Next Article in Journal
Interfacial Bonding Improvement through Nickel Decoration on Carbon Nanotubes in Carbon Nanotubes/Cu Composite Foams Reinforced Copper Matrix Composites
Next Article in Special Issue
Carbene Addition Isomers of C70 formed in the Flame of Low-Pressure Combustion
Previous Article in Journal
Revealing the Real Role of Etching during Controlled Assembly of Nanocrystals Applied to Electrochemical Reduction of CO2
Previous Article in Special Issue
Highly Active Amino-Fullerene Derivative-Modified TiO2 for Enhancing Formaldehyde Degradation Efficiency under Solar-Light Irradiation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Progress in Antiviral Fullerene Research

1
College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
2
Funano New Material Technology Company Ltd., Xiamen 361110, China
3
Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen 361005, China
*
Author to whom correspondence should be addressed.
Nanomaterials 2022, 12(15), 2547; https://doi.org/10.3390/nano12152547
Submission received: 30 June 2022 / Revised: 20 July 2022 / Accepted: 22 July 2022 / Published: 24 July 2022
(This article belongs to the Special Issue Fullerene Nanostructures: Synthesis, Functionalities and Applications)

Abstract

:
Unlike traditional small molecule drugs, fullerene is an all-carbon nanomolecule with a spherical cage structure. Fullerene exhibits high levels of antiviral activity, inhibiting virus replication in vitro and in vivo. In this review, we systematically summarize the latest research regarding the different types of fullerenes investigated in antiviral studies. We discuss the unique structural advantage of fullerenes, present diverse modification strategies based on the addition of various functional groups, assess the effect of structural differences on antiviral activity, and describe the possible antiviral mechanism. Finally, we discuss the prospective development of fullerenes as antiviral drugs.

1. Introduction

Fullerenes are all-carbon molecules discovered in 1985 [1]. They are spherical or ellipsoidal in shape, with a hollow cage structure. Three discoverers of fullerene C60 won the Nobel Prize in chemistry in 1996. Fullerene C60, the representative fullerene, is ~0.7 nm in diameter. In the past 30 years, with the continuous development of fullerene preparation technology [2,3,4,5], fullerenes have presented unprecedented opportunities in the fields of biomedicine, catalysis, superconduction, and photovoltaics. Nanomolecules have important applications in cancer treatment, diagnosis, imaging, drug delivery, catalysis, and biosensing [6,7,8,9,10,11,12,13,14,15]. Fullerene molecules not only have defined nanostructures, but also unique electronic characteristics, photophysical properties, and excellent biocompatibility. Fullerene molecules have properties that differ from those of traditional small molecule drugs, which make fullerenes nanodrug candidates [16], especially for diagnosis and treatment. For example, fullerenes and their derivatives can be used as antioxidants against inflammatory diseases, due to their rich conjugated double bonds, which scavenge free radicals [17,18]. Fullerene C60 activates tumor immunity by polarizing tumor-associated macrophages and combines with immune checkpoint inhibitors (PD-L1 monoclonal antibody) to achieve efficient tumor immunotherapy [19]. Fullerene C70 derivatives, as photosensitizers, produce singlet oxygen, which can effectively kill tumor cells [20]. Endohedral metal fullerenes serve as new nuclear magnetic resonance contrast agents [21,22] for treating liver steatosis [23] and tumors [24,25,26,27,28,29,30]. Additionally, some fullerene derivatives stabilize immune effector cells and prevent/inhibit the release of proinflammatory mediators. Therefore, they are potential drugs for a variety of diseases, such as asthma [31], arthritis [32], and multiple sclerosis [33]. Moreover, carboxylic acid derivatives of fullerenes can cut DNA under visible light irradiation, with the potential for use as photosensitive biochemical probes [34]. Fullerene C60 carboxylic acid derivatives also exhibit neuroprotective activity [35] and strongly inhibit tumor growth in a zebrafish xenograft model [36]. Because nanoparticles have been approved as drugs and drug carriers, fullerenes have great potential as drugs or gene delivery carriers [37,38].
Currently, more than 90% of infectious diseases in humans are caused by viruses. The most well-known include the influenza virus, human immunodeficiency virus (HIV), and Ebola virus, which have caused serious damage [39,40,41,42,43]. Although several anti-HIV and anti-Ebola drugs, such as saquinavir, ritonavir, T20, lopinavir, ribavirin, tenofovir, and remdesivir, have been developed, their efficacy is not satisfactory. Severe acute respiratory syndrome (SARS), which broke out in China in 2003, is a respiratory infection caused by a coronavirus. So far, there is no specific medicine for SARS. The novel coronavirus, SARS-CoV-2, now circulating worldwide, is more infectious than SARS or HIV. For patients infected with SARS-CoV-2, there are no specific antiviral drugs.
Fullerenes and their derivatives exert significant inhibitory effects against HIV [39], herpes simplex virus (HSV) [40], influenza virus [44], Ebola virus [45], cytomegalovirus (CMV) [46], and other viruses in vitro and in vivo (Figure 1). Fullerenes and their derivatives, as a class of new, broad-spectrum antiviral drugs, have attracted increasing attention as a potential treatment for SARS-CoV-2.
Because fullerenes are insoluble in water and polar media, their use in biomedicine is extremely complicated [47]. To increase biocompatibility, the cage structure of fullerenes needs to be modified with appropriate hydrophilic functional groups. The modified structure and properties of the carbon cage may facilitate new applications in different biological systems. Because the fullerene carbon cage has multiple modifiable reaction sites, many fullerene derivatives with well-defined structures have been synthesized using regioselective functional group derivatization strategies. Therefore, fullerenes serve as ideal scaffolds for different bioactive drugs. Studies on the synthesis and antiviral activity of fullerenes and their derivatives have facilitated a deeper understanding of the relationship between fullerene structure and bioactivity.
In the past, hundreds of fullerene derivatives have been synthesized and used to inhibit viruses in vitro. Most of these derivatives are water soluble. These fullerene derivatives can be classified as the following six types: (1) amino acid, peptide, and primary amine derivatives; (2) piperazine and pyrrolidine derivatives; (3) carboxyl derivatives; (4) hydroxyl derivatives; (5) glycofullerene derivatives; and (6) fullerene complexes. Numerous antiviral studies have been conducted to evaluate fullerene C60 and its derivatives; this review assesses the latest research on the ability of fullerene C60 and its derivatives to inhibit virus replication.

