Next Article in Journal
Physiologically Based Pharmacokinetic Modelling to Predict Pharmacokinetics of Enavogliflozin, a Sodium-Dependent Glucose Transporter 2 Inhibitor, in Humans
Next Article in Special Issue
Metal and Metal Oxides Nanoparticles and Nanosystems in Anticancer and Antiviral Theragnostic Agents
Previous Article in Journal
Size-Controllable Nanosystem with Double Responsive for Deep Photodynamic Therapy
Previous Article in Special Issue
One-Step Phytofabrication Method of Silver and Gold Nanoparticles Using Haloxylon salicornicum for Anticancer, Antimicrobial, and Antioxidant Activities
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Platinum-Nucleos(t)ide Compounds as Possible Antimetabolites for Antitumor/Antiviral Therapy: Properties and Perspectives

Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali, Università del Salento, Prov.le Lecce-Monteroni, Centro Ecotekne, 73100 Lecce, Italy
*
Author to whom correspondence should be addressed.
Pharmaceutics 2023, 15(3), 941; https://doi.org/10.3390/pharmaceutics15030941
Submission received: 9 February 2023 / Revised: 6 March 2023 / Accepted: 12 March 2023 / Published: 14 March 2023
(This article belongs to the Special Issue Novel Metal-Based Drugs for Anticancer and Antiviral Applications)

Abstract

:
Nucleoside analogues (NAs) are a family of compounds which include a variety of purine and pyrimidine derivatives, widely used as anticancer and antiviral agents. For their ability to compete with physiological nucleosides, NAs act as antimetabolites exerting their activity by interfering with the synthesis of nucleic acids. Much progress in the comprehension of their molecular mechanisms has been made, including providing new strategies for potentiating anticancer/antiviral activity. Among these strategies, new platinum-NAs showing a good potential to improve the therapeutic indices of NAs have been synthesized and studied. This short review aims to describe the properties and future perspectives of platinum-NAs, proposing these complexes as a new class of antimetabolites.

1. Introduction

Nucleos(t)ide analogues (NAs) antimetabolites are drugs structurally similar to either purine and pyrimidine bases or to the corresponding nucleos(t)ides [1,2,3,4,5]. In this context, the folate cofactors are also considered antimetabolites, because they are involved in several steps of the purine and pyrimidine biosynthesis [6,7]. Drugs of this class have been developed for more than 50 years and, nowadays, are commonly used in the treatment of cancers and viral diseases [2,4,6], Table 1. These antimetabolites act by interfering with the production or function of nucleic acids, RNA and DNA, and preventing cells’ growth and/or survival, since biological systems cannot generally use them in a productive manner [8].
In recent years, for antitumor/antiviral activity, pharmaceutical industries have prepared and evaluated several novel drugs based on nucleos(t)ides analogues (NAs), confirming the ongoing and also applicative interest for this important research line. The increasing knowledge of the physiological mechanisms operating in viral and cancer diseases development and progress helped the design of new active NAs to be addressed against specific biological targets.
The structural and chemical modifications of NAs are generally produced by N-conjugation or azotation and halogenation for the nucleobase, and by hydroxylation or dihydroxylation, methylation, halogenation, ring opening, and saturation for the ribose moiety [2]. In the case of nucleotides, the protection by polar groups or replacement of P-O bonds by P-N bonds may also occur, as shown in Figure 1.
Despite chemical changes, showing a structure very similar to that of endogenous nucleos(t)ides, the NAs can maintain the ability to enter cells by exploiting specific plasma membrane nucleoside transporters [34,35]. Organic cation and/or anion transporters, not specific for nucleosides, can also be involved in the cellular uptake of some NAs [2]. These classes of antitumor and/or antiviral compounds can differ for the types of transporters used and for their preferential interaction with specific targets. This can explain the effectiveness differences observed for these compounds in the treatment of neoplasia with protracted evolution, rapidly proliferating tumors, and viral diseases [36]. A major difference of the NAs used as antiviral drugs, with respect to those used as antitumor agents, consists in a generally lower affinity for the mammalian enzymatic systems. Another, but not secondary, aspect is the lower dosages generally required to produce an effective antiviral activity compared to those necessary for a relevant antitumor activity. Both these characteristics concur in determining a generally better tolerance profile when the NAs are used as antiviral drugs rather than as antitumors.
All NAs use similar mechanisms of action to exert their activity. They enter the cells through an active transport across the cell membrane by using specific membrane transporters. After entering the cells, NAs use the same metabolic pathways of the endogenous nucleos(t)ides [37]. Their mechanism of action includes a first step of phosphorylation, catalyzed from the nucleoside kinase, undergoing production of the nucleoside monophosphate analogue. Two further phosphorylation steps, performed, respectively, by nucleoside monophosphate kinases and nucleoside diphosphate kinases, produce the active triphosphate form [38] (see Figure 2). Cellular or viral nucleic acids can incorporate the triphosphate forms of nucleoside analogues which compete with their counterparts, eventually inhibiting essential enzymes such as human and viral polymerases, therefore interfering with cancer cell growth or viral replication [2].
As for most of the anticancer drugs used in chemotherapy, and also for NAs, some mechanisms of resistance may occur. The main causes of resistance to NAs in both cell lines and clinical samples are (i) inefficient cellular uptake and subsequent deficient intracellular concentration of the NAs triphosphates; (ii) prevented DNA strands and/or dNTP pools damaging; and (iii) faulty apoptosis induction [36,39].
The development of novel drugs candidates able to reduce the resistance, improve the oral bioavailability and overcome the inter-individual variability (which requires a dose adaptation) is still highly desired. One of the employed therapeutic approaches consists in combining the NAs therapy with other agents modulating the apoptotic responses. An example is combination chemotherapy. This approach, adopted for high rate malignant tumors, takes advantage of the combined use of different therapeutic agents, providing additional benefits to the treatments [36]. A multifactorial approach may allow significant progress in the treatment of cancer and viral diseases. In 1997, Mosconi et al. reported the effectiveness of combining gemcitabine and cisplatin in non-small cell lung cancer. By combining gemcitabine, a pyrimidine analogue, with cisplatin, one of the most potent anticancer agents used in the treatment of various solid tumors, the response rates significantly increased with respect to either drug used alone [40]. Nowadays, this combined treatment (cisplatin–gemcitabine) is highly recommend for the treatment of advanced (metastasis) non-small cell lung cancer [41].
In this context, metal-based drugs have shown great flexibility in their potential ranging from anticancer [42,43,44] to antiviral applications [45,46] and several other disorders [47,48]. Differences in ligands coordination to metal may have a great impact on their efficacy, strongly affecting pharmacodynamics and pharmacokinetics profiles. Metallodrugs are often prodrugs which undergo activation by ligand substitution or redox reactions. Both essential and non-essential metals are used in drug therapy and diagnosis [47,48]. Some of the first metallodrugs used in therapy were antimicrobial and antiparasitic agents based on arsenic. In fact, in 1912, Paul Ehrlich and co-workers discovered salvarsan, an arsenic-based drug effective against syphilis, signing the birth of modern chemotherapy [49]. Nowadays, melarsoprol, despite its severe side effect of encephalopathy, is an approved arsenic drug that is still used against trypanosomiasis. Two other types of antimicrobic and antiparasitic drugs are represented by antimony(III) and (IV) metallodrugs (e.g., sodium stibogluconate and melglumine antimoniate), which act against cutaneous and mucocutaneous leishmaniasis [48]. While arsenic- and antimony-based drugs showed high toxicity compared to their therapeutic benefit, bismuth is non-toxic and well tolerated, even at high doses [48]. Bismuth drugs are utilized as a broad-spectrum antibiotic; they have been demonstrated to be effective against antibiotic-resistant bacteria (e.g., Helicobacter pylori) thanks to their ability to bind and inhibit key enzymes such as metallo-β-lactamase (MBL) [50]. Bismuth subcitrate (CBS) or ranitidine bismuth citrate (RBC) are used for the treatment of peptic ulcers that are often associated with H. pylori, while tribromophenate–bismuth(III) (Xeroform) is an antimicrobial agent applied in the treatment of chronic wounds. Again, bismuth subsalicylate (BSS) is a widely used metal-based drug for the treatment of gastrointestinal disorders.
Complexes of copper, gold, and silver have been used extensively, since they demonstrate anticancer, anti-inflammatory, antimicrobial, and antiparasitic activity [51,52]. Silver sulphadiazine is an example of a silver-based drug approved for the prevention and treatment of infections resulting from second- or third-degree burns. On the other side, gold compounds, mainly gold(I) cyanide and thiosulfates, were introduced and approved for rheumatoid arthritis treatment. Among these, we might mention sodium aurothiomalate, aurothioglucose, sodium aurothiopropanol sulfonate, sodium aurothiosulfate, and the much newer gold(I) compound, auranofin (tetraacetyl-β-D-thioglucose-gold(I)-thioethylphosphine), approved by the FDA in 1985. These types of metallodrugs are defined as disease-modifying antirheumatic drugs (DMARDs), being able to slow the progression of rheumatoid arthritis by inhibiting several cathepsins implicated in these diseases [48].
Some metal-based drugs were applied for other types of disorders. For example, the superoxide dismutase-mimicking macrocycle (M40403) is a promising manganese(II) metallodrug candidate for the treatment of cardiovascular disorders [48]. Again, lithium-based drugs represent modern psychopharmacological agents and are the gold standard in the treatment of bipolar disorders and mania [53,54]; a lithium carbonate formulation was first approved in 1979, while a lithium citrate tetrahydrate is currently undergoing clinical trials for the treatment of Huntington’s disease [48].
An important step in the development of metallodrugs was the discovery of cisplatin and its clinical debut in 1978 [55]. Platinum drugs are used in 50% of cancer chemotherapies. Together with cisplatin, carboplatin and oxaliplatin are the only FDA-approved platinum drugs, but there are other platinum complexes which have been approved in single nations, such as nedaplatin, heptaplatin, lobaplatin, miriplatin, and dicycloplatin [56,57]. The mechanism of platinum-based anticancer drugs involves the formation of platinum-DNA adduct and inter- and intra-DNA strand cross-links, which interfere with DNA replication and DNA transcription, causing cell cycle arrest and cell apoptosis/necrosis [58]. Furthermore, platinum complexes have also demonstrated to be good antiviral and antibiotic agents and protect cells from different types of viruses and bacteria [59,60].
Focusing on their applications as anticancer or antiviral, antivirals metal-based drugs can block viral infections, relieve symptoms, regulate the human immune system, and inhibit virus replication enzymes [47]. When used as adjuvants, they can also improve the effectiveness of antiviral drugs and vaccines [61]. Anticancer metal-based drugs can act via metal–DNA or metal–protein adducts formation (thus, through a genomic or non-genomic target), then activating specific mechanisms leading to cancer cell death [62,63,64].
This review aims to highlight the properties and the potential of platinated nucleos(t)ides-based compounds, which may represent a new promising class of antimetabolites with merged properties between those of platinum compounds and those of nucleoside analogues. The perspectives in the treatment of cancer and viral diseases are discussed here.

2. Platinated Nucleos(t)ides

Metal-based antitumor drugs constitute a very successful class of drugs. In this class, the main clinical and therapeutic successes can be referred to the wide family of platinum-based antitumor drugs, starting from the first discovered platinum-based drug named cisplatin [65]. After cisplatin discovery, several platinum complexes have been synthesized but only few of them have been successfully marketed worldwide (e.g., cisplatin, carboplatin, oxaliplatin) as chemotherapeutic drugs [44,66,67,68,69,70]. Cisplatin induces cytotoxic effects through binding DNA, thus interfering with the physiological DNA transcription and/or replication [71,72,73] (see Figure 3).
Platinum compounds act as antiviral agents by interfering with DNA or RNA synthesis to exploit their mechanism of action. Moreover, it has recently been demonstrated that they are able to induce the virus’s death without affecting the host cell [74].
Starting from the wide research interest on platinum drugs as chemotherapeutic agents, thousands of new platinum compounds were synthesized [65,75,76,77,78]. Considering the high reactivity of platinum complexes with the DNA nucleobases, some new platinum-nucleoside analogues were synthesized [79]. These can be considered as special nucleobase analogues, where the modification of the nucleoside mainly consists of a coordination to a metal complex.
Here, we highlight the demonstrated antitumor and antiviral activities of platinated nucleos(t)ides, confirming the adaptability of this class of complexes to the different fields of application (as anticancer and/or antiviral drugs).

