanoformulation reduces metalloantibiotic-induced inhibition of hERG and the inhibition of cytochromes, significantly decreasing the potential liabilities of the metallodrug. The obtained metalloantibiotic liposomal formulation shows high stability and suitable properties for drug delivery, representing an effective strategy to fight against drug-resistant bacteria.
Despite the clear need for more antimicrobial agents, such drugs have not been forthcoming.7,8 There are currently only 42 antibiotic drugs in clinical development, which is clearly not enough considering the current rate of AMR.9 Furthermore, a closer analysis of their structures reveals an even more pressing problem: most of these compounds are simply variations of well-known antibiotic families, meaning that bacteria will quickly develop resistance to these antibiotics.10 The only long-term solution is to develop new chemical classes of antibiotic drugs to replace the increasingly ineffective ones.11 Since these new generations of antibiotics would have completely new structures, they would be insensitive to existing resistance mechanisms and effective against the most dangerous forms of antibiotic-resistant bacteria.12
Most of the lead compounds in preclinical and clinical antibiotic development share a common structural feature: they are purely organic compounds. However, a radically new approach that has emerged in recent years has been to explore the antimicrobial properties of metal complexes, the so-called metalloantibiotics.13,14 Although yet to be advanced to clinical trial stage, metalloantibiotics have several advantages over purely organic drug candidates.15 The great diversity of ligand types and geometries makes metal-based complexes very useful to access a very little explored chemical space for drug development, especially relevant for the design of new antimicrobials.16 Furthermore, metal-based complexes can provide unique modes of action: ligand exchange or release, redox activation and catalytic generation of toxic species (reactive oxygen species, ROS), as well as depletion of essential substrates, making them capable of abolishing enzyme activities, disrupt membrane function or damage DNA.17 As recently reported, metallodrugs display a 10-fold higher hit-rate towards resistant pathogens compared to purely organic molecules.18 Among the metal-containing species, gold complexes have attracted considerable attention for exploration as metalloantibiotics.19,20 This interest was sparked by the potent antibacterial activity discovered in the FDA-approved drug auranofin (Aur, Ridaura™), a gold(I) complex in clinical use since 1985 for the treatment of severe rheumatoid arthritis (Fig. 1).21 Since then, a wide variety of gold(I) and gold(III) complexes have reported for their antibiotic potential.22 However, despite the potential of gold complexes for antimicrobial drug development, the clinical application of metallodrugs poses some major challenges,23 including limited aqueous solubility and short in vivo half-lives, resulting in inadequate bioavailability and low accumulation at the therapeutic site.24 In addition, several metallodrugs exerted systemic toxicity, mainly related to hepatic damage and cardiotoxic effects.25–27 A general strategy to overcome these limitations focuses on the encapsulation of metallodrugs in nanotechnology-based formulations.28,29 Encapsulation systems limit the interaction of the drug with healthy cells and create a protective home for the drug, dramatically improving toxicity as well as the efficacy of the treatment.30,31
Among the synthetic nanocarriers used to encapsulate metallodrugs, liposomes are by far the most successful. Liposomes have a morphology similar to that of cellular membranes: they are spherical vesicles with an aqueous inner core surrounded by lipid bilayers.32 Liposomes are one of the first nanosized vehicles used in drug delivery applications, as they can encapsulate hydrophilic drugs in an aqueous core and hydrophobic derivatives in the lipidic bilayer.33,34 Liposomes are therefore ideal vehicles for the delivery of metallodrugs, and several studies has been published so far, mainly dedicated to anticancer platinum metallodrugs. In this regard, a liposomal formulation of cisplatin (Lipoplatin®) has reached Phase III clinical trials.35–37 Liposomal formulations for anticancer metallodrugs of ruthenium,38,39 iridium40,41 and gold42 have also received attention.
To address the global shortage of effective antibiotics, our research group is dedicated to the development of novel antibiotic classes based on Au(III) complexes. It was recently reported the synthesis of a series of cationic (dppta)Au(III) complexes (dppta = N,N-diisopropyl-P,P-diphenylphosphinothioic amide-κ2C,S) with κ2S,S’-dithiocarbamate (dtc) auxiliary ligands (AuDTC) displaying potent antibacterial activity against methicillin-resistant S. aureus (MRSA).43
These promising results indicate that the gold(III) (C^S, S^S)-cyclometallated center could be used as a scaffold to develop new classes of antibiotics. However, these metalloantibiotics present a very low aqueous solubility, considerable cytotoxicity in hepatic cell lines and potential cardiotoxicity, which have hampered its clinical application.