2. Synthesis and Antiviral Properties of Amino Acid, Peptide, and Primary Amine Fullerene Derivatives

HIV-1-specific protease (HIVP) is an effective target for antiviral therapy. Its active site can be roughly described as an open-ended cylindrical cavity composed almost entirely of hydrophobic amino acids [48]. Since spherical C60 derivatives are hydrophobic and have a similar radius as the cylindrical cavity, strong hydrophobic interactions may occur between the active site surface and C60 derivatives. Therefore, C60 derivatives are potential inhibitors of HIVP.
In 1993, Friedman et al. [39] reported a landmark study based on model building and experimental evidence. Theoretical calculations revealed that the hydrophobic cavity of HIVP accommodates C60 molecules; the spherical C60 molecules fit perfectly within the active site, facilitating strong interactions between HIVP and fullerene. However, because C60 spheres are insoluble in polar solvents, it is critical to dissolve C60 in a medium suitable for biological testing. By modifying fullerene with strong polar groups, Sijbesma et al. [49] synthesized the water-soluble fullerene derivative bis (phenylenaminosuccinic acid)-C60 (1) (Figure 2). In vitro studies revealed that 1 inhibited acute and chronic HIV-1 infection in human peripheral blood mononuclear (PBM) cells, with a half-effective concentration (EC50) of 7.0 µM, while showing no cytotoxicity to uninfected PBM cells. This work is of great significance because fullerene derivatives as virus inhibitors are unprecedented in the field of antiviral research.
According to the theoretical analysis, the main driving force behind the binding between HIVP and fullerene derivatives is the hydrophobic interaction between the non-polar active site surface of the protease and the non-polar fullerene surface [50,51]. In order to improve antiviral activity, Friedman et al. proposed the addition of appropriate functional groups at specific positions on the fullerene derivatives [39] that would interact with the protease, generate electrostatic and/or hydrogen bonds as well as van der Waals forces, and subsequently increase the binding constant by several orders of magnitude. Therefore, compound 2 with two amino groups (Figure 3) was designed as an ideal “second generation” fullerene derivative. In addition to forming van der Waals forces with the non-polar HIVP surface, the cationic sites on the fullerene surface can form salt bridges with the catalytic aspartates on the floor of the active site, thus improving the binding strength. Although compound 2 was an ideal model of a fullerene derivative, the pure isomer was difficult to obtain, owing to a lack of regioselectivity. Subsequently, Prato and co-workers [52] proposed the design, simulation, and synthesis of the C60 diamine derivative 3, which was similar to 2. In the PM3-minimized structure of 3, the N–N distance between the two amino groups is 0.51 nm, while that in 2 is 0.55 nm, suggesting that the spatial arrangement of the two amino groups in 3 is very similar to that in 2.
In order to determine the binding energy between 3 and HIVP, Prato and co-workers [52] conducted a simulation using the Discover (Biosym/MSI) program with a cvff force field. Figure 4a depicts 3 binding to the cavity region of HIVP to form a complex. Compared with unmodified fullerene C60, the complex formed by the binding of 3 to HIVP exhibited significant improvements. When fullerene was protonated with a monoamino group, the relative binding energy was approximately −134 kJ/mol, and when it was protonated with a diamino group, the relative binding energy was approximately −105 kJ/mol. The greater binding energy of compound 3, with a single amino group, is due to the hydrogen bond interactions between the neutral –NH2 and –COOH groups of neutral aspartic acid, rather than the hydrogen bond interactions between the –NH3+ group and –COOH. Figure 4b illustrates the active site of the complex, highlighting the hydrogen bonds between the HIVP cavity and 3. The interatomic distance between the N atom in each amine/ammonium molecule and the O atom in the carboxyl/carboxylic acid is about 0.28 nm. This strong interaction suggests the contribution of Coulombic attraction.
In the same time period, many research groups synthesized various fullerene derivatives with different functional groups and demonstrated effective virus inhibition by introducing appropriate carboxylic acid and amino acid groups at specific positions on fullerenes. Among them, the fullerene dendritic amino acid derivative 4, prepared by Brettreich and Hirsch [53] (Figure 5), is highly water soluble and has an EC50 of 0.22 µM in human lymphocytes acutely infected with HIV-1 [54]. This compound also shows no toxicity up to 100 μM in PBM, Vero, and CEM cells. The fullerene amino acid derivative 5 (Figure 5), synthesized by Mashino et al. [40], strongly inhibits HIV reverse transcriptase (HIV-RT), with an EC50 of 0.029 μM. Additionally, Toniolo et al. [55] synthesized fullerene peptide derivatives, which also exhibit anti-HIVP activity.
In 2012, Troshin and coworkers [43] reported that multi-functional C60 amine and amino acid derivatives could be readily prepared from hexachlorofullerene C60Cl6 (6). The synthesized fullerene derivative contains at least five hydrophilic functional groups (such as amino or carboxyl groups). As shown in Figure 6, a fullerene amino acid ester (8) can be obtained via the reaction of C60Cl6 with an amino acid ester (7). The amino acid ester groups in compound 8 can be hydrolyzed to obtain fullerene amino acid derivatives under acidic conditions. In order to further increase the water solubility of fullerene amino acid derivatives, fullerene amino acid potassium salts are formed by adding potassium carbonate.
In vitro cell experiments [43] showed that the carboxylic acid potassium salt 8 has low cytotoxicity to HSV-sensitive Vero cells (CC50 > 1.3 mM) and human CMV-sensitive human embryonic lung fibroblasts (CC50 > 0.5 mM). Meanwhile, compound 8 showed pronounced antiviral activity, with an EC50 of 0.26 µM for HSV and 37.6 µM for CMV. Combining CC50 and EC50 values, the selectivity indices of compound 8 for HSV and CMV are >5000 and 14, respectively, indicating that compound 8 has potential as a new antiviral drug against HSV and CMV.

3. Synthesis and Antiviral Studies of Fullerene Piperazine and Pyrrolidine Derivatives

As shown in Figure 7, the reaction of C60Cl6 with N-methylpiperazine (9) can efficiently generate a fullerene-piperazine derivative (10) in the absence of any base [43]. By adding six times the amount of compound 9 to a C60Cl6 toluene solution, 10 is precipitated immediately, with more than a 95% yield. In vivo cell experiments have indicated that 10 exhibits high acute toxicity when administered via intraperitoneal injection in mice, while 8 shows very low acute toxicity, suggesting the latter is safe for biomedical applications. This example also revealed that the toxicity of the water-soluble fullerene derivatives depends largely on the organic functional groups attached to the fullerene carbon cage.
According to the literature [51], a regioisomer mixture of C60 pyrrolidine derivatives exhibits good anti-HIV-1 activity. The pyrrolidine derivative has a low EC50 and exhibits little toxicity to Vero and PBM cells. However, a relationship between the structure and activity of the reported isomer mixture of fullerene pyrrolidine derivatives has not been established. In order to better understand which structural characteristics can be modified to improve anti-HIV activity, Prato and coworkers [56] synthesized a series of new, pure fullerene pyrrolidine derivatives through the Prato reaction (Figure 8).
The Bis-adduct fullerene pyrrolidine derivative has eight regioisomers. The yields of the eight isomers differ; cis-Bis-adducts are produced at extremely low yields and trans- and equatorial-adducts are produced at relatively high yields. Hence, Prato and coworkers were able to separate trans- and equatorial-isomers (1215 and 1619, respectively) and study their structure and activity. Additionally, a Mono-adduct (11) and other Bis-adducts (2021, 2223) were synthesized (Figure 8).
The anti-HIV activity and cytotoxicity of all fullerene pyrrolidine derivatives (1123) was tested in lymphocyte (CEM) cultures infected with HIV-1 or HIV-2. As shown in Table 1, mono-functionalized derivative 11 and Bis-adduct derivatives 1215, 20, 21 and 23 showed low anti-HIV-1 activity, while the corresponding quaternary ammonium pyrrolidine derivatives 1619 showed high anti-HIV-1 activity at low concentrations (EC50: 0.40–2.60 µM). These results suggest that the inhibitory effect on HIV-1 might be related to electrostatic interactions.
The biological activity of pyrrolidine derivatives of fullerene varies among the regioisomers. As shown in Table 1, the anti-HIV-1 activity of the trans-2 isomer (16) was 2.4~6.5 times that of the corresponding trans-3 (17), trans-4 (18), and equatorial (19) isomers. The trans-2-tetraacetic acid derivative (22) showed high anti-HIV-1 activity at low concentrations, while the equatorial isomer (23) showed low activity. The introduction of malonate resulted in the loss of anti-HIV-1 activity of pyrrolidine derivatives 2021. Most synthetic fullerene pyrrolidine derivatives exhibit pronounced toxicity in human CEM cells (CC50: 3.02~13.2 µM), but monofunctional derivative 11 exhibit relatively low toxicity (CC50: 44 µM). This toxicity most likely results from the strong amphiphilic properties of these pyrrolidine derivatives, which can cause the rupture of the cell membrane and subsequent cell death.
To define the structure-activity relationship, Prato and co-workers [57] prepared Bis-adduct fullerene pyrrolidine derivatives (2428) through a [3+2] dipolar cycloaddition reaction between azomethine ylides and C60 (Figure 9) and subsequently studied their anti-HIV-1 and anti-HIV-2 activity and cytotoxicity in CEM culture. As shown in Table 2, the anti-HIV-1 activity of the trans-isomers (2426) is about 2–10 times that of the corresponding cis-3 isomer (28), while the equatorial-isomer (27) exhibits no antiviral activity against HIV-1. Among the trans-isomers, trans-2 (24) and trans-3 (25) demonstrate higher levels of anti-HIV-1 (EC50 = 0.21 and 0.35 µM, respectively) and anti-HIV-2 activity (EC50 = 0.21–1.0 µM). The anti-HIV-1 activity of the trans-4 isomer (26) is significantly lower than that of compounds 24 and 25 (EC50 = 1.08 µM), and the anti-HIV-2 activity is also lower (EC50 = 2.5 µM), which is consistent with the experimental results for fullerene pyrrolidine derivatives 1123. Fullerene pyrrolidine derivatives 2428 also exhibit toxicity in human CEM cells (CC50 = 2.9–28.7 µM). The CC50/EC50 ratio of the trans-3 isomer (25) is 26, higher than that of the reference compound 16 (CC50/EC50 = 12).
As mentioned, Mashino et al. (2005) reported that fullerene amino acid-type derivatives exhibited HIV-RT inhibitory activity, while cationic fullerene derivatives such as the pyrrolidinium-type derivatives showed weaker HIV-RT inhibitory activity [40]. The carboxyl groups on the pyrrolidine-type fullerene derivatives were considered crucial to HIV-RT inhibitory activity [40,58]. However, ten years later, Mashino et al. found that fullerene pyrrolidine-pyridine and pyrrolidine-pyridinium salt derivatives without any carboxyl groups [42], such as 2940, which are functionalized with pyridine or pyridinium groups (Figure 10), exhibit strong HIV-RT inhibition. This is useful information for the future design of fullerene derivatives as HIV-RT inhibitors. In addition to HIV-RT inhibition, recently, Kobayashi et al. [59] found that compound 34 can inhibit HIV-PR, and HCV NS5B polymerase (HCV NS5B) with IC50 values in the micromolar range. Compound 41, the exo-substituent on the most potent derivative (34), exhibits no HIV-RT inhibitory activity in cell culture, indicating that HIV-RT inhibition is dependent on the fullerene skeleton. The conventional trypan blue dye exclusion test (Table 3) was used to evaluate the cytotoxicity of all fullerene pyrrolidine-pyridine and pyrrolidine-pyridinium salt derivatives (2940) to HL60 cells. The CC50 of all derivatives except 35 (CC50 = 39.4 µM) was greater than 50 µM, which suggests that the new fullerene-pyrrolidine-pyridine or pyridinium salt derivatives effectively inhibit HIV-RT activity without damaging living cells.
In 2016, Echegoyen and co-workers [41] reported a novel cationic N,N-dimethyl C70 fullerene-pyrrolidine iodized salt derivative (4244), with fullerene C70 as the starting material (Figure 11), that inhibits more than 99% of HIV-1 infectivity at a low micromolar concentration. These three compounds have an EC50 of 0.41, 0.33, and 0.54 µM, respectively. An analysis of the life cycle of HIV-1 suggested that these compounds inhibit viral maturation by influencing the processing of Gag and GAG-POL. Significantly, fullerene derivatives 4244 do not inhibit protease activity in vitro, and strongly interact with immature HIV capsid proteins in a pull-down assay. Moreover, these compounds may block infection by viruses carrying either a mutant protease that is resistant to multiple protease inhibitors, or the mutant Gag protein, which is resistant to the mature inhibitor bevirimat. Fullerenes 4244 do not inhibit HIV-1 proteases at doses that strongly inhibit HIV-1 infection in vitro, suggesting that this mechanism is independent of the HIV-1 protease. This finding differed from previous reports that fullerene derivatives affect HIV-1 protease activity in vitro. Echegoyen et al. [60] then proposed that fullerene derivatives 4244 act through a novel anti-HIV-1 mechanism, rather than interacting with other capsid proteins as previously reported. Unraveling the details of this mechanism will facilitate the discovery of novel anti-HIV-1 inhibitors.
Additionally, Tollas et al. [44] reported that fullerene pyrrolidine derivatives functionalized with hydrophilic sugar groups. While these derivatives have no inhibitory effect on influenza virus hemagglutinin, they exhibit a good inhibitory effect on neuraminidase.