2.1. Anticancer Activity

Despite the great progress made in oncology research, the lack of selectivity towards tumors and the high systemic toxicity of many approved drugs make this research field of great interest in the scientific community. The main objectives are focalized on the search for new drugs with a better bioavailability and fewer side effects.
In 1989, Hollis et al. prepared and screened 34 cationic compounds against in vivo murine tumor models [80]. They proposed a series of platinum antitumor agents violating the classical structure–activity relationships, being cationic Pt(II) complexes with three coordinated N-donor ligands, as reported in Figure 4. Among these compounds, some platinum(II)-nucleoside complexes of the type cis-[Pt(NH3)2(Am)Cl]+ demonstrated high stability and solubility in aqueous media and an interesting antitumor potential against different tumors, including Sl80a, P388, and L1210 in mice [80].
In detail, cis-[Pt(NH3)2(N3-cytosine)Cl]Cl and cis-[Pt(NH3)2(N7-2′-deoxyguanosine)Cl]Cl complexes showed the best combination of activity and potency in the S180a screen. In the L1210 leukemia screen, cis-[Pt(NH3)2(N3-cytosine)Cl]Cl and cis-[Pt(NH3)2(N3-cytosine)Cl]Cl demonstrated good antitumor activity. Cis-[Pt(NH3)2(N3-cytosine)Cl]Cl was also found active against P388 leukemia.
Other cis-dichloridoplatinum(II)-N-aminated nucleoside complexes (see Figure 5) were synthesized and tested for their antitumor effect against L1210 in vivo (in mice) and in vitro by Maeda et al. [81].
For all these types of complexes, higher activity and a much lower nephrotoxicity in mice with respect to cisplatin was observed. Examples are the 3-aminocytidine-dichloridoplatinum(II) and 3-amino-2′-deoxycytidine-dichloridoplatinum(II) (see Figure 6), which possess high antitumor activity against L1210 cells in vivo in a wide dose range from 10 to 200 mg/kg. The in vitro cytotoxic effects in L1210 cells demonstrated higher cytocidal effects than the parent not-platinated N-aminated nucleosides. The IC50 values of the complexes were higher than that of cis-DDP by factors of 10 to 103.
Extending the study on this class of cis-dichloridoplatinum(II)-N-aminated nucleoside complexes, researchers also established that their mechanism of action caused the inhibition of DNA and RNA synthesis, as occurs for widely used anticancer/antiviral antimetabolites. Maeda et al. demonstrated the role of sugar moiety in the action mechanism of the considered complexes: the Pt-nucleoside complexes having ribose inhibited RNA synthesis and those having deoxyribose or arabinose inhibited DNA synthesis [81].
Recently, Štarha et al. offered an overview of platinum(II) and platinum(IV) complexes containing adenine as ligands or its derivatives coordinated to platinum through N- or C-donor atom(s) of the adenine moiety [82]. Many complexes of this type have been synthesized, demonstrating the interest of researchers in this new class of nucleoside analogues [83,84]. Complexes containing monodentate N7 coordinating adenine-based ligand(s) are the most numerous with respect to the N6, N9, C8, or bidentate N1,N6, which are relatively rare (see Figure 7).
Summaries of the known biological activities of tested complexes were also reported. For the [PtCl2(Ado-N6,N7)] complex, in vivo tests against mice Sarcoma 180 cells were performed by Cleare and Hoeschele in 1973. Unfortunately, for the mentioned compound, low activity was showed [85].
Baranowska-Kortylewicz et al. studied the in vitro cytotoxicity of new complexes of the type dichlorido(6-aminoethylaminopurine)platinum(II) and dichlorido(6-hydroxyethylaminopurine)platinum(II) against different carcinoma cancer cells, while also investigating nephrotoxicity in the mouse kidney [86]. Despite the lower cytotoxicity on tumor cells, an interesting, relatively lower renal toxicity was also observed compared to cisplatin.
Other synthesized platinum complexes containing adenosine showed anticancer activity in in vitro assays. An example is the antineoplastic agent cis-K4[PtCl2ATP] synthesized by Nayak et al. (see Figure 8), which demonstrated active against Dalton’s lymphoma cells [87].
Kirschner et al. reported that platinum(IV) complex of the antimetabolite 6-mercaptopurine (mp), [Pt(mp2)Cl4], was effective against the murine tumors S180 (Sarcoma 180) [88,89]. In this study, researchers demonstrated that the platinum complex with 6-mercaptopurine as ligand showed a more enhanced anticancer activity than the free ligands. The used ligand, 6-mercaptopurine (see Table 1), is an antimetabolite with known antineoplastic and immunosuppressant properties [90]. It is generally used in combination with other drugs, inhibiting purine metabolism and, as consequence, the nucleic acid synthesis.
Other thio and selenoguanine–platinum complexes were reported from Maeda et al. [89,91], as shown in Figure 9. In detail, aquated cis-diamminoplatinum(II) reacted in slightly acidic conditions with stoichiometric amounts of selenopurine or thiopurine and the resulting complexes were studied for their antitumor activity against L1210 cells in mice. These compounds exhibited a satisfying antitumor activity and very low toxicity. Although their mechanisms of action were not fully understood, the antitumor activity of these complexes seem to also be due to a slow release of the ligands, therefore resulting in a merged action of both the residual platinum complex moiety and the purine analogues.
One of the reasons for the continued study of platinum complexes is the search for new drugs able to circumvent the resistance phenomena that often occurs during cisplatin treatment. In addition to Pt(II) complexes, Pt(IV) complexes also showed high potential as anticancer drugs, even for their ability to work as prodrugs.
Model platinum-nucleobase and -nucleoside complexes were synthesized and studied for their antitumor activity by Ali et al. The mentioned study concerns a series of platinum(II) and (IV) monoadducts of the type [Pt(DACH)LCl]NO3 and [Pt(DACH){trans-(Y)2}LCl]NO3, respectively. A total of 30 platinum monoadducts nucleobases complexes, where DACH = trans-1R,2R-diaminocyclohexane, L = adenine, guanine, hypoxanthine, cytosine, adenosine, guanosine, inosine, cytidine, 9-ethylguanine or 1-methylcytosine, and Y = hydroxo or acetate ligand (see Figure 10), were synthesized.
These platinum-nucleoside monoadducts may also have the ability to further interact with DNA. The cytotoxicity for some of the synthesized compounds against A2780 and A2780CP human ovarian cell lines that were, respectively, sensitive and resistant to cisplatin was also evaluated in comparison with the latter.
Although able to show cytotoxicity on cisplatin-resistant A2780CP cells, also in a low concentration range, the nucleobase adducts proved less effective against A2780 cells then cisplatin (4 to 29 fold). Consistently, cisplatin needs a higher concentration against A2780CP-resistant cells with respect to the sensitive, showing the used A2780CP strain, high levels of resistance (resistance factor of 44.4). Among the nucleobase derivatives, [Pt(DACH){trans-(acetate)2}(9-EtGua)Cl]NO3 proved the most potent and biologically active, with an IC50 of 1.1 and 3.23 µM on the A2780 and A2780CP, respectively. The authors also highlighted the ability of the complex to circumvent the cisplatin resistance [92].