Herein we report the synthesis and chemical and biological characterization of a (C^S, S^S)-cycloaurated gold(III) dithiocarbamate complex with improved properties. Furthermore, the encapsulation of this gold(III) complex in liposomes was investigated. The obtained liposomal formulations were physico-chemically characterized and subsequently analyzed for their antibacterial activity, toxicity and relevant pharmacological properties.
The complex was characterized by 1H, 13C{1H} and 31P{1H} NMR, IR, X-ray and HRMS. Complex formation with the dithiocarbamate ligand was confirmed by the upfield shift in the characteristic signal of the H ortho relative to Au (from δH 8.47 to 7.84 ppm) in the 1H NMR. On the other hand, the 31P NMR spectra of AuPyPZ display a significant downfield shift from δP 68.2 to 74.6 ppm. The 13C NMR resonance for the NCS2 carbon appeared at δC 195.0 ppm, the signals corresponding to the pyridine ring at δC 107.0, 113.7, 138.7, 149.5 and 157.8 ppm and the quaternary carbon directly attached to the Au(III) center in the dppta ligand at δC 144.7 ppm (1JPC 27.9 Hz). The band in the IR spectra at 1527 cm−1 is attributed to the delocalized dithiocarbamate NCS2 system, while the band around 840 cm−1 is due to the PF6− anion. The formation of the desired complex is further supported by the ion [M]+ observed in mass spectrometry (calculated for C28H35AuN4PS3+: 751.1421; found: 751.1414). The lack of fragmentation indicates appreciable stability, and is consistent with other gold(III) complexes containing dithiocarbamate ligands.
Slow vapour diffusion of hexane into a concentrated chloroform solution of AuPyPZ gold(III) complex gave crystals suitable for X-ray diffraction. The molecular structure is shown in Fig. 2, and the most significant bond lengths and bond angles are given in Table 1. The unit cell contains the two enantiomers of chiral phosphorous-containing complex AuPyPZ; one of the enantiomers is omitted in Fig. 2 for clarity. Diffraction studies revealed a mononuclear compound where the dithio ligand is κ2-S,S′ coordinated to the gold centre; however, the S2–Au1 and S3–Au1 involved distances are 2.377 Å and 2.235 Å, respectively, and this difference is attributed to the stronger trans influence of the C6H4 aromatic ring compared to that of the S = P moiety. The bite angle of the dithiocarbamate ligand is 74.63°, which is in agreement with the values reported in the literature for related dithiocarbamate complexes.44,45
Bond lengths | |||
---|---|---|---|
Au1–S1 | 2.3229(13) | Au1–S2 | 2.3772(13) |
Au1–S3 | 2.3347(11) | Au1–C13 | 2.0625(5) |
Bond angles | |||
S1–Au1–C18 | 91.10(1) | C13–Au1–S2 | 173.02(1) |
S2–Au1–S1 | 95.89(5) | C13–Au1–S3 | 98.50(14) |
S2–Au1–S3 | 74.63(5) | S1–Au1–S3 | 169.84(5) |
Fig. 2 Molecular structure of complex AuPyPZ. Ellipsoids are shown at 50% probability. Hydrogen atoms, chloroform molecule and PF6 ion are omitted for clarity. |
The metallacycle is slightly displaced from planarity, with the phosphorus atom pointing out the plane with a torsion angle S1–P1–C18–C13 of 26.23°. Still, the value of the four-coordinate geometry index (τ4 = 0.10) is close to the ideal square planar geometry (τ4 = 0).46
In a biomedical context, a truthful chemical characterization requires not only an unambiguous description of the structure but also of the purity. Regardless of development stage, purity assessment is critical whenever chemistry is linked with biological activity and therapeutic application. Impurity analysis in a drug should include organic impurities and inorganic impurities.47 Organic impurities for AuPyPZ complex were analyzed by quantitative NMR (qNMR) using 1,3,5-trimethoxybenzene as internal reference,48 indicating high purity (≥99.5%) and an impurity level below the accepted limits.49 On other hand, inorganic impurities were assessed by ICP-MS, considering the most relevant class 1 elemental impurities (Cd, Hg, Pb) and also Sn, as the preparation of the starting complex involves a Sn–Au transmetallation step. The concentration of elemental impurities is in all cases below the accepted limits for any route of administration, as shown by the results compiled in Table 2.50