4. Synthesis and Antiviral Studies of Fullerene Carboxyl Derivatives

As mentioned, hydrophilic functional groups such as amino or amino acid groups can be directly or indirectly attached to the skeleton of fullerenes to increase water solubility. These water-soluble fullerene derivatives have exhibited high levels of antiviral activity. Instead of amino or amino acid groups, a single carboxylic acid group can be used to modify fullerenes to increase their water solubility and improve their antiviral activity. In 1996, Schuster et al. [51] synthesized 11 new water-soluble fullerene derivatives, among which fullerene carboxylic acid derivatives 45 and 46 (Figure 12) exhibit antiviral activity against HIV-1 at low micromolar concentrations, with an IC50 of 2.2 and 6.3 μM, respectively.
In 2007, Troshin and coworkers [61] proposed an effective method for the synthesis of water-soluble fullerene carboxylic acid derivatives. With C60Cl6 as the starting point, C60(Ar)5Cl, with ester groups linked to aryl groups, was obtained via the simple and efficient Friedel–Crafts arylation of C60Cl6 (6) with methyl esters of phenylacetate at 100 °C. The fullerene carboxylic acid derivative (48) was prepared in almost a quantitative yield by removing the methyl group from the methyl ester under acidic conditions, as shown in Figure 13. Compound 48, with five carboxyl groups, is insoluble in water but soluble in DMSO. In order to improve the water solubility, potassium carbonate was added to neutralize the carboxylic acid group of 48 and form the corresponding ionic potassium salts, with a water solubility of up to 50–100 mg/mL at pH < 7.5. A virus-induced cytopathicity assay revealed that the fullerene carboxylic acid potassium derivative has pronounced anti-HIV-1 activity, with an IC50 of 1.20 ± 0.44 μM and a low cytotoxicity (>52 μM).
Usually, only five chlorine atoms in 6 are replaced; the sixth chlorine atom cannot be substituted due to steric hindrance. Recently, Troshin and coworkers [62] found that the sixth chlorine atom can be substituted by an alkyl group through a reverse Arbuzov reaction between trialkyl phosphite (P(OR)3) and the fullerene derivative C60(Ar)5Cl (Figure 14). More significantly, the introduction of different R groups through the reverse Arbuzov reaction affects the antiviral activity of the carboxyl fullerene derivatives, establishing a fundamental correlation between the structure of carboxyl fullerene derivatives and their antiviral activity. Experiments on the inhibition of the influenza H3N2 virus showed that compounds 49 and 50 (R = Et and Me, respectively) were quite active, while the fullerene derivative 48 (R = Cl) was completely inactive. Specifically, 49 and 50 effectively inhibited influenza virus H3N2 at nanomolar concentrations, with an EC50 of 500 nM and 100 nM, respectively; both compounds were more active against H3N2 than the commercial drugs zanamivir (EC50 = 3.0 μM) and amantadine (EC50 = 1.3 μM). However, there were no significant differences in the HIV-1 inhibitory activity of the fullerene derivative C60(Ar)5Cl after the alkyl substitution of chlorine atoms.
Recently, Troshin and coworkers [63] synthesized the fullerene carboxylic acid derivative C70[p-C6H4(CH2)nCOOH]8 by Friedel–Crafts arylation of chlorofullerene C70Cl8 with unprotected carboxylic acids. The obtained carboxylic acid fullerene C70 derivatives showed significant antiviral effects against HIV and the influenza viruses H1N1 and H3N2. The EC50 value of anti-HIV-1/NL4.3 (X4) is close to 1.0 µM, which indicates that this derivative is more effective against the virus than the commercial drug tenofovir. Additionally, Troshin and coworkers synthesized a variety of carboxylic acid fullerene derivatives, such as the carboxylic acid thiofullerene derivative, 51 [64]; polycarboxylic acid fullerene derivative, 52 [65]; and tetracarboxylic acid methanofullerene derivative, 53 (Figure 15) [66]. All these compounds exhibit inhibitory effects against HIV-1, HIV-2, influenza A (H3N2), HSV, and CMV, with low toxicity.
Modern antiviral drugs have extended patients’ lives and improved their quality of life, but unsolved problems remain, such as toxicity, limited bioavailability, and drug resistance (rapid cyclic changes in influenza strains reduce the effectiveness of commonly used vaccines). Therefore, finding new antiviral drugs has become an urgent problem. The research by Troshin and coworkers on carboxylic acid fullerene derivatives has provided more possibilities for the development of new and effective antiviral drugs [61,62,63,64,65,66].