2.2. Antiviral Activity

The appearance of new epidemic viruses requires the search for new effective antiviral drugs. Nucleoside and nucleotide analogues belong to the largest class of agents active as antivirals. These usually follow similar metabolic pathways as the endogenous nucleos(t)ides, creating interferences and alterations of the normal metabolic pathways [1,5,38,93,94,95]. In fact, they are widely used for the treatment of both acute and chronic viral infections since they may inhibit virus replication, by interfering with the synthesis of DNA and/or RNA in infected cells. Nucleos(t)ide analogues drugs generally cause the inhibition of specific intracellular enzymes (e.g., viral polymerases) acting as a substrate for the viral enzymes. In this way, nucleoside/nucleotide analogues can be incorporated into newly synthesized nucleic acids in competition with their endogenous congeners.
The incorporation of nucleoside or nucleotide analogues into DNA and/or RNA can lead to the termination of chain elongation or the occurrence of genetic mutations in the viral progeny [96]. The nucleoside analogues-based drugs can be based on structurally modified nucleos(t)ides, in the nucleobase and/or sugar moieties, with the aim to enhance the biological interactions able to generate antiviral activity [43]. In this optic, Acyclovir (ACV) was the first modified nucleoside-based drug effective for the therapeutic treatment of herpes virus (HSV) infections [97]. For its structural analogies with guanosine, ACV’s coordination ability toward metals generated a wide interest among researchers. Coluccia et al. first reported the synthesis of “hybrid drugs” based on the combination of cis-amine platinum antitumor compounds with antiviral nucleotide analogues [59]. In particular, they investigated the in vitro antiviral properties of cis-[Pt(N7-ACV)Cl(NH3)2]NO3 (cisPt-ACV) on VERO cells infected with HSV-1. The cisPt-ACV complex showed good antiviral activity against HSV-1, albeit less effective than acyclovir. This could be explained by the possible low affinity of the viral thymidine kinase and/or of the host cell kinases for the cisPt-ACV complex compared to the simple ACV. More in-depth studies also showed that the incorporation of monofunctional cisPt-ACV inhibits DNA synthesis and blocks RNA polymerases at platinated guanine residues [98]. Subsequently, Margiotta et al. studied four novel hybrid Pt-complexes containing the phenanthroline (phen) or neocuproine (Me2phen) aromatic diimines as platinum carrier ligands, and the antiviral nucleotide analogues ACV or penciclovir (PEN), coordinated to the metal through N(7). In particular, the [Pt(ACV)2(phen)](NO3)2], [Pt(PEN)2(phen)](NO3)2, [Pt(ACV)2(Me2phen)]I2, and [Pt(PEN)2(Me2phen)](NO3)2 complexes were synthesized and tested against different strains of virus (HSV, HIV, CMV) [99,100]. It emerged from the analysis that ACV-containing compounds had antiviral activity similar to ACV against HSV-1 (strain KOS) and HSV-1 (TK-KOS-ACVr strain). Furthermore, they also seemed to be more effective than ACV against HSV-2. Indeed, the [Pt(ACV)2(Me2phen)]I2 compound showed antiviral activity similar to ACV, but at a concentration five times lower. On the other hand, PEN-containing complexes show a capability to inhibit viral replication similar to that of the sole PEN. All examined complexes induced significant microscopic alteration of cell morphology at concentrations comparable to those of PEN, except for [Pt(PEN)2(Me2phen)](NO3)2 which was less toxic compared to the other tested analogues. Differently, the ACV/PEN and Me2phen-containing complexes demonstrated higher antiviral activity against CMV compared to the antiviral drugs ACV or PEN [59]. On the contrary, all four tested platinum compounds were not effective against HIV. Finally, it is interesting to note that the Me2phen-containing complexes were generally more active with respect to the phen-containing analogues. These results could be ascribed to a different stability in the biological medium of phen and neocuproine derivatives [99].
The nucleoside tubercidin (7-deaza-adenosine), extracted from the bacterium Streptomyces tubercidicus, is a potent antibiotic, antiviral, and antitumor agent, but it is highly toxic against humans. It is phosphorylated by cellular kinases to the corresponding triphosphate, and incorporated into DNA/RNA, causing damage to nucleic acids functions. In addition, tubercidin takes part in several cellular processes, such as pre-mRNA processing, mitochondrial respiration, and the synthesis of purines. In the last few years, many tubercidin analogues were synthetized, with the aim to reduce its strong toxicity. The main modifications affect the C6, C7, and C8 positions of the purine tubercidin. D’Errico et al. tested the anticancer potential of some neutral platinum(II) complexes carrying the cisplatin-like moiety linked to tubercidin through a N-alkyl-amide diamino spacer at the purine C6 position. Thanks to their capability to react more quickly than cisplatin with model duplex DNA chains, it would be interesting to also evaluate the antiviral activity of such complexes [101,102,103,104,105].
Farrell et al. synthesized other platinum-nucleosides analogues having the general structural formula [PtXAmB3-m] (X= anionic ligand, A= acyclic nitrogen containing monodentate ligand, B= nucleobase derivative). They proposed and patented a method based on the administration of Pt-complexes for viral infection treatment and prevention in a host or a patient [106]. Among the examined 9-ethylguanine analogues, trans-[PtCl(N7-9-ethylguanine)(NH3)2]NO3 and trans-[PtCl(N7-9-ethylguanine)(NH3)(quinoline)]NO3 demonstrated protective capabilities in the host cells against HIV infection, after administration [106,107]. The affinity towards S-donor ligands of trans-[PtCl(N7-9-ethylguanine)(NH3)(quinoline)]NO3 complex was exploited to eject zinc from the zinc finger site of the C-terminal finger of the HIV-nucleocapsid protein (NCp7), which is critical for the viral life cycle [107,108]. Therefore, the ability of this platinum-nucleosides analogue to remove zinc makes it a good candidate for antiviral therapy. On the other hand, trans-[PtCl(N7-9-ethylguanine)(NH3)2]NO3 showed good viral inhibition at subtoxic concentration against other type of viruses, such as HCMV, VCV, HSV-1 and 2, EBV, and VZV [107].
Among nucleotide analogues directed against the RNA polymerases of RNA-dependent viruses (i.e., HCV and SARS-CoV-2), remdesivir is the only one that is FDA approved [109]. It is a monophosphoramidate prodrug of an adenosine analogue with a broad antiviral spectrum including negative-sense (filoviruses, paramyxoviruses, pneumoviruses) and positive-sense (coronaviruses) single-stranded viruses [108,110,111]. It also demonstrated to be highly effective against coronaviruses (CoVs), including MERS-Cov and SARS-CoV-2 [94,112,113,114,115,116,117,118,119,120]. Abacavir, zidovudine, lamivudine, tenofovir, stavudine, didanosine, zalcitabine, and emtricitabine are nucleoside/nucleotide analogues approved by the FDA for the treatment of HIV infections. Each of these analogues (except for tenofovir) is administered as a nucleoside and metabolized to a nucleoside triphosphate, becoming active in vivo. They lack a 3′-OH and thereby induce chain termination after being incorporated by HIV reverse transcriptase [109]. Recently, Shahabadi et al. synthetized a new fluorescent Pt-complex containing the anti-HIV didanosine drug ddI (i.e., K[PtCl(OCH3)2(ddI)] complex), with the purpose of designing a new nucleoside analogue with improved clinical efficacy. The application of spectroscopic techniques such as UV absorption, fluorescence spectroscopy, and dynamic viscosity measurements allowed the study of the interaction of K[PtCl(OCH3)2(ddI)] complex with calf thymus DNA (ct-DNA). Experiments showed that this complex can give formation of adducts with DNA through hydrogen and van der Waals interactions [121].
The discovery of “Platinblau” was attributed to Hofmann and Bugge in 1908 [122], who, in aqueous solution, reacted Ag2SO4 with [Pt(CH3CN)2Cl2] and isolated deep blue complexes. Interest in these platinum complexes derived from their antitumor activity, radio-sensitizing action, high water solubility, and low levels of renal toxicity [123,124]. In 1975, Rosenberg and collaborators reported on a new class of “platinum blues” (also called “Platinblau”), obtained by reacting the diaquo species of the antitumor agent cisplatin with pyrimidine nucleobases (such as uracil or thymine, as well as their N1-blocked derivatives, e.g., 1-methyluracil). Their DNA-binding properties and unusually high solubility make these platinum compounds particularly promising for cancer therapy, as well as bacterial and viral infections treatment. Among the studied “Platinblau” compounds, the most interesting was the cis-diaquodiammineplatinum(II)-uracil complex, which was demonstrated to be highly effective against the fowl pox virus (FPV), given its entrance into the cell and inhibition of the viral replication process [124].
The potential of platinum complexes in viral diseases remains an unexplored field. Nevertheless, since the discovery of the antitumor activity of platinum compounds, these molecules were studied worldwide in order to improve their efficacy and selectivity. Considering that most of the currently approved antiviral drugs are based on nucleos(t)ide analogues, the study of the ligand properties of modified nucleos(t)ides towards metal centers is an interesting promising field in the design of novel antivirals [102,121]. DNA is a primary target for many antiviral drugs because inhibition of DNA and/or RNA synthesis during viral replication is an important step, sometimes necessary, for the design of new antiviral drugs. For these reasons, the ability of some nucleos(t)ides-based platinum(II) compounds to interfere with the production, function, and/or metabolism of nucleic acids, even at low dosages, could enhance the efficacy and tolerance profiles of antiviral drugs based on modified nucleos(t)ides [59,107,124].

3. Cellular Processing of Platinated Nucleotides

It is well known that cisplatin, cis-[Pt(NH3)2Cl2], i.e., the most studied platinum-based antitumor drug, affects the replication of tumor cells by bonding purines into DNA in the N7 position [72]. Such interactions seem pivotal for antitumor activity, explaining why most of the studies on cisplatin’s mechanism of action are centered on a better understanding of cisplatin–DNA interactions [125]. On the other hand, the cisplatin analogues are known to bind many biological targets (beside DNA) and for this, a comprehensive understanding of the interactions resulting in therapeutic or side effects is still in progress. The similarity with natural nucleosides makes platinum-nucleos(t)ide complexes suitable biomimetic substrates belonging to the nucleos(t)ide analogous family. Indeed, for some platinum-nucleoside compounds, cellular processing like that of the NAs was hypothesized (see Figure 2). They can act as prodrugs and, once introduced inside cells by the plasma membrane nucleoside transporters, require intracellular phosphorylation to generate their pharmacologically active triphosphate form [38]. Many investigations have focused on the study of the synthesis, structural features, and antitumor and/or antiviral biological effects of platinated nucleotides [74,79,89,126,127,128,129,130,131,132,133]. Recently, our research efforts were devoted to a deeper comprehension of the cellular metabolic mechanisms involving some model platinated nucleos(t)ides, to confirm their potential as antimetabolites. The obtained results are summarized hereafter.

3.1. Incorporation of Model Platinum-Nucleotide Complexes into Newly Synthesized DNA by DNA Polymerases

Nucleotides are physiologically present in the cell cytoplasm for important processes, in particular, for the synthesis of nucleic acids [134]. A requirement to consider platinated nucleotides as antimetabolites is their recognition as substrates from the cellular enzymes, as occurs for the natural counterpart. For this purpose, we synthesized a purine–platinum complex, in the form of [Pt(dien)(5′-dGTP)], to be used as a model for cellular processing studies. The considered compound has a platinum atom attached to the diethylene-triamine tridentate ligand and to a N7-bonded 5′-dGTP (see Figure 11). The lack of labile chlorido ligands in its structure makes the complex unable to give mono- or bis-adducts by direct reaction with DNA, as generally occurs for cisplatin derivatives. Thus, the presence of platinum in the newly synthesized DNA can only be ascribed to the recognition by cell enzymes of the platinated nucleotide, with consequent insertion in the newly synthesized DNA, as occurs for natural nucleotides. Therefore, the [Pt(dien)(5′-dGTP)] complex can work as an antimetabolite.
This was recently studied by some of us, reporting clear experimental evidence that the DNA polymerases can recognize and incorporate the N7-platinated guanines in the in vitro and in vivo synthesis of DNA, to give platinated DNA (Figure 12) [135].
Moreover, by incorporating the metal-modified nucleotides into DNA, a possible site-specific metalation was also proposed. This possibility to obtain a polynucleotide sequence containing a nucleotide covalently linked to metal in a specific position could be of particular interest for different applications ranging from pharmacological to technological [135,136].

3.2. Effects of N7-Platinated Ribonucleotides on RNA Polymerases Activity

DNA and RNA strongly differ in their biological functions in relation to information storage and cellular processing [137]. A limited amount of research has focused on platinum drugs binding to RNA, rather than on the study of the stalling of transcription activities of RNA polymerases induced by platinum complexes when bonded to DNA.
We tested the T7 RNA polymerase for the incorporation of the model compound, i.e., [Pt(dien)(N7-5′-GTP)] (dien = diethylenetriamine; GTP = 5′-guanosine triphosphate), into a natural RNA sequence (Escherichia coli lacZ gene encoding for β-galactosidase) by in vitro transcription [138]. The considered complex is unable to form platinum chelates with RNA. The metalated nucleotide was tested as substrate for T7 RNA polymerase under standard in vitro transcription conditions. Our findings suggest that RNA polymerases, unlike DNA polymerases [135], are unable to incorporate the N7-platinated nucleotides into newly synthesized nucleic acids. This result suggests higher activation energy and selectivity for the NTPs with respect to dNTPs insertion since the nucleotide triphosphate insertion catalyzed by RNA polymerases is generally slower with respect to the analogous reaction promoted by DNA polymerases. Indeed, the RNA polymerases have lower processivity with respect to DNA polymerases and frequently abort the synthesis after the incorporation of the first few nucleotides [139,140]. The use of platinated deoxy ribonucleotides could than offer a strategy to specifically target DNA, without affecting the RNA. This class of compounds could be specifically designed and used as an alternative to the classical platinum-based drugs, with a possible reduction of both systemic toxicity and side effects, generally produced by platinum drugs for the possible direct binding to RNA.

3.3. Plasma Membrane Transport of Platinated Nucleos(t)ides

The uptake across the plasma membrane is one of the fundamental steps at the base of the mechanism of drugs’ pharmacological activity and resistance processes. Nucleotide analogues-induced cell toxicity is frequently limited by inefficient cellular uptake. This can determine a too-low concentration level in the cytoplasm to have a useful pharmacological activity [141]. Indeed, the ability of platinated nucleos(t)ide derivatives to mimic the natural cellular nucleos(t)ides depends on their bioavailability in cells, which is also conditioned by their efficiency of transport at the membrane cell level. Two families of transporters are involved in the natural nucleotides uptake: equilibrative transporters (ENT), which act according to the substrate concentration, and concentrative nucleotide transporters (CNT), which act against the concentration gradients in a Na+-dependent manner. In this frame, we experimentally studied the cellular uptake of three N7-platinated purines, differing in the number of phosphates carried by the nucleoside moiety and overall molecular charge. Such complexes were of the following types: [Pt(dien)(N7-dGuo)]2+ (dGuo = 2′-deoxy-guanosine), [Pt(dien)(N7-5′-dGMP)] (5′-dGMP = 5′-(2′-deoxy)-guanosine monophosphate), and [Pt(dien)(N7-5′-dGTP)]2−. Interestingly, from these experiments, a higher intracellular uptake for the two electrically charged [Pt(dien)(N7-dGuo)]2+ and [Pt(dien)(N7-5′-dGTP)]2− complexes resulted, while the uptake of the neutral [Pt(dien)(N7-5′-dGMP)] was negligible [129]. Consistently, the electrically charged complexes [Pt(dien)(N7-dGuo)]2+ and [Pt(dien)(N7-5′-dGTP)]2− demonstrated higher cytotoxic activity on human cervical adenocarcinoma epithelial cells (HeLa), with respect to the neutral analogue. In detail, the cell membrane transporters involved in the uptake of considered platinated nucleos(t)ides resulted in being Na+-dependent, which occurred for the concentrative transporters of natural nucleosides. These results suggest possible applications of the electrically charged platinated nucleos(t)ides as antiviral/antitumor drugs for more effective therapeutic strategies [142].