Fig. 3 31P{1H} NMR spectra of AuPyPZ in 0.5 mL of (CD3)2SO/PBS (1:1) (a) at t = 0; (b) after heating at 37 °C for 24 h; (c) after heating at 37 °C for 48 h. |
Despite the promising biological profile of gold(III) complexes, their application in pharmacology has been limited by their ability to oxidize biologically relevant molecules. If the metallic centre is not adequately stabilized, Au(III) complexes can be easily reduced to Au(I)/Au(0) in the presence of thiol containing amino acids. This reaction not only results in the loss of the chemical integrity of the complex, but also in the oxidation of sulfur-containing proteins. In fact, the oxidation of thiol containing amino acids by metal ions has been studied for decades because of their important role in both protein structure and function.51 One of the most abundant thiol-containing peptide in biological systems is the tetrapeptide glutathione (GSH, L-γ-Glu-Cys-Gly), present at mM concentrations in mammalian cells. To determine the redox stability of AuPyPZ complex in the presence of biological reductants, the behaviour of the complex in the presence of GSH was studied. A solution of complex AuPyPZ in DMSO-d6 and PBS was treated with an excess of GSH. The 31P NMR spectra of the resulting solution showed that the metallo-complex maintained its chemical integrity (Fig. 4).
Fig. 4 31P{1H} NMR NMR spectra of AuPyPZ in 0.5 mL of (CD3)2SO/PBS (1:1) (a) at t = 0; (b) after addition of aq. GSH (5 equiv.). |
Entry | Bacterial speciesa | No. tested strains | MIC (mg L−1) | ||
---|---|---|---|---|---|
AuPyPZ | CIP | TOB | |||
a MDR, multidrug-resistant bacteria; ATCC, American Type Culture Collection. | |||||
1 | MRSA (MDR clinical) | 4 | 0.5 | 32– > 128 | 2– > 128 |
2 | MSSA (ATCC 25923) | 1 | 1 | 0.5 | 0.5 |
3 | S. epidermidis (MDR clinical) | 3 | 1 | 128 | 0.25–32 |
4 | P. aeruginosa (MDR clinical) | 2 | 16 | 0.5– > 128 | 1– > 128 |
5 | P. aeruginosa (ATCC 27853) | 1 | 16 | 1 | 1 |
6 | S. malthophilia (MDR clinical) | 2 | 4 | 4–64 | 4– > 128 |
7 | E. coli (MDR clinical) | 1 | 8–16 | >128 | >128 |
8 | E. coli (ATCC 25922) | 1 | 8 | 0.25 | 0.25 |
9 | A. baumanii (MDR clinical) | 3 | 2 | >128 | 64– > 128 |
10 | A. baumanii (ATCC 19606) | 1 | 2 | 0.5 | 1 |
Considering the potential of AuPyPZ as a therapeutic antimicrobial agent, we evaluated the in vitro toxicity profile on tumor and non-tumor eukaryotic cell lines from liver tissue. The in vitro cytotoxicity results show an IC50 value of 1.48 μM (1.33 mg L−1) on the tumor cell line Hep G2, whereas the toxicity profile using the non-tumor cell line THLE-2 was slightly less favorable with an IC50 value of 1.12 μM (1.00 mg L−1). Overall, these results point to an acceptable but narrow therapeutic window, with a Therapeutic Index (TI) in the range of 2–4 for S. aureus and S. epidermidis.
Early assessment of absorption, distribution, metabolism and excretion (ADME) properties and safety is critical, not only to guide the initial selection of a drug lead and avoid failures in later stages of development, but also to establish benchmarks against which subsequent drug optimizations can be evaluated. Considering the previous information from other metallodrugs, we decided to evaluate three critical parameters: water solubility, potential cardiotoxicity and cytochrome inhibition.