5. Synthesis and Antiviral Studies of Fullerene Hydroxyl Derivatives

Fullerenol is a polyhydroxylated fullerene C60 derivative with suitable water solubility and biocompatibility. In 2013, Eropkin et al. [67] synthesized a series of fullerenols (Figure 16). Three different groups of fullerenols, C60(OH)12–14, C60(OH)18–24, and C60(OH)30–38, can be prepared by changing the reaction conditions to control the number of hydroxyl groups attached to the fullerene skeleton. Experimental studies revealed that fullerenols containing 12~14 hydroxyl groups are insoluble in water and have no biological activity when introduced into cell culture as suspensions. The other two groups of fullerenols show broad spectrum antiviral activity in vitro against the human influenza viruses H1N1 and H3N2, avian influenza virus A (H5N1), adenovirus, human HSV, and respiratory syncytial virus. C60(OH)18–24 demonstrates better antiviral activity than C60(OH)30–38. Moreover, the three water-soluble fullerenols exhibit no toxicity in vitro to human and animal cells.
Compared with other fullerene derivatives, fullerenols are relatively easy to prepare, requiring only one organic reaction step. So far, only one example of polyhydroxyfullerene (C60(OH)8) with a well-defined structure has been reported, by Gan and co-workers [68]. However, the insufficient number of hydroxyl groups in C60(OH)8 limits its biological applications. At present, even by purification via high performance liquid chromatography, it is impossible to obtain pure regioisomer fullerenols from the mixture of polyhydroxyfullerenes. Therefore, the fullerenols used in current biological studies have been a mixture of regioisomers, which restricts the potential of fullerenols as standard drug candidate molecules.

6. Synthesis and Antiviral Studies of Glycofullerene Derivatives

Carbohydrate and protein interactions dominate many biological processes, including inflammation, embryogenesis, tumor development and metastasis, and pathogen infection [69]. These interactions are characterized by high selectivity, metal ion dependence, and compensation for low affinity through multivalent interactions [70]. Finding a suitable system to realize the polyvalent expression of sugars has been a subject of extensive research [71,72]. Due to the lack of information on the proper orientation of the ligands required to obtain the strongest interactions, chemists have experimented with all possible scaffolds. Calixarene [73], gold nanoparticles [74], polymers [75], liposomes [76], dendritic macromolecules [77], and fullerenes [78] are most commonly used as scaffolds to construct multivalent glycoconjugates. The advantages associated with fullerenes over other nanostructures are their three-dimensional (3D) structure and the ability to functionalize different positions of the C60 cage in a controlled way [79]. In this sense, fullerenes can be thought of as a special class of spherical scaffolds ideal for building a multivalent spherical ligand. Martin and coworkers [78] proposed that spherical carbohydrate derivatives of fullerenes, with fullerenes used as scaffolds, could serve as an interesting sugar analog. The rigid spherical scaffold allows distance to be maintained between the two ligands (the diameter of fullerenes is 1 nm, plus the distance provided by the dendritic moiety), thus increasing the chances of obtaining effective multivalent interactions. In addition, due to the symmetry of the multivalent system, the 3D orientation of these ligands at 360° better mimics the surface of a pathogen, such as HIV; thus, the molecule is more likely to encounter a receptor.
Based on an octahedral addition pattern, soluble hexakis-adduct glycofullerene derivatives can act as spherical carbohydrates, thus serving as a potential multivalent spherical ligand. In 2013, Martin and coworkers [79] designed and synthesized a class of hexakis-adduct glycofullerene derivatives (54), containing 36 mannoses (Figure 17), and used them to inhibit cell infection by pseudotyped Ebola virus particles. This was the first time that glycofullerene derivatives were demonstrated to effectively inhibit cell infection. In the pseudotyped Ebola infection model, the antiviral activity of 54 was in the low nanomolar range, with an IC50 of 0.3 μM. The glycofullerene with 12 galactosyl had no inhibitory effect on virus infection, indicating that the inhibitory effect is dependent on mannose. Interestingly, only an increase in the valence of glycofullerene resulted in a loss of the antiviral effect. This phenomenon is related to the spatial crowding of sugars at the surface of glycofullerene. Martin et al. speculated that the high binding affinity occurs not only because of the extensive spatial presentation of multivalent ligands, but also the frequent interactions between the ligands and corresponding receptors. They demonstrated that a rational design of compounds with the same valency but longer spacers can significantly increase the antiviral activity, likely due to more efficient interactions with receptors. Therefore, the selection of suitable scaffolds (such as spherical fullerenes) to achieve multivalence, as well as the accessibility and flexibility of ligands, are key factors for improving antiviral activity.
The aforementioned studies have demonstrated that using multivalent glycofullerenes to block lectin receptors on the cell surface is a promising method for inhibiting virus entry into cells. However, creating large enough multivalent glycofullerenes to improve the binding ability between ligands and virus receptors remains a challenge. In 2015, Martin and coworkers [45] conducted an impressive study of water-soluble fullerenes. They synthesized three water-soluble glycofullerene derivatives (5658) with 12 fullerene spheres modified with 120 sugars, starting from the hex-adduct fullerene derivative (55), namely, the “super sphere” (Figure 18). In addition to the core fullerene sphere, the other 12 fullerenes in the super sphere contain 10 sugars each, totaling 120 sugars, and have diameters up to 4 nm.
The mannose in compounds 5658 is critical in blocking virus entry into cells. As expected, 57, with 120 galactosyl species, did not inhibit the infection process. Glycofullerene derivatives 56 and 58, with 120 mannoses, exhibited high levels of antiviral activity in the pM~nM range. As shown in Table 4, 56 can effectively block Ebola virus infection in the nM range, with an IC50 of 20.4 nM. Compound 58 is almost 10 times more potent in the antiviral infection process, with an IC50 of 667 pM. Fullerene 54, linked with 36 mannoses, produced relative inhibitory effect (RIP) values of at least two orders of magnitude smaller than those of 56 and 58. Compared to previously reported results, 56 and 58 are the most effective compounds against Ebola virus infection in vitro. Compounds 5658 showed no appreciable cell cytotoxicity at the concentrations used in the virus inhibition experiments. Owing to their biocompatibility and spherical structure, fullerenes have become ideal scaffolds for studying multivalent interactions. However, further research is needed to determine whether the obtained glycofullerene derivatives can be used for practical applications.
In order to enhance multivalency and improve the biocompatibility of glycofullerene derivatives, Martin and coworkers [80] (2019) synthesized tridecafullerene derivatives containing up to 360 1,2-mannobiosides via the strain-promoted azide–alkyne cycloaddition method. The obtained glycofullerene derivative showed pronounced antiviral activity against Zika virus and dengue virus, with an IC50 of 67 and 35 pM, respectively.
Additionally, Martin and coworkers [81] synthesized a series of amphiphilic glycodendrofullerene [60] monoadducts (59 and 60) through the “click chemistry” reaction. In aqueous media, the glycodendrofullerenes can self-assemble into large, compact micelles with a uniform size and spherical shape. Antiviral tests showed that these aggregates of 59 and 60 can effectively inhibit Ebola virus infection in the nM range, with an IC50 of 424 nM and 196 nM, respectively. However, these compounds are inferior to 56 and 68.
Ebola virus has a filament-like structure and is similar to single-walled carbon nanotubes (SWCNTs) and multi-walled carbon nanotubes (MWCNTs) in shape. SWCNTs are usually 1.5 to 3.0 nm in diameter. They match Ebola virus in length, at 20 to 1000 nm. On the other hand, HIV has a roughly spherical shape of about 120 nm in diameter and is very similar in shape and size to spherical fullerenes and single-walled nanocones (SWCNHs) [82]. Therefore, Martin and coworkers proposed that SWCNTs, MWCNTS, and SWCNHs could be used as virus-mimicking nanocarbon platforms, and after chemical modification, could interact with the receptors in a multivalent manner. To employ the novel strategy for designing antiviral agents against HIV or Ebola virus (designing glycofullerene and nanocarbon complexes that mimic the virus surface and interfere with the infection of receptors on the corresponding cell surface), Martin et al. [83] (2018) used the “click chemistry” reaction to covalently connect glycofullerene to SWCNTs, MWCNTs, or SWCNHs. The multivalent hybrid glycoconjugate consisting of MWCNTs and glycofullerene (61) is shown in Figure 19. After chemical modification, the water solubility of the MWCNTs was significantly improved. In tests of efficiency in blocking artificial Ebola virus infection with three types of multivalent hybrid glycoconjugates, 61, based on MWCNTs and fullerene functionalization, was the most effective inhibitor of viral infection, with an IC50 of 0.37 µg/mL.
Cyclodextrins (CDs) are a series of cyclic oligosaccharides containing 6–12 D-glucopyranose units. They have a slightly conical, hollow, cylindrical three-dimensional ring structure and have been widely studied because of their significant solubility in water. The CD molecules containing 6, 7, and 8 glucose units are called α-, β- and γ-CD, respectively. In 2012, Zhang and coworkers [84] prepared a water-soluble α-CD-C60 conjugate (62), in which C60 and α-CD were linked by two flexible alkyl chains (Figure 20). Interestingly, this water-soluble compound showed pronounced anti-HCV entry activity, with an IC50 of 0.17 µM.
In 2018, Zhang and coworkers [85] continued to design and synthesize seven α-CD-C60 copolymers and two γ-CD-C60 copolymers [85]. However, the newly obtained α-CD-C60 conjugates did not exhibit inhibitory activity against HCV. Subsequently, all nine CD-C60 conjugates were assessed in terms of their activity against the influenza virus H1N1. No conjugates exhibited cytotoxicity at 100.0 µM. The two γ-Cd-C60 conjugates demonstrated higher anti-H1N1 activity, with IC50 values of 87.7 µM and 75.0 µM, respectively. Because they exhibit less aggregation in aqueous solutions, the two γ-Cd-C60 conjugates are the most water soluble of the nine conjugates. This trait might be related to their higher inhibitory efficiency against H1N1.