3.4. Transport and Incorporation in Mitochondrial DNA (mtDNA) of Platinated Nucleotide Analogues

Mitochondria play an important role in cell death induction [143]. Recently, we also evaluated the possibility of metalated deoxynucleotide analogues being transported into mitochondria and then incorporated into mitochondrial DNA (mtDNA) [144,145]. We first demonstrated the direct involvement of a mitochondrial carrier (Drosophila melanogaster thiamine pyrophosphate carrier protein, DmTpc1) in metalated purines transport [145]. DmTpc1 is a protein of the inner mitochondrial membrane that promotes mitochondrial uptake of thiamine pyrophosphate and nucleotides [146]. It was shown that the DmTpc1 transporter is able to promote the specific uptake of N7-platinated purines in the form of complexes [Pt(dien)(N7-5′-dGTP)]2− (dien = diethylenetriamine; 5′-dGTP = 5′-(2′-deoxy)-guanosine triphosphate) and cis-[Pt(NH3)2(py)(N7-5′-dGTP)]2− (py = pyridine), which are related to the mechanism of action of cisplatin. DmTpc1 showed a lower affinity for the two platinated purines compared to the dATP, which is the preferred substrate. However, the good affinity of DmTpc1 for platinated purines could explain the correlation between the eventual cell toxicity of platinum-bonded nucleos(t)ides and their mitochondrial import. Moreover, it was shown that a lower steric hindrance of platinated nucleos(t)ides could enhance the transport at the mitochondrial membrane level, as demonstrated by the higher uptake of complex [Pt(dien)(N7-5′-dGTP)]2− compared to cis-[Pt(NH3)2(py)(N7-5′-dGTP)]2− [145]. Similar results were obtained by Lunetti et al. [144], even with the affinity of the human mitochondrial deoxynucleotide carrier (DNC) and its possible involvement in the mitochondrial uptake of platinated nucleos(t)ides. Kinetic characterization, followed by ICP-AES (inductively coupled plasma atomic emission spectroscopy), showed that the [Pt(dien)(N7-5′-dGTP)]2− was highly and selectively accumulated into proteoliposomes reconstituted with the purified recombinant DNC, demonstrating the possible involvement of this transporter in the transport of platinated nucleotides across the inner mitochondrial membrane. Even in this case, the cis-[Pt(NH3)2(py)(N7-5′-dGTP)]2− complex is transported less efficiently, probably due to its higher steric hindrance. Finally, experiments in organello were permitted to observe the incorporation of the [Pt(dien)(N7-5′-dGTP)] complex into the mtDNA of freshly prepared rat liver mitochondria by DNA polymerase γ, in a time-dependent manner [144]. Thus, the DNC and the mitochondrial polymerase γ could be directly involved in the cytotoxicity mechanism of platinated nucleobases, suggesting the possibility of applying metalated nucleoside analogues-based drugs in the treatment of a wide range of diseases.

4. Conclusions

Metalated nucleos(t)ides are a new class of potential antitumor/antiviral drugs that merge the antitumor and/or antiviral properties of metal compounds and of nucleos(t)ides analogues antimetabolites in the same compound. The study of this new class of promising antitumor/antiviral compounds is still an unexplored field. This review aimed to collect all the scientific research concerned with the evidenced properties as antitumor/antiviral agents of platinated nucleotide complexes.
As previously discussed, platinum-bonded nucleos(t)ides, considered as nucleoside analogues, could act as biomimetic substrates and be selectively internalized and/or processed in cells. Platinated nucleos(t)ides could follow a pathway similar to that of other nucleoside analogues-based drugs. This entails that after their eventual uptake by membrane transporters (cellular and/or mitochondrial) and after phosphorylation, they could also be recognized by nuclear and/or mitochondrial DNA polymerases and incorporated into newly synthesized DNA (see Figure 13). This could induce cancer cells or viruses’ death.
In conclusion, anticancer and/or antiviral metalated nucleotide analogues could act as a new generation of metal-based drugs, characterized by more specific cellular targets and fewer side effects. The possibility of controlled mechanisms of both cell uptake and cellular processing could be a relevant improvement with respect to common metal-based drugs. Indeed, these latter drugs generally undergo a wide set of mechanisms of action and toxicity often not easily defined and controlled. This situation is at the basis of low specificity and relevant side effects generally shown by metal-based drugs. Low-specificity problems are particularly felt for platinum antitumor drugs, where the reduction and control of side effects remains one of the main issues to be addressed. The studies reviewed here suggest considering platinum-nucleotide adducts as a potential new class of pharmacologically active antitumor/antiviral drugs and metal-based antimetabolites, to be further explored.

Author Contributions

Conceptualization, M.B. and F.P.F.; writing-original draft preparation, M.B., F.D.C.; writing-review and editing, F.D.C., M.B., E.S., E.D.L. and F.P.F.; supervision, M.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created.