Oral ingestion is the most convenient route of drug delivery due to its ease of administration, cost effectiveness, and patient comfort.52 However, a major challenge in the design of oral dosage forms of metallodrugs lies in their low bioavailability, mainly related to low solubility. This is the case for many gold metallodrugs, whose poor water solubility has hampered their pharmaceutical use.53 The water solubility determination of AuPyPZ was performed by turbidimetric measurement, showing intermediate to low solubility (11–33 μM).
One of the main reasons for drug withdrawal or drug label revision is drug-induced sudden cardiac death associated with QT prolongation in the electrocardiogram. Inhibition of the hERG (human ether-a-go-go related gene) potassium channel is the most common mechanism responsible for drug-induced QT interval prolongation in humans; therefore, testing the interaction of a compound with the hERG potassium channel is crucial to assess the safety of a drug. Several gold-based drugs showed significant inhibition of the hERG potassium channel, indicating a potential risk for cardiac problems.54AuPyPZ was tested as an hERG potassium channel inhibitor, displaying an IC50 value of 6.2 μM. As drugs with an IC50 < 10 μM are classified as potentially cardiotoxic, AuPyPZ is below safe limits, indicating a potential risk for sudden cardiac death.
Another relevant parameter to determine the safety of a drug compound is the inhibition of cytochrome P450 (CYP450) enzymes, as this is the most common mechanism leading to drug–drug interaction. For early assessment, the inhibition of cytochrome CYP3A4 is used as a reference. In this regard complex AuPyPZ shows a moderate inhibition of CYP3A4, with an IC50 value of 3.57 μM (3.20 mg L−1).
For the preparation of the liposomal formulations loaded with the AuPyPZ metalloantibiotic, the optimal conditions were carefully investigated, selecting the best components to preserve the structure of the encapsulated metallodrug and achieve suitable nanoparticle size, drug loading and release properties. In this regard, liposomes were prepared using thin film hydration method.58 Phosphatidylcholine, 1-hexadecanol and oleic acid in a molar ratio of 64/28/8 were dissolved in ethanol for empty liposomes. In the case of loaded liposomes, AuPyPZ metalloantibiotic was added to the solution. The organic solvent was removed using a rotary evaporator at 50 °C and decreasing the pressure for 45 minutes. Then, the residue was hydrated with 3 mL of ultrapure water and sonicated in an ultrasound bath for 3 minutes. The solution was then homogenised for 5 minutes at 11.000 rpm using a Micron homogenizer, obtaining a yellow solution of liposomes of bilayer structure.
Diameter (nm) | PDI | Zeta potential (mV) | EE (%) | |
---|---|---|---|---|
Unloaded | 185.0 nm ± 19.1 | 0.322 ± 0.070 | −30.9 mV ± 0.8 | — |
AuPyPZ loaded | 181.8 nm ± 25.5 | 0.246 ± 0.023 | 9.54 mV ± 1.22 | 94% ± 2 |
Particle size was determined by dynamic light scattering (DLS), confirming a similar size between unloaded liposomes and gold complex-loaded liposomes. Unloaded liposomes have an average diameter of 185.0 nm ± 19.1 while loaded liposomes had an average diameter of 181.8 nm ± 25.5. The polydispersity index (PDI) is below 0.35, indicating that the size distribution of liposomes is monodisperse.
Zeta potential analysis was performed by combining laser Doppler velocimetry (LDV) and phase analysis light scattering (PALS).59 LDV measurements are performed using a procedure called “mixed mode measurement” (M3).60 Unloaded liposomes exhibit a high negative value of −30.9 mV ± 0.8, whereas loaded liposomes exhibit a positive zeta potential of 9.54 mV ± 1.22. The negative charge of unloaded liposomes can be explained by the presence of oleic acid in the liposome membrane. The gold complex in the liposome membrane modifies this charge from negative to positive.
The efficacy of encapsulation (EE%) of AuPyPZ was determined by high-performance liquid chromatography with ultraviolet/visible light detection (HPLC-UV/VIS). The amount of free metallodrug was analyzed and the EE% was calculated by comparison with the total concentration. The high value found for the encapsulation efficiency (94%) shows that the designed experimental procedure succeeded in incorporating the drug almost completely in the lipoformulation.