7. Synthesis and Antiviral Studies of Fullerene Complexes

As mentioned, to enhance the water solubility of pristine fullerenes, various hydrophilic functional groups (amino acids, sugars, or calixarene) can be directly or indirectly used to modify fullerenes through covalent bonds. Alternatively, pristine fullerenes can be dispersed in polyvinylpyrrolidone (PVP), Triton X-100, dioctadecyldimethylammonium bromide, or lecithin to form complexes.
Sirotkin et al. [86] added an aqueous solution of the C60/PVP complex to a suspension of H1N1 particles. As a result, the number of virus particles with a broken lipoprotein envelope increased dramatically, possibly due to the fusion of the C60/PVP complex with the influenza virus.
Yang and coworkers [87] prepared a fullerene [60] liposome complex (Figure 21) and studied its anti-H1N1 activity in vivo. The fullerene liposome complex significantly reduces the average lung virus yields and lung index; prolongs the mean time to death; and decreases the mortality of mice infected with H1N1. In addition, the fullerene liposome complex has good water solubility and low toxicity, and its anti-influenza activity in vivo is much higher than that of rimantadine. Therefore, the fullerene [60] liposome complex is a promising clinical candidate drug against influenza infection.

8. Conclusions

This review summarized the latest antiviral research conducted on fullerenes and their derivatives. Numerous water-soluble fullerene derivatives or fullerene complexes have shown great antiviral potential, mainly because fullerenes have three advantages. First, pristine fullerenes are hydrophobic, which is conducive to the formation of strong hydrophobic interactions with the active site surfaces of viruses. Second, hydrophilic groups with various functions (such as amino, carboxyl, amino acid, hydroxyl, pyrrolidine, and sugar groups) can be used to selectively modify the unique spherical skeleton of fullerenes via organic reactions. Third, fullerenes and their derivatives exhibit no or low cytotoxicity at relatively high concentrations. Although fullerenes are promising prospective antiviral drugs, antiviral research on fullerenes requires improvement. Most fullerene derivatives exhibit good antiviral effects in vitro, but the antiviral mechanism has not been thoroughly studied. Additionally, most of the studies on fullerenes have only focused on virus inhibition in vitro; there have been few antiviral studies in vivo, and relevant clinical studies involving fullerenes have not been conducted.
Viruses constantly threaten human health. Fullerenes have become an important molecular platform for the development of antiviral drugs. Research on fullerenes as antiviral drugs urgently needs the joint efforts of scientists working in synthesis, molecular design, biology, and medicine. Some fullerene derivatives display inhibitory activity against multiple types of viruses. Therefore, fullerene derivatives have the potential to become a class of broad-spectrum antiviral drugs effective against SARS-CoV-2, which remains a global threat. We believe that this review will encourage more researchers to synthesize fullerene derivatives and study their antiviral properties and applications.