Acknowledgments

The authors acknowledge the PON “BIO-D” project “Sviluppo di Biomarcatori Diagnostici per la medicina di precisione e la terapia personalizzata” (ARS01_00876).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mahmoud, S.; Hasabelnaby, S.; Hammad, S.; Sakr, T. Antiviral Nucleoside and Nucleotide Analogs: A Review. J. Adv. Pharm. Res. 2018, 2, 73–88. [Google Scholar] [CrossRef] [Green Version]
  2. Jordheim, L.P.; Durantel, D.; Zoulim, F.; Dumontet, C. Advances in the development of nucleoside and nucleotide analogues for cancer and viral diseases. Nat. Rev. Drug Discov. 2013, 12, 447–464. [Google Scholar] [CrossRef] [PubMed]
  3. Kataev, V.E.; Garifullin, B.F. Antiviral nucleoside analogs. Chem. Heterocycl. 2021, 57, 326–341. [Google Scholar] [CrossRef]
  4. Galmarini, C.M.; Mackey Dumontet, C., Jr.; Dumontet, C. Nucleoside analogues and nucleobases in cancer treatment. Lancet Oncol. 2002, 3, 415–424. [Google Scholar] [CrossRef]
  5. Yates, M.K.; Seley-Radtke, K.L. The evolution of antiviral nucleoside analogues: A review for chemists and non-chemists. Part II: Complex modifications to the nucleoside scaffold. Antivir. Res. 2019, 162, 5–21. [Google Scholar] [CrossRef]
  6. Mathews, C.K. Deoxyribonucleotide metabolism, mutagenesis and cancer. Nat. Rev. Cancer 2015, 15, 528–539. [Google Scholar] [CrossRef]
  7. Kovalev, I.S.; Zyryanov, G.V.; Santra, S.; Majee, A.; Varaksin, M.A.-O.; Charushin, V.A.-O.X. Folic Acid Antimetabolites (Antifolates): A Brief Review on Synthetic Strategies and Application Opportunities. Molecules 2022, 27, 6229. [Google Scholar] [CrossRef]
  8. Wang, F.; Li, P.; Chu, H.C.; Lo, P.K. Nucleic Acids and Their Analogues for Biomedical Applications. Biosensors 2022, 12, 93. [Google Scholar] [CrossRef] [PubMed]
  9. Gearry, R.B.; Barclay, M.L. Azathioprine and 6-mercaptopurine pharmacogenetics and metabolite monitoring in inflammatory bowel disease. J. Gastroenterol. Hepatol. 2005, 20, 1149–1157. [Google Scholar] [CrossRef]
  10. Roberts, C.; Strauss, V.Y.; Kopijasz, S.; Gourley, C.; Hall, M.; Montes, A.; Abraham, J.; Clamp, A.; Kennedy, R.; Banerjee, S.; et al. Results of a phase II clinical trial of 6-mercaptopurine (6MP) and methotrexate in patients with BRCA-defective tumours. Br. J. Cancer 2020, 122, 483–490. [Google Scholar] [CrossRef] [Green Version]
  11. Ali, S.S.; Raj, R.; Kaur, T.; Weadick, B.; Nayak, D.; No, M.; Protos, J.; Odom, H.; Desai, K.; Persaud, A.K.; et al. Solute Carrier Nucleoside Transporters in Hematopoiesis and Hematological Drug Toxicities: A Perspective. Cancers 2022, 14, 3113. [Google Scholar] [CrossRef] [PubMed]
  12. Leist, T.P.; Weissert, R. Cladribine: Mode of Action and Implications for Treatment of Multiple Sclerosis. Clin. Neuropharmacol. 2011, 34, 28–35. [Google Scholar] [CrossRef]
  13. Xie, J.; Span, M.; van Maarseveen, E.; Langenhorst, J.; Boddy, A.V.; Sia, K.C.S.; Sutton, R.; Venn, N.; Punt, A.M.; Tyrrell, V.; et al. Optimization of a clofarabine-based drug combination regimen for the preclinical evaluation of pediatric acute lymphoblastic leukemia. Pediatr. Blood Cancer 2020, 67, e28133. [Google Scholar] [CrossRef]
  14. Ewert de Oliveira, B.; Junqueira Amorim, O.H.; Lima, L.L.; Rezende, R.A.; Mestnik, N.C.; Bagatin, E.; Leonardi, G.R. 5-Fluorouracil, innovative drug delivery systems to enhance bioavailability for topical use. J. Drug Deliv. Sci. Technol. 2021, 61, 102155. [Google Scholar] [CrossRef]
  15. Di Francia, R.; Crisci, S.; De Monaco, A.; Cafiero, C.; Re, A.; Iaccarino, G.; De Filippi, R.; Frigeri, F.; Corazzelli, G.; Micera, A.; et al. Response and Toxicity to Cytarabine Therapy in Leukemia and Lymphoma: From Dose Puzzle to Pharmacogenomic Biomarkers. Cancers 2021, 13, 966. [Google Scholar] [CrossRef]
  16. Müller, A.; Florek, M. 5-Azacytidine/Azacitidine. In Small Molecules in Oncology; Martens, U.M., Ed.; Springer: Berlin/Heidelberg, Germany, 2010; pp. 159–170. [Google Scholar]
  17. Bedoui, Y.; Guillot, X.; Sélambarom, J.; Guiraud, P.; Giry, C.; Jaffar-Bandjee, M.C.; Ralandison, S.; Gasque, P. Methotrexate an Old Drug with New Tricks. Int. J. Mol. Sci. 2019, 20, 5023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. de Rouw, N.; Piet, B.; Derijks, H.J.; van den Heuvel, M.M.; ter Heine, R. Mechanisms, Management and Prevention of Pemetrexed-Related Toxicity. Drug Saf. 2021, 44, 1271–1281. [Google Scholar] [CrossRef] [PubMed]
  19. Andrei, G.; Snoeck, R. Advances and Perspectives in the Management of Varicella-Zoster Virus Infections. Molecules 2021, 26, 1132. [Google Scholar] [CrossRef]
  20. Pott Junior, H.; de Oliveira, M.F.B.; Gambero, S.; Amazonas, R.B. Randomized clinical trial of famciclovir or acyclovir for the treatment of herpes zoster in adults. Int. J. Infect. Dis. 2018, 72, 11–15. [Google Scholar] [CrossRef] [Green Version]
  21. Märtson, A.-G.; Edwina, A.E.; Kim, H.Y.; Knoester, M.; Touw, D.J.; Sturkenboom, M.G.G.; Alffenaar, J.-W.C. Therapeutic Drug Monitoring of Ganciclovir: Where Are We? Ther. Drug Moni. 2022, 44, 138–147. [Google Scholar] [CrossRef]
  22. Chuchkov, K.; Chayrov, R.; Hinkov, A.; Todorov, D.; Shishkova, K.; Stankova, I.G. Modifications on the heterocyclic base of ganciclovir, penciclovir, acyclovir—Syntheses and antiviral properties. Nucleos. Nucl. Nucl. 2020, 39, 979–990. [Google Scholar] [CrossRef] [PubMed]
  23. Bodilsen, J.; Nielsen, H.; Whitley, R.J. Valaciclovir therapy for herpes encephalitis: Caution advised. J. Antimicrob. Chemother. 2019, 74, 1467–1468. [Google Scholar] [CrossRef] [PubMed]
  24. Murray, J.; Hilbig, A.; Soe, T.T.; Ei, W.L.S.S.; Soe, K.P.; Ciglenecki, I. Treating HIV-associated cytomegalovirus retinitis with oral valganciclovir and intra-ocular ganciclovir by primary HIV clinicians in southern Myanmar: A retrospective analysis of routinely collected data. BMC Infect. Dis. 2020, 20, 842. [Google Scholar] [CrossRef] [PubMed]
  25. Haque, N.; Parveen, S.; Tang, T.; Wei, J.; Huang, Z. Marine Natural Products in Clinical Use. Mar. Drugs 2022, 20, 528. [Google Scholar] [CrossRef] [PubMed]
  26. De Clercq, E. Clinical Potential of the Acyclic Nucleoside Phosphonates Cidofovir, Adefovir, and Tenofovir in Treatment of DNA Virus and Retrovirus Infections. Clin. Microbiol. Rev. 2003, 16, 569–596. [Google Scholar] [CrossRef] [Green Version]
  27. Rabie, A.M. Efficacious Preclinical Repurposing of the Nucleoside Analogue Didanosine against COVID-19 Polymerase and Exonuclease. ACS Omega 2022, 7, 21385–21396. [Google Scholar] [CrossRef]
  28. Li, J.; Hu, C.; Chen, Y.; Zhang, R.; Fu, S.; Zhou, M.; Gao, Z.; Fu, M.; Yan, T.; Yang, Y.; et al. Short-term and long-term safety and efficacy of tenofovir alafenamide, tenofovir disoproxil fumarate and entecavir treatment of acute-on-chronic liver failure associated with hepatitis B. BMC Infect. Dis. 2021, 21, 567. [Google Scholar] [CrossRef]
  29. De Clercq, E. Tribute to John C. Martin at the Twentieth Anniversary of the Breakthrough of Tenofovir in the Treatment of HIV Infections. Viruses 2021, 13, 2410. [Google Scholar] [CrossRef]
  30. Ude, I.N.; Yeh, S.; Shantha, J.G. Cytomegalovirus retinitis in the highly active anti-retroviral therapy era. Ann. Eye Sci. 2022, 7, 5. [Google Scholar] [CrossRef]
  31. Chodosh, J.; Ung, L. Adoption of Innovation in Herpes Simplex Virus Keratitis. Cornea 2020, 39, S7–S18. [Google Scholar] [CrossRef]
  32. Anderson, P.L.; Kiser, J.J.; Gardner, E.M.; Rower, J.E.; Meditz, A.; Grant, R.M. Pharmacological considerations for tenofovir and emtricitabine to prevent HIV infection. J. Antimicrob. Chemother. 2011, 66, 240–250. [Google Scholar] [CrossRef] [Green Version]
  33. Zamora, F.J.; Dowers, E.; Yasin, F.; Ogbuagu, O. Dolutegravir And Lamivudine Combination For The Treatment Of HIV-1 Infection. HIV/AIDS-Res. Palliat. Care 2019, 11, 255–263. [Google Scholar]
  34. Cano-Soldado, P.; Pastor-Anglada, M. Transporters that translocate nucleosides and structural similar drugs: Structural requirements for substrate recognition. Med. Res. Rev. 2012, 32, 428–457. [Google Scholar] [CrossRef]
  35. Pastor-Anglada, M.; Pérez-Torras, S. Emerging Roles of Nucleoside Transporters. Front. Pharmacol. 2018, 9, 606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Galmarini, C.M.; Mackey, J.R.; Dumontet, C. Nucleoside analogues: Mechanisms of drug resistance and reversal strategies. Leukemia 2001, 15, 875–890. [Google Scholar] [CrossRef] [Green Version]
  37. Abdullah Al Awadh, A. Nucleotide and nucleoside-based drugs: Past, present, and future. Saudi J. Biol. Sci. 2022, 29, 103481. [Google Scholar] [CrossRef]
  38. Van Rompay, A.R.; Johansson, M.; Karlsson, A. Substrate specificity and phosphorylation of antiviral and anticancer nucleoside analogues by human deoxyribonucleoside kinases and ribonucleoside kinases. Pharmacol. Ther. 2003, 100, 119–139. [Google Scholar] [CrossRef] [PubMed]
  39. Tsesmetzis, N.; Paulin, C.B.J.; Rudd, S.G.; Herold, N. Nucleobase and Nucleoside Analogues: Resistance and Re-Sensitisation at the Level of Pharmacokinetics, Pharmacodynamics and Metabolism. Cancers 2018, 10, 240. [Google Scholar] [CrossRef] [Green Version]
  40. Yang, Q.; Nie, Y.H.; Cai, M.B.; Li, Z.M.; Zhu, H.B.; Tan, Y.R. Gemcitabine Combined with Cisplatin Has a Better Effect in the Treatment of Recurrent/Metastatic Advanced Nasopharyngeal Carcinoma. Drug Des. Dev. Ther. 2022, 16, 1191–1198. [Google Scholar] [CrossRef]
  41. Mosconi, A.M.; Crinò, L.; Tonato, M. Combination therapy with gemcitabine in non-small cell lung cancer. Eur. J. Cancer 1997, 33, S14–S17. [Google Scholar] [CrossRef]
  42. Englinger, B.; Pirker, C.; Heffeter, P.; Terenzi, A.; Kowol, C.R.; Keppler, B.K.; Berger, W. Metal Drugs and the Anticancer Immune Response. Chem. Rev. 2019, 119, 1519–1624. [Google Scholar] [CrossRef]
  43. 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]
  44. Stefàno, E.; De Castro, F.; De Luca, E.; Muscella, A.; Marsigliante, S.; Benedetti, M.; Fanizzi, F.P. Synthesis and comparative evaluation of the cytotoxic activity of cationic organometallic complexes of the type [Pt(η1-CH2-CH2-OR)(DMSO)(phen)]+ (R = Me, Et, Pr, Bu). Inorg. Chim. Acta 2023, 546, 121321. [Google Scholar] [CrossRef]
  45. Cirri, D.; Pratesi, A.; Marzo, T.; Messori, L. Metallo therapeutics for COVID-19. Exploiting metal-based compounds for the discovery of new antiviral drugs. Expert Opin. Drug Discov. 2020, 16, 39–46. [Google Scholar] [CrossRef] [PubMed]
  46. Marzo, T.; Messori, L. A Role for Metal-Based Drugs in Fighting COVID-19 Infection? The Case of Auranofin. ACS Med. Chem. Lett. 2020, 11, 1067–1068. [Google Scholar] [CrossRef] [PubMed]
  47. Anthony, E.J.; Bolitho, E.M.; Bridgewater, H.E.; Carter, O.W.L.; Donnelly, J.M.; Imberti, C.; Lant, E.C.; Lermyte, F.; Needham, R.J.; Palau, M.; et al. Metallodrugs are unique: Opportunities and challenges of discovery and development. Chem. Sci. 2020, 11, 12888–12917. [Google Scholar] [CrossRef]