Another important feature of liposomes is their stability over time. Stability studies performed over a week (Fig. 5) demonstrated that the liposomal formulations remained stable with no observable clumps or precipitations. Measurements show that there are minimal differences in size, PDI and z-potential over the days measured.
Fig. 5 Stability in (a) size, (b) z-potential and (c) PDI of loaded and unloaded liposomes over a week. |
Entry | Bacterial species | No. tested strains | IC50 (mg L−1) | ||
---|---|---|---|---|---|
Free AuPyPZ | Liposomal AuPyPZ | Unloaded liposomes | |||
1 | MRSA (MDR clinical) | 4 | 0.5 | 0.5–1 | >50 |
2 | MSSA (ATCC 25923) | 1 | 1 | 1 | >50 |
3 | S. epidermidis (MDR clinical) | 3 | 1 | 0.25–0.5 | >50 |
In view of the excellent antibacterial activity of the metalloantibiotic-loaded liposomes, in vitro toxicity was next investigated. Considering that S. aureus and S. epidermidis are responsible for most of the diagnosed resistant skin infections and that liposomal formulations are especially useful for topical use, cell viability for both skin and liver cells was investigated.
As depicted in Table 6, unloaded liposomes have no appreciable effect on cell viability for any of the cell lines studied. Surprisingly, the liposomal metalloantibiotic formulation also has no effect on cell viability, in contrast to the relatively low IC50 values obtained for the free metallodrug for all liver and skin eukaryotic cell lines studied. Even though the antibacterial activity is similar to that of the free drug in most cases, the steep decrease in cytotoxicity results in an optimal Therapeutic Index for the metalloantibiotic liposomal formulation.
Entry | Cell line | IC50 (mg L−1) | ||
---|---|---|---|---|
Free AuPyPZ | Liposomal AuPyPZ | Unloaded liposomes | ||
1 | Hep G2 (tumoral, liver) | 1.33 | >50 | >50 |
2 | THLE-2 (non-tumoral, liver) | 1.00 | >50 | >50 |
3 | CCD-1064-sk (tumoral, skin) | 0.86 | >50 | >50 |
Furthermore, liposomes ameliorate metalloantibiotic-induced inhibition of hERG, significantly decreasing the cardiac liability of the metallodrug. Thus, AuPyPZ-loaded liposomes display a 26 ± 2% inhibition of hERG at a maximum concentration of 1.12 μM. Cytochrome inhibition is also improved by encapsulation, as the metalloantibiotic-loaded liposomes show low CYP3A4 inhibition (27 ± 5% inhibition at a maximum concentration of 1.12 μM).
The minimum inhibitory concentrations (MICs) of AuPyPZ complex were determined in triplicate by the broth microdilution method recommended by the Clinical and Laboratory Standards Institute (CLSI) in 96-well round-bottom microtiter plates. Auranofin, a reference Au(I) metalloantibiotic, was included for comparison. The assays were performed in ISO-Sensitest broth (Oxoid, Madrid, Spain). The plates were incubated at 37 °C and read after 18 h to check the absence of turbidity. The same procedure was performed to determine the MICs of unloaded and AuPyPZ-loaded liposomes. MIC values were defined as the lowest concentration of the compound that inhibited visible growth.
Cytotoxicity was evaluated using the MTT metabolic test. Cells were seeded at a density of 10.000 per well in a 96-wells plate and were incubated in a humidified atmosphere at 37 °C with 5% CO2 for 24 h. Solutions of AuPyPZ and AuPyPZ-loaded liposomes were prepared at 25 mM in 100% DMSO and analyzed in an increasing dose curve from 100 to 0.2 μM. After 72 h of treatment, plates were treated with MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, ACROS Organics) at 5 μg mL−1 in Minimum Essential Medium Eagle (MEM) for 2 h. Then, DMSO was added to the plates to solubilizing the formazan crystals formed in viable cells and plates were stirred for 5 minutes to homogenize the solution. Absorbance was measured at 570 nm by Envision Multiplate Reader (PerkinElmer), and AC50 calculations were done using GeneData Screener Software.
Footnote |
† Electronic supplementary information (ESI) available. CCDC 2361824. For ESI and crystallographic data in CIF or other electronic format see DOI: https://doi.org/10.1039/d4dt01867b |
This journal is © The Royal Society of Chemistry 2024 |