Funding

This research was funded by the National Natural Science Foundation of China (NSFC) (91961113, 21721001, 92061204, 21827801 and 92061000); Xiamen Youth Innovation Fund (3502Z20206054); the Funds of the Science and Technology Project of Yunnan Province-Major Project (202101AS070049).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kroto, H.W.; Heath, J.R.; O’Brien, S.C.; Curl, R.F.; Smalley, R.E. C60: Buckminsterfullerene. Nature 1985, 318, 162–163. [Google Scholar] [CrossRef]
  2. Krätschmer, W.; Lamb, L.D.; Fostiropoulos, K.; Huffman, D.R. Solid C60: A new form of carbon. Nature 1990, 347, 354–358. [Google Scholar] [CrossRef]
  3. Howard, J.B.; McKinnon, J.T.; Makarovsky, Y.; Lafleur, A.L.; Johnson, M.E. Fullerenes C60 and C70 in flames. Nature 1991, 352, 139–141. [Google Scholar] [CrossRef]
  4. Tian, H.-R.; Chen, M.-M.; Wang, K.; Chen, Z.-C.; Fu, C.-Y.; Zhang, Q.; Li, S.-H.; Deng, S.-L.; Yao, Y.-R.; Xie, S.-Y.; et al. An Unconventional Hydrofullerene C66H4 with Symmetric Heptagons Retrieved in Low-Pressure Combustion. J. Am. Chem. Soc. 2019, 141, 6651–6657. [Google Scholar] [CrossRef] [PubMed]
  5. Zhang, H.-G.; Zhuo, Y.-Q.; Zhang, X.-M.; Zhang, L.; Xu, P.-Y.; Tian, H.-R.; Lin, S.-C.; Zhang, Q.; Xie, S.-Y.; Zheng, L.-S. Synthesis of Fullerenes from a Nonaromatic Chloroform through a Newly Developed Ultrahigh-Temperature Flash Vacuum Pyrolysis Apparatus. Nanomaterials 2021, 11, 3033. [Google Scholar] [CrossRef] [PubMed]
  6. Yang, B.; Chen, Y.; Shi, J. Reactive Oxygen Species (ROS)-Based Nanomedicine. Chem. Rev. 2019, 119, 4881–4985. [Google Scholar] [CrossRef]
  7. Rosi, N.L.; Mirkin, C.A. Nanostructures in Biodiagnostics. Chem. Rev. 2005, 105, 1547–1562. [Google Scholar] [CrossRef]
  8. Ni, K.; Luo, T.; Nash, G.T.; Lin, W. Nanoscale Metal–Organic Frameworks for Cancer Immunotherapy. Acc. Chem. Res. 2020, 53, 1739–1748. [Google Scholar] [CrossRef]
  9. Kirtane, A.R.; Verma, M.; Karandikar, P.; Furin, J.; Langer, R.; Traverso, G. Nanotechnology approaches for global infectious diseases. Nat. Nanotechnol. 2021, 16, 369–384. [Google Scholar] [CrossRef]
  10. Li, S.; Jiang, Q.; Liu, S.; Zhang, Y.; Tian, Y.; Song, C.; Wang, J.; Zou, Y.; Anderson, G.J.; Han, J.-Y.; et al. A DNA nanorobot functions as a cancer therapeutic in response to a molecular trigger in vivo. Nat. Biotechnol. 2018, 36, 258–264. [Google Scholar] [CrossRef]
  11. Ji, T.; Zhao, Y.; Ding, Y.; Wang, J.; Zhao, R.; Lang, J.; Qin, H.; Liu, X.; Shi, J.; Tao, N.; et al. Transformable Peptide Nanocarriers for Expeditious Drug Release and Effective Cancer Therapy via Cancer-Associated Fibroblast Activation. Angew. Chem. Int. Ed. 2016, 55, 1050–1055. [Google Scholar] [CrossRef] [PubMed]
  12. Han, X.; Li, Y.; Xu, Y.; Zhao, X.; Zhang, Y.; Yang, X.; Wang, Y.; Zhao, R.; Anderson, G.J.; Zhao, Y.; et al. Reversal of pancreatic desmoplasia by re-educating stellate cells with a tumour microenvironment-activated nanosystem. Nat. Commun. 2018, 9, 3390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Lu, F.; Gu, L.; Meziani, M.J.; Wang, X.; Luo, P.G.; Veca, L.M.; Cao, L.; Sun, Y.-P. Advances in Bioapplications of Carbon Nanotubes. Adv. Mater. 2009, 21, 139–152. [Google Scholar] [CrossRef]
  14. Feng, L.; Zhang, S.; Liu, Z. Graphene based gene transfection. Nanoscale 2011, 3, 1252–1257. [Google Scholar] [CrossRef] [PubMed]
  15. Zhu, M.; Nie, G.; Meng, H.; Xia, T.; Nel, A.; Zhao, Y. Physicochemical Properties Determine Nanomaterial Cellular Uptake, Transport, and Fate. Acc. Chem. Res. 2013, 46, 622–631. [Google Scholar] [CrossRef] [Green Version]
  16. Dellinger, A.; Zhou, Z.; Connor, J.; Madhankumar, A.; Pamujula, S.; Sayes, C.M.; Kepley, C.L. Application of fullerenes in nanomedicine: An update. Nanomedicine 2013, 8, 1191–1208. [Google Scholar] [CrossRef] [Green Version]
  17. McEwen, C.N.; McKay, R.G.; Larsen, B.S. C60 as a radical sponge. J. Am. Chem. Soc. 1992, 114, 4412–4414. [Google Scholar] [CrossRef]
  18. Maas, M. Carbon Nanomaterials as Antibacterial Colloids. Materials 2016, 9, 617. [Google Scholar] [CrossRef] [Green Version]
  19. Li, L.; Zhen, M.; Wang, H.; Sun, Z.; Jia, W.; Zhao, Z.; Zhou, C.; Liu, S.; Wang, C.; Bai, C. Functional Gadofullerene Nanoparticles Trigger Robust Cancer Immunotherapy Based on Rebuilding an Immunosuppressive Tumor Microenvironment. Nano Lett. 2020, 20, 4487–4496. [Google Scholar] [CrossRef]
  20. Liu, Q.; Guan, M.; Xu, L.; Shu, C.; Jin, C.; Zheng, J.; Fang, X.; Yang, Y.; Wang, C. Structural Effect and Mechanism of C70-Carboxyfullerenes as Efficient Sensitizers against Cancer Cells. Small 2012, 8, 2070–2077. [Google Scholar] [CrossRef]
  21. MacFarland, D.K.; Walker, K.L.; Lenk, R.P.; Wilson, S.R.; Kumar, K.; Kepley, C.L.; Garbow, J.R. Hydrochalarones: A Novel Endohedral Metallofullerene Platform for Enhancing Magnetic Resonance Imaging Contrast. J. Med. Chem. 2008, 51, 3681–3683. [Google Scholar] [CrossRef] [Green Version]
  22. Zheng, J.-p.; Liu, Q.-l.; Zhen, M.-m.; Jiang, F.; Shu, C.-y.; Jin, C.; Yang, Y.; Alhadlaq, H.A.; Wang, C.-r. Multifunctional imaging probe based on gadofulleride nanoplatform. Nanoscale 2012, 4, 3669–3672. [Google Scholar] [CrossRef] [PubMed]
  23. Zhou, C.; Zhen, M.; Yu, M.; Li, X.; Yu, T.; Liu, J.; Jia, W.; Liu, S.; Li, L.; Li, J.; et al. Gadofullerene inhibits the degradation of apolipoprotein B100 and boosts triglyceride transport for reversing hepatic steatosis. Sci. Adv. 2020, 6, eabc1586. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, Y.; Chen, C.; Qian, P.; Zhu, T.; Zhao, Y. Gd-metallofullerenol nanomaterial as non-toxic breast cancer stem cell-specific inhibitor. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 510. [Google Scholar] [CrossRef] [Green Version]
  25. Kang, S.-G.; Zhou, G.; Yang, P.; Liu, Y.; Sun, B.; Huynh, T.; Meng, H.; Zhao, L.; Xing, G.; Chen, C.; et al. Molecular mechanism of pancreatic tumor metastasis inhibition by Gd@C82(OH)22 and its implication for de novo design of nanomedicine. Proc. Natl. Acad. Sci. USA 2012, 109, 15431–15436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Deng, R.; Wang, Y.; Zhen, M.; Li, X.; Zou, T.; Li, J.; Yu, T.; Zhou, Y.; Lu, Z.; Xu, H.; et al. Real-time monitoring of tumor vascular disruption induced by radiofrequency assisted gadofullerene. Sci. China Mater. 2018, 8, 1101. [Google Scholar] [CrossRef] [Green Version]
  27. Zhen, M.; Shu, C.; Li, J.; Zhang, G.; Wang, T.; Luo, Y.; Zou, T.; Deng, R.; Fang, F.; Lei, H.; et al. A highly efficient and tumor vascular-targeting therapeutic technique with size-expansible gadofullerene nanocrystals. Sci. China Mater. 2015, 58, 799–810. [Google Scholar] [CrossRef] [Green Version]