  48. Mjos, K.D.; Orvig, C. Metallodrugs in Medicinal Inorganic Chemistry. Chem. Rev. 2014, 114, 4540–4563. [Google Scholar] [CrossRef]
  49. Vernon, G. Syphilis and Salvarsan. Br. J. Gen. Pract. 2019, 69, 246. [Google Scholar] [CrossRef]
  50. Li, H.; Wang, R.; Sun, H. Systems Approaches for Unveiling the Mechanism of Action of Bismuth Drugs: New Medicinal Applications beyond Helicobacter Pylori Infection. Acc. Chem. Res. 2019, 52, 216–227. [Google Scholar] [CrossRef]
  51. Wang, H.; Yang, X.; Wang, M.; Hu, M.; Xu, X.; Yan, A.; Hao, Q.; Li, H.; Sun, H. Atomic differentiation of silver binding preference in protein targets: Escherichia coli malate dehydrogenase as a paradigm. Chem. Sci. 2020, 11, 11714–11719. [Google Scholar] [CrossRef] [PubMed]
  52. de Souza, C.C.; de Azevedo-França, J.A.; Barrias, E.; Cavalcante, S.C.F.; Vieira, E.G.; Ferreira, A.M.D.C.; de Souza, W.; Navarro, M. Silver and copper-benznidazole derivatives as potential antiparasitic metallodrugs: Synthesis, characterization, and biological evaluation. J. Inorg. Biochem. 2023, 239, 112047. [Google Scholar] [CrossRef]
  53. Volkmann, C.; Bschor, T.; Köhler, S. Lithium Treatment Over the Lifespan in Bipolar Disorders. Front. Psychiatry 2020, 11, 377. [Google Scholar] [CrossRef] [PubMed]
  54. Fountoulakis, K.N.; Tohen, M.; Zarate, C.A. Lithium treatment of Bipolar disorder in adults: A systematic review of randomized trials and meta-analyses. Eur. Neuropsychopharmacol. 2022, 54, 100–115. [Google Scholar] [CrossRef] [PubMed]
  55. Frei, A.; Ramu, S.; Lowe, G.J.; Dinh, H.; Semenec, L.; Elliott, A.G.; Zuegg, J.; Deckers, A.; Jung, N.; Bräse, S.; et al. Platinum Cyclooctadiene Complexes with Activity against Gram-positive Bacteria. ChemMedChem 2021, 16, 3165–3171. [Google Scholar] [CrossRef] [PubMed]
  56. Wheate, N.J.; Walker, S.; Craig, G.E.; Oun, R. The status of platinum anticancer drugs in the clinic and in clinical trials. Dalton Trans. 2010, 39, 8113–8127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Famurewa, A.C.; Mukherjee, A.G.; Wanjari, U.R.; Sukumar, A.; Murali, R.; Renu, K.; Vellingiri, B.; Dey, A.; Valsala Gopalakrishnan, A. Repurposing FDA-approved drugs against the toxicity of platinum-based anticancer drugs. Life Sci. 2022, 305, 120789. [Google Scholar] [CrossRef]
  58. Garutti, M.; Pelizzari, G.; Bartoletti, M.; Malfatti, M.C.; Gerratana, L.; Tell, G.; Puglisi, F. Platinum Salts in Patients with Breast Cancer: A Focus on Predictive Factors. Int. J. Mol. Sci. 2019, 20, 3390. [Google Scholar] [CrossRef] [Green Version]
  59. Coluccia, M.; Boccarelli, A.; Cermelli, C.; Portolani, M.; Natile, G. Platinum(II)-Acyclovir Complexes: Synthesis, Antiviral and Antitumour Activity. Met Based Drugs 1995, 2, 249–256. [Google Scholar] [CrossRef] [Green Version]
  60. Bailly, B.; Gorle, A.K.; Dirr, L.; Malde, A.K.; Farrell, N.P.; Berners-Price, S.J.; von Itzstein, M. Platinum complexes act as shielding agents against virus infection. Chem. Commun. 2021, 57, 4666–4669. [Google Scholar] [CrossRef]
  61. Abate, C.; Carnamucio, F.; Giuffrè, O.; Foti, C. Metal-Based Compounds in Antiviral Therapy. Biomolecules 2022, 12, 933. [Google Scholar] [CrossRef]
  62. Jia, S.; Wang, R.; Wu, K.A.-O.; Jiang, H.; Du, Z. Elucidation of the Mechanism of Action for Metal Based Anticancer Drugs by Mass Spectrometry-Based Quantitative Proteomics. Molecules 2019, 24, 581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. De Castro, F.; Stefàno, E.; De Luca, E.; Muscella, A.; Marsigliante, S.; Benedetti, M.; Fanizzi, F.P. A NMR-Based Metabolomic Approach to Investigate the Antitumor Effects of the Novel [Pt(η1-C2H4OMe)(DMSO)(phen)]+(phen = 1,10-Phenanthroline) Compound on Neuroblastoma Cancer Cells. Bioinorg. Chem. Appl. 2022, 2022, 8932137. [Google Scholar] [CrossRef] [PubMed]
  64. De Castro, F.; Benedetti, M.; Antonaci, G.; Del Coco, L.; De Pascali, A.S.; Muscella, A.; Marsigliante, S.; Fanizzi, P.F. Response of Cisplatin Resistant Skov-3 Cells to [Pt(O,O′-Acac)(γ-Acac)(DMS)] Treatment Revealed by a Metabolomic 1H-NMR Study. Molecules 2018, 23, 2301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Benedetti, M.; Girelli, C.R.; Antonucci, D.; De Pascali, S.A.; Fanizzi, F.P. New method for the synthesis of [PtCl{η1-CH2C(O)R}(N-N)] ketonyl derivatives starting from the Zeise’s salt. Inorg. Chim. Acta 2014, 413, 109–114. [Google Scholar] [CrossRef]
  66. Schoch, S.; Gajewski, S.; Rothfuß, J.; Hartwig, A.; Köberle, B. Comparative Study of the Mode of Action of Clinically Approved Platinum-Based Chemotherapeutics. Int. J. Mol. Sci. 2020, 21, 6928. [Google Scholar] [CrossRef]
  67. Tylkowski, B.; Jastrząb, R.; Odani, A. Developments in platinum anticancer drugs. Phys. Sci. Rev. 2018, 3, 20160007. [Google Scholar] [CrossRef]
  68. Sarkisyan, Z.M.; Shkutina, I.V.; Srago, I.A.; Kabanov, A.V. Relevance of Using Platinum-Containing Antitumor Compounds (A Review). Pharm. Chem. J. 2022, 56, 729–735. [Google Scholar] [CrossRef]
  69. De Castro, F.; Vergaro, V.; Benedetti, M.; Baldassarre, F.; Del Coco, L.; Dell’Anna, M.M.; Mastrorilli, P.; Fanizzi, F.P.; Ciccarella, G. Visible Light-Activated Water-Soluble Platicur Nanocolloids: Photocytotoxicity and Metabolomics Studies in Cancer Cells. ACS Appl. Bio Mater. 2020, 3, 6836–6851. [Google Scholar] [CrossRef]
  70. De Castro, F.; Stefàno, E.; Migoni, D.; Iaconisi, G.N.; Muscella, A.; Marsigliante, S.; Benedetti, M.; Fanizzi, F.P. Synthesis and Evaluation of the Cytotoxic Activity of Water-Soluble Cationic Organometallic Complexes of the Type [Pt(η1-C2H4OMe)(L)(Phen)]+ (L = NH3, DMSO.; Phen = 1,10-Phenanthroline). Pharmaceutics 2021, 13, 642. [Google Scholar] [CrossRef]
  71. Johnstone, T.C.; Suntharalingam, K.; Lippard, S.J. The Next Generation of Platinum Drugs: Targeted Pt(II) Agents, Nanoparticle Delivery, and Pt(IV) Prodrugs. Chem. Rev. 2016, 116, 3436–3486. [Google Scholar] [CrossRef] [Green Version]
  72. Martinho, N.; Santos, T.C.B.; Florindo, H.F.; Silva, L.C. Cisplatin-Membrane Interactions and Their Influence on Platinum Complexes Activity and Toxicity. Front. Physiol. 2019, 9, 1898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Gabano, E.; Ravera, M.; Osella, D. The drug targeting and delivery approach applied to pt-antitumour complexes. A coordination point of view. Curr. Med. Chem. 2009, 16, 4544–4580. [Google Scholar] [CrossRef] [PubMed]
  74. De Castro, F.; De Luca, E.; Benedetti, M.; Fanizzi, F.P. Platinum compounds as potential antiviral agents. Coord. Chem. Rev. 2022, 451, 214276. [Google Scholar] [CrossRef]
  75. Benedetti, M.; Antonucci, D.; Girelli, C.R.; Capitelli, F.; Fanizzi, F.P. Reactivity of [PtCl(η2-C2H4)(N-N)]+, N-N=diimine ligand, with phenol derivatives and first comparison between single crystal X-ray structures of syn- and anti-[Pt(N-N)(phenolate)2] rotamers in the solid state. Inorg. Chim. Acta 2014, 409, 427–432. [Google Scholar] [CrossRef]
  76. Benedetti, M.; Barone, C.R.; Girelli, C.R.; Fanizzi, F.P.; Natile, G.; Maresca, L. H/D exchange at sp3 carbons in the coordination sphere of platinum(ii). Dalton Trans. 2014, 43, 3669–3675. [Google Scholar] [CrossRef]
  77. Benedetti, M.; Girelli, C.R.; Antonucci, D.; Fanizzi, F.P. [PtCl(η1-CH2–CH2OR)(NˆN)] and [PtCl(η2-CH2CH2)(NˆN)]+, NˆN = dinitrogen ligand, complexes. Sterical and electronic effects evidenced by NMR analysis. J. Organomet. Chem. 2014, 771, 40–46. [Google Scholar] [CrossRef]
  78. Benedetti, M.; Barone, C.R.; de Pinto, S.; De Castro, F.; Natile, G.; Fanizzi, F.P. Cationic olefin complexes of platinum(II): Aspects of availability and reactivity. Inorg. Chim. Acta 2018, 470, 172–180. [Google Scholar] [CrossRef]
  79. Müller, J. Nucleic acid duplexes with metal-mediated base pairs and their structures. Coord. Chem. Rev. 2019, 393, 37–47. [Google Scholar] [CrossRef]
  80. Hollis, L.S.; Amundsen, A.R.; Stern, E.W. Chemical and biological properties of a new series of cis-diammineplatinum(II) antitumor agents containing three nitrogen donors: Cis-[Pt(NH3)2(N-donor) Cl]+. J. Med. Chem. 1989, 32, 128–136. [Google Scholar] [CrossRef]
  81. Maeda, M.; Abiko, N.; Uchida, H.; Sasaki, T. Synthesis and antitumor activity of cis-dichloroplatinum(II)-N-aminated nucleoside. J. Med. Chem. 1984, 27, 444–449. [Google Scholar] [CrossRef]
  82. Štarha, P.; Vančo, J.; Trávníček, Z. Platinum complexes containing adenine-based ligands: An overview of selected structural features. Coord. Chem. Rev. 2017, 332, 1–29. [Google Scholar] [CrossRef]
  83. Eastman, A. Separation and characterization of products resulting from the reaction of cis-diamminedichloroplatinum(II) with deoxyribonucleosides. Biochemistry 1982, 21, 6732–6736. [Google Scholar] [CrossRef] [PubMed]
  84. Ibáñez, S.; Albertí, F.M.; Sanz Miguel, P.J.; Lippert, B. Exploring the Metal Coordination Properties of the Pyrimidine Part of Purine Nucleobases: Isomerization Reactions in Heteronuclear PtII/PdII of 9-Methyladenine. Inorg. Chem. 2011, 50, 10439–10447. [Google Scholar] [CrossRef] [PubMed]
  85. Cleare, M.J.; Hoeschele, J.D. Studies on the antitumor activity of group VIII transition metal complexes. Part I. Platinum (II) complexes. Bioinorg. Chem. 1973, 2, 187–210. [Google Scholar] [CrossRef]
  86. Baranowska-Kortylewicz, J.; Pavlik, E.J.; Smith, W.T.; Flanigan, R.C.; Van Nagell, J.R.; Ross, D.; Kenady, D.E. Dichloro(6-aminoethylaminopurine)platinum(II) and its hydroxy analogues: Synthesis and preliminary evaluation. Inorg. Chim. Acta 1985, 108, 91–98. [Google Scholar] [CrossRef]
  87. Nayak, K.K.; Bhattacharyya, R.; Maity, P. Synthesis, characterization, and in vitro cytotoxic effects of K4 [PtCl2ATP]. J. Inorg. Biochem. 1991, 41, 293–298. [Google Scholar] [CrossRef]
  88. Kirschner, S.; Wei, Y.K.; Francis, D.; Bergman, J.G. Anticancer and potential antiviral activity of complex inorganic compounds. J. Med. Chem. 1966, 9, 369–372. [Google Scholar] [CrossRef]
  89. Pasini, A.; Zunino, F. New Cisplatin Analogues—On the Way to Better Antitumor Agents. Angew. Chem. Int. Ed. 1987, 26, 615–624. [Google Scholar] [CrossRef]
  90. Vasta, L.M.; Zanetti, R.C.; Parekh, D.S.; Warwick, A.B.; Lieuw, K. A Retrospective Review of Mercaptopurine Metabolism Reveals High Rate of Patients With Suboptimal Metabolites Successfully Corrected With Allopurinol. J. Pediatr. Hematol. Oncol. 2021, 43, e1003–e1009. [Google Scholar]
  91. Maeda, M.; Abiko, N.; Sasaki, T. Synthesis and antitumor activity of seleno- and thio-purines complexed with cis-diamminoplatinum (II). J. Pharmacobiodyn. 1982, 5, 81–87. [Google Scholar] [CrossRef] [Green Version]
  92. Ali, M.S.; Ali Khan, S.R.; Ojima, H.; Guzman, I.Y.; Whitmire, K.H.; Siddik, Z.H.; Khokhar, A.R. Model platinum nucleobase and nucleoside complexes and antitumor activity: X-ray crystal structure of [PtIV(trans-1R,2R-diaminocyclohexane)trans-(acetate)2(9-ethylguanine)Cl]NO3·H2O. J. Inorg. Biochem. 2005, 99, 795–804. [Google Scholar] [CrossRef]
  93. Lapponi, M.J.; Rivero, C.W.; Zinni, M.A.; Britos, C.N.; Trelles, J.A. New developments in nucleoside analogues biosynthesis: A review. J. Mol. Catal. B-Enzym. 2016, 133, 218–233. [Google Scholar] [CrossRef]
  94. De Clercq, E. New Nucleoside Analogues for the Treatment of Hemorrhagic Fever Virus Infections. Chem.-Asian J. 2019, 14, 3962–3968. [Google Scholar] [CrossRef] [PubMed]
  95. Varga, A.; Lionne, C.; Fau-Roy, B.; Roy, B. Intracellular Metabolism of Nucleoside/Nucleotide Analogues: A Bottleneck to Reach Active Drugs on HIV Reverse Transcriptase. Curr. Drug Metab. 2016, 17, 237–252. [Google Scholar] [CrossRef] [PubMed]
  96. Huchting, J. Targeting viral genome synthesis as broad-spectrum approach against RNA virus infections. Antivir. Chem. Chemother. 2020, 28, 2040206620976786. [Google Scholar] [CrossRef] [PubMed]
  97. Chang, W.T.; Liu, P.Y.; Gao, Z.H.; Lee, S.W.; Lee, W.K.; Wu, S.N. Evidence for the Effectiveness of Remdesivir (GS-5734), a Nucleoside-Analog Antiviral Drug in the Inhibition of I (K(M)) or I (K(DR)) and in the Stimulation of I (MEP). Front. Pharmacol. 2020, 11, 1091. [Google Scholar] [CrossRef]
  98. Balcarova, Z.; Kasparkova, J.; Zakovska, A.; Novakova, O.; Sivo, M.F.; Natile, G.; Brabec, V. DNA interactions of a novel platinum drug, cis-[PtCl(NH3)2(N7-acyclovir)]+. Mol. Pharm. 1998, 53, 846–855. [Google Scholar]
  99. Margiotta, N.; Bergamo, A.; Sava, G.; Padovano, G.; de Clercq, E.; Natile, G. Antiviral properties and cytotoxic activity of platinum(II) complexes with 1,10-phenanthrolines and acyclovir or penciclovir. J. Inorg. Biochem. 2004, 98, 1385–1390. [Google Scholar] [CrossRef]
  100. Margiotta, N.; Fanizzi, F.P.; Kobe, J.; Natile, G. Synthesis, Characterisation and Antiviral Activity of Platinum(II) Complexes with 1,10-Phenanthrolines and the Antiviral Agents Acyclovir and Penciclovir. Eur. J. Inorg. Chem. 2001, 2001, 1303–1310. [Google Scholar] [CrossRef]
  101. De Clercq, E.; Li, G. Approved Antiviral Drugs over the Past 50 Years. Clin. Microbiol. Rev. 2016, 29, 695. [Google Scholar] [CrossRef] [Green Version]
  102. D’Errico, S.; Falanga, A.P.; Capasso, D.; Di Gaetano, S.; Marzano, M.; Terracciano, M.; Roviello, G.; Piccialli, G.; Oliviero, G.; Borbone, N. Probing the DNA Reactivity and the Anticancer Properties of a Novel Tubercidin-Pt(II) Complex. Pharmaceutics 2020, 12, 627. [Google Scholar] [CrossRef] [PubMed]
  103. D’Errico, S.; Borbone, N.; Piccialli, V.; Di Gennaro, E.; Zotti, A.; Budillon, A.; Vitagliano, C.; Piccialli, I.; Oliviero, G. Synthesis and Evaluation of the Antitumor Properties of a Small Collection of PtII Complexes with 7-Deazaadenosine as Scaffold. Eur. J. Org. Chem. 2017, 2017, 4935–4947. [Google Scholar] [CrossRef]
  104. D’Errico, S.; Oliviero, G.; Borbone, N.; Di Gennaro, E.; Zotti, A.I.; Budillon, A.; Cerullo, V.; Nici, F.; Mayol, L.; Piccialli, V.; et al. Synthesis and Evaluation of the Antiproliferative Properties of a Tethered Tubercidin–Platinum(II) Complex. Eur. J. Org. Chem. 2015, 2015, 7550–7556. [Google Scholar] [CrossRef]
  105. Ami, E.-i.; Ohrui, H. Intriguing Antiviral Modified Nucleosides: A Retrospective View into the Future Treatment of COVID-19. ACS Med. Chem. Lett. 2021, 12, 510–517. [Google Scholar] [CrossRef]
  106. Farrell, N.; Bierbach, U. Preparation of Platinum Nucleobase Complexes with Anti-Viral Activity and Method of Using Same. WO9964019A1, 16 December 1999. [Google Scholar]
  107. Sartori, D.A.; Miller, B.; Bierbach, U.; Farrell, N. Modulation of the chemical and biological properties of trans platinum complexes: Monofunctional platinum complexes containing one nucleobase as potential antiviral chemotypes. J. Biol. Inorg. Chem. 2000, 5, 575–583. [Google Scholar] [CrossRef]
  108. Wang, M.; Cao, R.; Zhang, L.; Yang, X.; Liu, J.; Xu, M.; Shi, Z.; Hu, Z.A.-O.; Zhong, W.; Xiao, G. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020, 30, 269–271. [Google Scholar] [CrossRef]
  109. Johnson, K.A.; Dangerfield, T. Mechanisms of inhibition of viral RNA replication by nucleotide analogs. Enzymes 2021, 49, 39–62. [Google Scholar]
  110. Tchesnokov, E.P.; Feng, J.Y.; Porter, D.P.; Götte, M. Mechanism of Inhibition of Ebola Virus RNA-Dependent RNA Polymerase by Remdesivir. Viruses 2019, 11, 326. [Google Scholar] [CrossRef] [Green Version]
  111. Gordon, C.J.; Tchesnokov, E.P.; Feng, J.Y.; Porter, D.P.; Götte, M. The antiviral compound remdesivir potently inhibits RNA-dependent RNA polymerase from Middle East respiratory syndrome coronavirus. J. Biol. Chem. 2020, 295, 4773–4779. [Google Scholar] [CrossRef] [Green Version]
  112. Beigel, J.A.-O.; Tomashek, K.M.; Dodd, L.E.; Mehta, A.K.; Zingman, B.S.; Kalil, A.A.-O.; Hohmann, E.; Chu, H.Y.; Luetkemeyer, A.; Kline, S.; et al. Remdesivir for the Treatment of COVID-19—Final Report. N. Engl. J. Med. 2020, 383, 1813–1826. [Google Scholar] [CrossRef]
  113. Ferren, M.; Horvat, B.; Mathieu, C. Measles Encephalitis: Towards New Therapeutics. Viruses 2019, 11, 1017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Hoenen, T.; Groseth, A.; Feldmann, H. Therapeutic strategies to target the Ebola virus life cycle. Nat. Rev. Microbiol. 2019, 17, 593–606. [Google Scholar] [CrossRef] [PubMed]
  115. Al-Tawfiq, J.A.; Al-Homoud, A.H.; Memish, Z.A. Remdesivir as a possible therapeutic option for the COVID-19. Travel Med. Infect. Dis. 2020, 34, 101615. [Google Scholar] [CrossRef] [PubMed]
  116. de Wit, E.A.-O.; Feldmann, F.; Cronin, J.; Jordan, R.; Okumura, A.; Thomas, T.; Scott, D.; Cihlar, T.; Feldmann, H. Prophylactic and therapeutic remdesivir (GS-5734) treatment in the rhesus macaque model of MERS-CoV infection. Proc. Natl. Acad. Sci. USA 2020, 117, 6771–6776. [Google Scholar] [CrossRef] [Green Version]
  117. Dong, Y.; Mo, X.; Hu, Y.; Qi, X.; Jiang, F.; Jiang, Z.; Tong, S. Epidemiology of COVID-19 Among Children in China. Pediatrics 2020, 145, e20200702. [Google Scholar] [CrossRef] [Green Version]
  118. Dong, L.; Hu, S.; Gao, J. Discovering drugs to treat coronavirus disease 2019 (COVID-19). Drug Discov. Ther. 2020, 14, 58–60. [Google Scholar] [CrossRef] [Green Version]
  119. Khot, W.Y.; Nadkar, M.Y. The 2019 Novel Coronavirus Outbreak—A Global Threat. J. Assoc. Physicians India 2020, 68, 67–71. [Google Scholar]
  120. Ko, W.C.; Rolain, J.M.; Lee, N.Y.; Chen, P.L.; Huang, C.T.; Lee, P.I.; Hsueh, P.R. Arguments in favour of remdesivir for treating SARS-CoV-2 infections. Int. J. Antimicrob. Agents. 2020, 55, 105933. [Google Scholar] [CrossRef]
  121. Shahabadi, N.; Abbasi, A.R.; Moshtkob, A.; Hadidi, S. Design, synthesis and DNA interaction studies of new fluorescent platinum complex containing anti-HIV drug didanosine. J. Biomol. Struct. Dyn. 2020, 38, 2837–2848. [Google Scholar] [CrossRef]
  122. Hofmann, K.A.; Bugge, G. Platinblau. Ber. Der Dtsch. Chem. Ges. 1908, 41, 312–314. [Google Scholar] [CrossRef] [Green Version]
  123. Pullen, S.; Hiller, W.G.; Lippert, B. Regarding the diamagnetic components in Rosenberg’s “platinum pyrimidine blues”: Species in the cis-Pt(NH3)2-1-methyluracil system. Inorg. Chim. Acta 2019, 494, 168–180. [Google Scholar] [CrossRef]
  124. Rosenberg, B.; Mansy, S.A.L.A.; Van Camp, L.L.; Peresie, H.J.; Fischer, R.G.; Davidson, J.P. Platinum-(2,4-dioxopyrimidine) Complex. U.S. Patent 4,419,351, 1 April 1976. [Google Scholar]
  125. Gąsior-Głogowska, M.; Malek, K.; Zajac, G.; Baranska, M. A new insight into the interaction of cisplatin with DNA: ROA spectroscopic studies on the therapeutic effect of the drug. Analyst 2016, 141, 291–296. [Google Scholar] [CrossRef] [PubMed]
  126. Kowalski, K. Organometallic nucleosides—Synthesis, transformations, and applications. Coord. Chem. Rev. 2021, 432, 213705. [Google Scholar] [CrossRef]
  127. Sigel, A.; Sigel, H.; Sigel, R.K.O. Coordination Chemistry of Nucleotides and Antivirally Active Acyclic Nucleoside Phosphonates, including Mechanistic Considerations. Molecules 2022, 27, 2625. [Google Scholar] [CrossRef] [PubMed]
  128. Lippert, B.; Sanz Miguel, P.J. Beyond sole models for the first steps of Pt-DNA interactions: Fundamental properties of mono(nucleobase) adducts of PtII coordination compounds. Coord. Chem. Rev. 2022, 465, 214566. [Google Scholar] [CrossRef]
  129. Benedetti, M.; Antonucci, D.; De Castro, F.; Girelli, C.R.; Lelli, M.; Roveri, N.; Fanizzi, F.P. Metalated nucleotide chemisorption on hydroxyapatite. J. Inorg. Biochem. 2015, 153, 279–283. [Google Scholar] [CrossRef] [PubMed]
  130. Benedetti, M.; De Castro, F.; Fanizzi, F.P. Square-Planar PtII versus Octahedral PtIV Halido Complexes: 195Pt NMR Explained by a Simple Empirical Approach. Eur. J. Inorg. 2016, 2016, 3957–3962. [Google Scholar] [CrossRef]
  131. Lippert, B.; Sanz Miguel, P.J. Chapter Six—Merging Metal–Nucleobase Chemistry with Supramolecular Chemistry. In Advances in Inorganic Chemistry; van Eldik, R., Puchta, R., Eds.; Academic Press: Cambridge, MA, USA, 2018; Volume 71, pp. 277–326. [Google Scholar]
  132. Kahlfuss, C.; Starck, E.; Tufenkjian, E.; Kyritsakas, N.; Jouaiti, A.; Baudron, S.A.; Hosseini, M.W.; Bulach, V. Construction of hydrogen bonding and coordination networks based on ethynylpyridine-appended nucleobases. CrystEngComm 2021, 23, 944–954. [Google Scholar] [CrossRef]
  133. Bertuzzi, D.L.; Perli, G.; Braga, C.B.; Ornelas, C. Synthesis, characterization, and anticancer activity of folate γ-ferrocenyl conjugates. New J. Chem. 2020, 44, 4694–4703. [Google Scholar] [CrossRef]
  134. Diehl, F.F.; Miettinen, T.P.; Elbashir, R.; Nabel, C.S.; Darnell, A.M.; Do, B.T.; Manalis, S.R.; Lewis, C.A.; Vander Heiden, M.G. Nucleotide imbalance decouples cell growth from cell proliferation. Nat. Cell Biol. 2022, 24, 1252–1264. [Google Scholar] [CrossRef]
  135. Benedetti, M.; Ducani, C.; Migoni, D.; Antonucci, D.; Vecchio, V.M.; Ciccarese, A.; Romano, A.; Verri, T.; Ciccarella, G.; Fanizzi, F.P. Experimental Evidence That a DNA Polymerase Can Incorporate N7-Platinated Guanines To Give Platinated DNA. Angew. Chem. Int. Ed. 2008, 47, 507–510. [Google Scholar] [CrossRef] [PubMed]
  136. Benedetti, M.; Ducani, C.; Migoni, D.; Antonucci, D.; Vecchio, V.M.; Romano, A.; Verri, T.; Fanizzi, F.P. Possible Incorporation of Free N7-Platinated Guanines in DNA by DNA Polymerases, Relevance for the Cisplatin Mechanism of Action. In Platinum and Other Heavy Metal Compounds in Cancer Chemotherapy: Molecular Mechanisms and Clinical Applications; Bonetti, A., Leone, R., Muggia, F.M., Howell, S.B., Eds.; Humana Press: Totowa, NJ, USA, 2009; pp. 125–132. [Google Scholar]
  137. Minchin, S.; Lodge, J. Understanding biochemistry: Structure and function of nucleic acids. Essays Biochem. 2019, 63, 433–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Benedetti, M.; Romano, A.; De Castro, F.; Girelli, C.R.; Antonucci, D.; Migoni, D.; Verri, T.; Fanizzi, F.P. N7-platinated ribonucleotides are not incorporated by RNA polymerases. New perspectives for a rational design of platinum antitumor drugs. J. Inorg. Biochem. 2016, 163, 143–146. [Google Scholar] [CrossRef] [PubMed]