  28. Zhou, Y.; Deng, R.; Zhen, M.; Li, J.; Guan, M.; Jia, W.; Li, X.; Zhang, Y.; Yu, T.; Zou, T.; et al. Amino acid functionalized gadofullerene nanoparticles with superior antitumor activity via destruction of tumor vasculature in vivo. Biomaterials 2017, 133, 107–118. [Google Scholar] [CrossRef]
  29. Zhou, Y.; Li, J.; Ma, H.; Zhen, M.; Guo, J.; Wang, L.; Jiang, L.; Shu, C.; Wang, C. Biocompatible [60]/[70] Fullerenols: Potent Defense against Oxidative Injury Induced by Reduplicative Chemotherapy. ACS Appl. Mater. Interfaces 2017, 9, 35539–35547. [Google Scholar] [CrossRef]
  30. Zhou, Y.; Zhen, M.; Ma, H.; Li, J.; Shu, C.; Wang, C. Inhalable gadofullerenol/[70] fullerenol as high-efficiency ROS scavengers for pulmonary fibrosis therapy. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 1361–1369. [Google Scholar] [CrossRef]
  31. Norton, S.K.; Wijesinghe, D.S.; Dellinger, A.; Sturgill, J.; Zhou, Z.; Barbour, S.; Chalfant, C.; Conrad, D.H.; Kepley, C.L. Epoxyeicosatrienoic acids are involved in the C70 fullerene derivative–induced control of allergic asthma. J. Allergy Clin. Immunol. 2012, 130, 761–769.e762. [Google Scholar] [CrossRef] [Green Version]
  32. Zhou, Z.; Lenk, R.P.; Dellinger, A.; Wilson, S.R.; Sadler, R.; Kepley, C.L. Liposomal Formulation of Amphiphilic Fullerene Antioxidants. Bioconjugate Chem. 2010, 21, 1656–1661. [Google Scholar] [CrossRef] [Green Version]
  33. Basso, A.S.; Frenkel, D.; Quintana, F.J.; Costa-Pinto, F.A.; Petrovic-Stojkovic, S.; Puckett, L.; Monsonego, A.; Bar-Shir, A.; Engel, Y.; Gozin, M.; et al. Reversal of axonal loss and disability in a mouse model of progressive multiple sclerosis. J. Clin. Investig. 2008, 118, 1532–1543. [Google Scholar] [CrossRef]
  34. Tokuyama, H.; Yamago, S.; Nakamura, E.; Shiraki, T.; Sugiura, Y. Photoinduced biochemical activity of fullerene carboxylic acid. J. Am. Chem. Soc. 1993, 115, 7918–7919. [Google Scholar] [CrossRef]
  35. Dugan, L.L.; Turetsky, D.M.; Du, C.; Lobner, D.; Wheeler, M.; Almli, C.R.; Shen, C.K.-F.; Luh, T.-Y.; Choi, D.W.; Lin, T.-S. Carboxyfullerenes as neuroprotective& agents. Proc. Natl. Acad. Sci. USA 1997, 94, 9434–9439. [Google Scholar] [CrossRef] [Green Version]
  36. Wong, C.-W.; Zhilenkov, A.V.; Kraevaya, O.A.; Mischenko, D.V.; Troshin, P.A.; Hsu, S.-h. Toward Understanding the Antitumor Effects of Water-Soluble Fullerene Derivatives on Lung Cancer Cells: Apoptosis or Autophagy Pathways? J. Med. Chem. 2019, 62, 7111–7125. [Google Scholar] [CrossRef]
  37. Sigwalt, D.; Holler, M.; Iehl, J.; Nierengarten, J.-F.; Nothisen, M.; Morin, E.; Remy, J.-S. Gene delivery with polycationic fullerene hexakis-adducts. Chem. Commun. 2011, 47, 4640–4642. [Google Scholar] [CrossRef] [Green Version]
  38. Fan, J.; Fang, G.; Zeng, F.; Wang, X.; Wu, S. Water-Dispersible Fullerene Aggregates as a Targeted Anticancer Prodrug with both Chemo- and Photodynamic Therapeutic Actions. Small 2013, 9, 613–621. [Google Scholar] [CrossRef]
  39. Friedman, S.H.; DeCamp, D.L.; Sijbesma, R.P.; Srdanov, G.; Wudl, F.; Kenyon, G.L. Inhibition of the HIV-1 protease by fullerene derivatives: Model building studies and experimental verification. J. Am. Chem. Soc. 1993, 115, 6506–6509. [Google Scholar] [CrossRef]
  40. Mashino, T.; Shimotohno, K.; Ikegami, N.; Nishikawa, D.; Okuda, K.; Takahashi, K.; Nakamura, S.; Mochizuki, M. Human immunodeficiency virus-reverse transcriptase inhibition and hepatitis C virus RNA-dependent RNA polymerase inhibition activities of fullerene derivatives. Bioorg. Med. Chem. Lett. 2005, 15, 1107–1109. [Google Scholar] [CrossRef]
  41. Castro, E.; Martinez, Z.S.; Seong, C.-S.; Cabrera-Espinoza, A.; Ruiz, M.; Hernandez Garcia, A.; Valdez, F.; Llano, M.; Echegoyen, L. Characterization of New Cationic N,N-Dimethyl [70]fulleropyrrolidinium Iodide Derivatives as Potent HIV-1 Maturation Inhibitors. J. Med. Chem. 2016, 59, 10963–10973. [Google Scholar] [CrossRef]
  42. Yasuno, T.; Ohe, T.; Takahashi, K.; Nakamura, S.; Mashino, T. The human immunodeficiency virus-reverse transcriptase inhibition activity of novel pyridine/pyridinium-type fullerene derivatives. Bioorg. Med. Chem. Lett. 2015, 25, 3226–3229. [Google Scholar] [CrossRef]
  43. Kornev, A.B.; Khakina, E.A.; Troyanov, S.I.; Kushch, A.A.; Peregudov, A.; Vasilchenko, A.; Deryabin, D.G.; Martynenko, V.M.; Troshin, P.A. Facile preparation of amine and amino acid adducts of [60]fullerene using chlorofullerene C60Cl6 as a precursor. Chem. Commun. 2012, 48, 5461–5463. [Google Scholar] [CrossRef]
  44. Tollas, S.; Bereczki, I.; Borbás, A.; Batta, G.; Vanderlinden, E.; Naesens, L.; Herczegh, P. Synthesis of a cluster-forming sialylthio-d-galactose fullerene conjugate and evaluation of its interaction with influenza virus hemagglutinin and neuraminidase. Bioorg. Med. Chem. Lett. 2014, 24, 2420–2423. [Google Scholar] [CrossRef]
  45. Muñoz, A.; Sigwalt, D.; Illescas, B.M.; Luczkowiak, J.; Rodríguez-Pérez, L.; Nierengarten, I.; Holler, M.; Remy, J.-S.; Buffet, K.; Vincent, S.P.; et al. Synthesis of giant globular multivalent glycofullerenes as potent inhibitors in a model of Ebola virus infection. Nat. Chem. 2016, 8, 50–57. [Google Scholar] [CrossRef] [Green Version]
  46. Fedorova, N.E.; Klimova, R.R.; Tulenev, Y.A.; Chichev, E.V.; Kornev, A.B.; Troshin, P.A.; Kushch, A.A. Carboxylic Fullerene C60 Derivatives: Efficient Microbicides Against Herpes Simplex Virus and Cytomegalovirus Infections In Vitro. Mendeleev Commun. 2012, 22, 254–256. [Google Scholar] [CrossRef]
  47. Nakamura, E.; Isobe, H. Functionalized Fullerenes in Water. The First 10 Years of Their Chemistry, Biology, and Nanoscience. Acc. Chem. Res. 2003, 36, 807–815. [Google Scholar] [CrossRef]
  48. Debouck, C. The HIV-1 Protease as a Therapeutic Target for AIDS. AIDS Res. Hum. Retrovir. 1992, 8, 153–164. [Google Scholar] [CrossRef]
  49. Sijbesma, R.; Srdanov, G.; Wudl, F.; Castoro, J.A.; Wilkins, C.; Friedman, S.H.; DeCamp, D.L.; Kenyon, G.L. Synthesis of a fullerene derivative for the inhibition of HIV enzymes. J. Am. Chem. Soc. 1993, 115, 6510–6512. [Google Scholar] [CrossRef]
  50. Da Ros, T.; Prato, M. Medicinal chemistry with fullerenes and fullerene derivatives. Chem. Commun. 1999, 663–669. [Google Scholar] [CrossRef]
  51. Schuster, D.I.; Wilson, S.R.; Schinazi, R.F. Anti-human immunodeficiency virus activity and cytotoxicity of derivatized buckminsterfullerenes. Bioorg. Med. Chem. Lett. 1996, 6, 1253–1256. [Google Scholar] [CrossRef]
  52. Marcorin, G.L.; Da Ros, T.; Castellano, S.; Stefancich, G.; Bonin, I.; Miertus, S.; Prato, M. Design and Synthesis of Novel [60]Fullerene Derivatives as Potential HIV Aspartic Protease Inhibitors. Org. Lett. 2000, 2, 3955–3958. [Google Scholar] [CrossRef]