  139. Hieb, A.R.; Baran, S.; Goodrich, J.A.; Kugel, J.F. An 8 nt RNA triggers a rate-limiting shift of RNA polymerase II complexes into elongation. EMBO J. 2006, 25, 3100–3109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Wang, Q.; Tullius, T.D.; Levin, J.R. Effects of Discontinuities in the DNA Template on Abortive Initiation and Promoter Escape by Escherichia coli RNA Polymerase. J. Biol. Chem. 2007, 282, 26917–26927. [Google Scholar] [CrossRef] [Green Version]
  141. Eljack, N.D.; Ma, H.-Y.M.; Drucker, J.; Shen, C.; Hambley, T.W.; New, E.J.; Friedrich, T.; Clarke, R.J. Mechanisms of cell uptake and toxicity of the anticancer drug cisplatin. Metallomics 2014, 6, 2126–2133. [Google Scholar] [CrossRef] [Green Version]
  142. De Castro, F.; De Luca, E.; Girelli, C.R.; Barca, A.; Romano, A.; Migoni, D.; Verri, T.; Benedetti, M.; Fanizzi, F.P. First evidence for N7-Platinated Guanosine derivatives cell uptake mediated by plasma membrane transport processes. J. Inorg. Biochem. 2022, 226, 111660. [Google Scholar] [CrossRef]
  143. Tian, C.; Liu, Y.; Li, Z.; Zhu, P.; Zhao, M. Mitochondria Related Cell Death Modalities and Disease. Front. Cell Dev. Biol. 2022, 10, 832356. [Google Scholar] [CrossRef]
  144. Lunetti, P.; Romano, A.; Carrisi, C.; Antonucci, D.; Verri, T.; De Benedetto, G.E.; Dolce, V.; Fanizzi, F.P.; Benedetti, M.; Capobianco, L. Platinated Nucleotides are Substrates for the Human Mitochondrial Deoxynucleotide Carrier (DNC) and DNA Polymerase γ: Relevance for the Development of New Platinum-Based Drugs. ChemistrySelect 2016, 1, 6616. [Google Scholar] [CrossRef] [Green Version]
  145. Carrisi, C.; Antonucci, D.; Lunetti, P.; Migoni, D.; Girelli, C.R.; Dolce, V.; Fanizzi, F.P.; Benedetti, M.; Capobianco, L. Transport of platinum bonded nucleotides into proteoliposomes, mediated by Drosophila melanogaster thiamine pyrophosphate carrier protein (DmTpc1). J. Inorg. Biochem. 2014, 130, 28–31. [Google Scholar] [CrossRef]
  146. Curcio, R.; Lunetti, P.; Zara, V.; Ferramosca, A.A.-O.; Marra, F.; Fiermonte, G.; Cappello, A.R.; De Leonardis, F.; Capobianco, L.; Dolce, V. Drosophila melanogaster Mitochondrial Carriers: Similarities and Differences with the Human Carriers. Int. J. Mol. Sci. 2020, 21, 6052. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation indicating the most common chemical modifications of nucleobases, nucleosides, and nucleotides suitable to give pharmacologically active antitumor and/or antiviral analogues. Adapted from [2], published by Nature Publishing Group, 2013.
Figure 1. Schematic representation indicating the most common chemical modifications of nucleobases, nucleosides, and nucleotides suitable to give pharmacologically active antitumor and/or antiviral analogues. Adapted from [2], published by Nature Publishing Group, 2013.
Pharmaceutics 15 00941 g001
Figure 2. Mechanism of action of NAs. The NAs generally enter cells crossing the cell membrane by an active transport mechanism operated by plasma membrane transporters. Once inside cells, the NAs are progressively phosphorylated and so produce the final triphosphate derivatives.
Figure 2. Mechanism of action of NAs. The NAs generally enter cells crossing the cell membrane by an active transport mechanism operated by plasma membrane transporters. Once inside cells, the NAs are progressively phosphorylated and so produce the final triphosphate derivatives.
Pharmaceutics 15 00941 g002
Figure 3. Schematic representation of cisplatin mechanism of action.
Figure 3. Schematic representation of cisplatin mechanism of action.
Pharmaceutics 15 00941 g003
Figure 4. Chemical structure of cationic triamine complexes. A = ammonia; Am = heterocyclic amine based on pyridine, pyrimidine, purine, piperidine, or saturated amine.
Figure 4. Chemical structure of cationic triamine complexes. A = ammonia; Am = heterocyclic amine based on pyridine, pyrimidine, purine, piperidine, or saturated amine.
Pharmaceutics 15 00941 g004
Figure 5. Schematic representation of the cis-dichloridoplatinum(II)-N-aminated nucleoside complexes. (a) Pt-cytosine analogues; (b) Pt-guanosine-like derivatives. R = deoxyribosyl, ribofuranosyl, arabinofuranosyl.
Figure 5. Schematic representation of the cis-dichloridoplatinum(II)-N-aminated nucleoside complexes. (a) Pt-cytosine analogues; (b) Pt-guanosine-like derivatives. R = deoxyribosyl, ribofuranosyl, arabinofuranosyl.
Pharmaceutics 15 00941 g005
Figure 6. Chemical structure of 3-amino-2′-deoxycytidine-dichloridoplatinum(II) and 3-aminocytidine-dichloridoplatinum(II).
Figure 6. Chemical structure of 3-amino-2′-deoxycytidine-dichloridoplatinum(II) and 3-aminocytidine-dichloridoplatinum(II).
Pharmaceutics 15 00941 g006
Figure 7. Structural formula of adenine, with the preferred coordination sites of platinum to nitrogen evidenced. The atom numbering scheme, for nucleotides, is also reported.
Figure 7. Structural formula of adenine, with the preferred coordination sites of platinum to nitrogen evidenced. The atom numbering scheme, for nucleotides, is also reported.
Pharmaceutics 15 00941 g007
Figure 8. Chemical structure of the cis-K4[PtCl2ATP] complex, adapted from [10], published by Nature Publishing Group, 2020.
Figure 8. Chemical structure of the cis-K4[PtCl2ATP] complex, adapted from [10], published by Nature Publishing Group, 2020.
Pharmaceutics 15 00941 g008
Figure 9. Schematic representation of the platinum complexes synthesized by Maeda et al.
Figure 9. Schematic representation of the platinum complexes synthesized by Maeda et al.
Pharmaceutics 15 00941 g009
Figure 10. Chemical structures of model platinum(II) and (IV) nucleoside monoadducts of the type (a) [Pt(DACH)LCl]NO3 and (b) [Pt(DACH){trans-(Y)2}LCl]NO3, respectively. DACH = trans-1R,2R-diaminocyclohexane; L = adenine, guanine, hypoxanthine, cytosine, adenosine, guanosine, inosine, cytidine, 9-ethylguanine, or 1-methylcytosine; Y = OH, acetate.
Figure 10. Chemical structures of model platinum(II) and (IV) nucleoside monoadducts of the type (a) [Pt(DACH)LCl]NO3 and (b) [Pt(DACH){trans-(Y)2}LCl]NO3, respectively. DACH = trans-1R,2R-diaminocyclohexane; L = adenine, guanine, hypoxanthine, cytosine, adenosine, guanosine, inosine, cytidine, 9-ethylguanine, or 1-methylcytosine; Y = OH, acetate.
Pharmaceutics 15 00941 g010
Figure 11. Schematic representation of the [Pt(dien)(N7-5′-GTP)] and [Pt(dien)(N7-5′-dGTP)], with R = OH and H, respectively.
Figure 11. Schematic representation of the [Pt(dien)(N7-5′-GTP)] and [Pt(dien)(N7-5′-dGTP)], with R = OH and H, respectively.
Pharmaceutics 15 00941 g011
Figure 12. Schematic representation of the insertion mechanism of platinated nucleotides operated by DNA polymerases, during the synthesis of the complementary DNA chain, in the presence of platinated guanines in the nucleotides cellular pool.
Figure 12. Schematic representation of the insertion mechanism of platinated nucleotides operated by DNA polymerases, during the synthesis of the complementary DNA chain, in the presence of platinated guanines in the nucleotides cellular pool.
Pharmaceutics 15 00941 g012
Figure 13. Proposed mechanism of action of platinated nucleos(t)ides.
Figure 13. Proposed mechanism of action of platinated nucleos(t)ides.
Pharmaceutics 15 00941 g013
Table 1. Selection of clinically used antitumor and antiviral nucleos(t)ide analogues (NAs) antimetabolites.
Table 1. Selection of clinically used antitumor and antiviral nucleos(t)ide analogues (NAs) antimetabolites.
Antitumor AntimetabolitesMolecular FormulaClinical UseReferences
Purine analogues
6-MercaptopurineC5H4N4SAcute lymphocytic and acute myelogenous leukemias and small cell non-Hodgkin’s lymphoma[9,10]
FludarabineC10H12FN5O4Chronic lymphocytic leukemia and low-grade lymphomas[11]
CladribineC10H12ClN5O3Hairy cell leukemia and low-grade lymphomas[12]
ClofarabineC10H11ClFN5O3Pediatric acute leukemia[13]
Pyrimidine analogues
5-FluorouracilC4H3FN2O2Head and neck, colon, breast, esophageal, stomach, pancreas, premalignant skin[14]
CytarabineC9H13N3O5Acute lymphocytic leukemia, non-Hodgkin’s lymphoma, and acute myelogenous leukemia[15]
5-AzacytidineC8H12N4O5Myelodysplasia[16]
Folate analogues
MethotrexateC20H22N8O5Acute lymphocytic leukemia, osteosarcoma, bladder cancer, head, neck, and lung cancer, breast[17]
PemetrexedC20H21N5O6Mesothelioma and lung cancer[18]
Antiviral antimetabolites
Purine analogues
AciclovirC8H11N5O3Chickenpox, shingles, herpes virus[19]
FamciclovirC14H19N5O4Shingles[20]
GanciclovirC9H13N5O4Cytomegalovirus (CMV) infection of the eyes in people whose immune system is compromised[21]
PenciclovirC10H15N5O3Herpes simplex virus infections around the mouth (cold sores)[22]
ValaciclovirC13H20N6O4Herpes virus infections, shingles, and herpes simplex in adults[23]
ValganciclovirC14H22N6O5Cytomegalovirus (CMV) retinitis in people who have acquired immunodeficiency syndrome (AIDS)[24]
VidarabineC10H15N5O5Acute keratoconjunctivitis and recurrent epithelial keratitis due to herpes simplex virus types l and 2.[25]
AdefovirC8H12N5O4PChronic (long-term) hepatitis B infection[26]
DidanosineC10H12N4O3Human immunodeficiency virus (HIV) infection[27]
Entecavir C12H15N5O3Hepatitis B virus[28]
TenofovirC9H14N5O4PHIV infection in adults and children[29]
Pyrimidine analogues
CidofovirC8H14N3O6PCytomegaloviral retinitis in people with acquired immunodeficiency syndrome (AIDS)[30]
TrifluridineC10H11F3N2O5Keratoconjunctivitis and recurrent epithelial keratitis due to herpes simplex virus, types 1 and 2[31]
Emtricitabine C8H10FN3O3SHuman immunodeficiency virus type 1 (HIV-1) (in combination with at least one other HIV drug)[32]
Lamivudine C8H11N3O3SHIV-1 infection, hepatitis B (HBV) virus infection[33]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

De Castro, F.; Stefàno, E.; De Luca, E.; Benedetti, M.; Fanizzi, F.P. Platinum-Nucleos(t)ide Compounds as Possible Antimetabolites for Antitumor/Antiviral Therapy: Properties and Perspectives. Pharmaceutics 2023, 15, 941. https://doi.org/10.3390/pharmaceutics15030941

AMA Style

De Castro F, Stefàno E, De Luca E, Benedetti M, Fanizzi FP. Platinum-Nucleos(t)ide Compounds as Possible Antimetabolites for Antitumor/Antiviral Therapy: Properties and Perspectives. Pharmaceutics. 2023; 15(3):941. https://doi.org/10.3390/pharmaceutics15030941

Chicago/Turabian Style

De Castro, Federica, Erika Stefàno, Erik De Luca, Michele Benedetti, and Francesco Paolo Fanizzi. 2023. "Platinum-Nucleos(t)ide Compounds as Possible Antimetabolites for Antitumor/Antiviral Therapy: Properties and Perspectives" Pharmaceutics 15, no. 3: 941. https://doi.org/10.3390/pharmaceutics15030941

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

Article Metrics

Back to TopTop