  53. Brettreich, M.; Hirsch, A. A highly water-soluble dendro [60]fullerene. Tetrahedron Lett. 1998, 39, 2731–2734. [Google Scholar] [CrossRef]
  54. Schuster, D.; Wilson, S.; Kirschner, A.; Schinazi, R.; Schlueter-Wirtz, S.; Barnett, T.; Martin, S.; Brettreich, M.; Hirsch, A. Evaluation of the anti-HIV Potency of a Water- Soluble Dendrimeric Fullerene Derivative. Proc. Electrochem. Soc. 2000. [Google Scholar]
  55. Toniolo, C.; Bianco, A.; Maggini, M.; Scorrano, G.; Prato, M.; Marastoni, M.; Tomatis, R.; Spisani, S.; Palu, G.; Blair, E.D. A Bioactive Fullerene Peptide. J. Med. Chem. 1994, 37, 4558–4562. [Google Scholar] [CrossRef]
  56. Bosi, S.; Da Ros, T.; Spalluto, G.; Balzarini, J.; Prato, M. Synthesis and Anti-HIV properties of new water-soluble bis-functionalized [60]fullerene derivatives. Bioorg. Med. Chem. Lett. 2003, 13, 4437–4440. [Google Scholar] [CrossRef]
  57. Marchesan, S.; Da Ros, T.; Spalluto, G.; Balzarini, J.; Prato, M. Anti-HIV properties of cationic fullerene derivatives. Bioorg. Med. Chem. Lett. 2005, 15, 3615–3618. [Google Scholar] [CrossRef]
  58. Nakamura, S.; Mashino, T. Water-soluble Fullerene Derivatives for Drug Discovery. J. Nippon Med. Sch. 2012, 79, 248–254. [Google Scholar] [CrossRef] [Green Version]
  59. Kobayashi, T.; Yasuno, T.; Takahashi, K.; Nakamura, S.; Mashino, T.; Ohe, T. Novel pyridinium-type fullerene derivatives as multitargeting inhibitors of HIV-1 reverse transcriptase, HIV-1 protease, and HCV NS5B polymerase. Bioorg. Med. Chem. Lett. 2021, 49, 128267. [Google Scholar] [CrossRef]
  60. Martinez, Z.S.; Castro, E.; Seong, C.-S.; Cerón, M.R.; Echegoyen, L.; Llano, M. Fullerene Derivatives Strongly Inhibit HIV-1 Replication by Affecting Virus Maturation without Impairing Protease Activity. Antimicrob. Agents Chemother. 2016, 60, 5731–5741. [Google Scholar] [CrossRef] [Green Version]
  61. Troshina, O.A.; Troshin, P.A.; Peregudov, A.S.; Kozlovskiy, V.I.; Balzarini, J.; Lyubovskaya, R.N. Chlorofullerene C60Cl6: A precursor for straightforward preparation of highly water-soluble polycarboxylic fullerene derivatives active against HIV. Org. Biomol. Chem. 2007, 5, 2783–2791. [Google Scholar] [CrossRef] [PubMed]
  62. Kraevaya, O.g.A.; Peregudov, A.S.; Troyanov, S.I.; Godovikov, I.; Fedorova, N.E.; Klimova, R.R.; Sergeeva, V.A.; Kameneva, L.V.; Ershova, E.S.; Martynenko, V.M.; et al. Diversion of the Arbuzov reaction: Alkylation of C–Cl instead of phosphonic ester formation on the fullerene cage. Org. Biomol. Chem. 2019, 17, 7155–7160. [Google Scholar] [CrossRef] [PubMed]
  63. Kraevaya, O.A.; Peregudov, A.S.; Godovikov, I.A.; Shchurik, E.V.; Martynenko, V.M.; Shestakov, A.F.; Balzarini, J.; Schols, D.; Troshin, P.A. Direct arylation of C60Cl6 and C70Cl8 with carboxylic acids: A synthetic avenue to water-soluble fullerene derivatives with promising antiviral activity. Chem. Commun. 2020, 56, 1179–1182. [Google Scholar] [CrossRef] [PubMed]
  64. Khakina, E.A.; Yurkova, A.A.; Peregudov, A.S.; Troyanov, S.I.; Trush, V.V.; Vovk, A.I.; Mumyatov, A.V.; Martynenko, V.M.; Balzarini, J.; Troshin, P.A. Highly selective reactions of C60Cl6 with thiols for the synthesis of functionalized [60]fullerene derivatives. Chem. Commun. 2012, 48, 7158–7160. [Google Scholar] [CrossRef] [Green Version]
  65. Voronov, I.I.; Martynenko, V.M.; Chernyak, A.V.; Balzarini, J.; Schols, D.; Troshin, P.A. Synthesis and Antiviral Activity of Water-Soluble Polycarboxylic Derivatives of [60]Fullerene Loaded with 3,4-Dichlorophenyl Units. Chem. Biodivers. 2018, 15, e1800293. [Google Scholar] [CrossRef]
  66. Kornev, A.B.; Peregudov, A.S.; Martynenko, V.M.; Balzarini, J.; Hoorelbeke, B.; Troshin, P.A. Synthesis and antiviral activity of highly water-soluble polycarboxylic derivatives of [70]fullerene. Chem. Commun. 2011, 47, 8298–8300. [Google Scholar] [CrossRef] [Green Version]
  67. Eropkin, M.Y.; Melenevskaya, E.Y.; Nasonova, K.V.; Bryazzhikova, T.S.; Eropkina, E.M.; Danilenko, D.M.; Kiselev, O.I. Synthesis and Biological Activity of Fullerenols with Various Contents of Hydroxyl Groups. Pharm. Chem. J. 2013, 47, 87–91. [Google Scholar] [CrossRef]
  68. Zhang, G.; Liu, Y.; Liang, D.; Gan, L.; Li, Y. Facile Synthesis of Isomerically Pure Fullerenols and Formation of Spherical Aggregates from C60(OH)8. Angew. Chem. Int. Ed. 2010, 49, 5293–5295. [Google Scholar] [CrossRef]
  69. Bertozzi, C.R.; Kiessling, A.L.L. Chemical Glycobiology. Science 2001, 291, 2357–2364. [Google Scholar] [CrossRef] [Green Version]
  70. Lundquist, J.J.; Toone, E.J. The Cluster Glycoside Effect. Chem. Rev. 2002, 102, 555–578. [Google Scholar] [CrossRef]
  71. Houseman, B.T.; Mrksich, M. Model Systems for Studying Polyvalent Carbohydrate Binding Interactions. In Host-Guest Chemistry: Mimetic Approaches to Study Carbohydrate Recognition; Penadés, S., Ed.; Springer: Berlin/Heidelberg, Germany, 2002; pp. 1–44. [Google Scholar] [CrossRef]
  72. Chabre, Y.M.; Roy, R. Chapter 6—Design and Creativity in Synthesis of Multivalent Neoglycoconjugates. In Advances in Carbohydrate Chemistry and Biochemistry; Horton, D., Ed.; Academic Press: Cambridge, MA, USA, 2010; Volume 63, pp. 165–393. [Google Scholar] [CrossRef]
  73. Dondoni, A.; Marra, A.; Scherrmann, M.-C.; Casnati, A.; Sansone, F.; Ungaro, R. Synthesis and Properties of O-Glycosyl Calix [4]Arenes (Calixsugars). Chemistry—A Eur. J. 1997, 3, 1774–1782. [Google Scholar] [CrossRef]
  74. de la Fuente, J.M.; Barrientos, A.G.; Rojas, T.C.; Rojo, J.; Cañada, J.; Fernández, A.; Penadés, S. Gold Glyconanoparticles as Water-Soluble Polyvalent Models to Study Carbohydrate Interactions. Angew. Chem. Int. Ed. 2001, 40, 2257–2261. [Google Scholar] [CrossRef]
  75. Roy, R. Syntheses and some applications of chemically defined multivalent glycoconjugates. Curr. Opin. Struct. Biol. 1996, 6, 692–702. [Google Scholar] [CrossRef]
  76. Tewart, R.J.; Boggs, J.M. A carbohydrate-carbohydrate interaction between galactosylceramide-containing liposomes and cerebroside sulfate-containing liposomes: Dependence on the glycolipid ceramide composition. Biochemistry 1993, 32, 10666–10674. [Google Scholar] [CrossRef] [PubMed]
  77. Imberty, A.; Chabre, Y.M.; Roy, R. Glycomimetics and Glycodendrimers as High Affinity Microbial Anti-adhesins. Chemistry—A Eur. J. 2008, 14, 7490–7499. [Google Scholar] [CrossRef]
  78. Luczkowiak, J.; Muñoz, A.; Sánchez-Navarro, M.; Ribeiro-Viana, R.; Ginieis, A.; Illescas, B.M.; Martín, N.; Delgado, R.; Rojo, J. Glycofullerenes Inhibit Viral Infection. Biomacromolecules 2013, 14, 431–437. [Google Scholar] [CrossRef]
  79. Hirsch, A.; Vostrowsky, O. C60 Hexakisadducts with an Octahedral Addition Pattern—A New Structure Motif in Organic Chemistry. Eur. J. Org. Chem. 2001, 2001, 829–848. [Google Scholar] [CrossRef]
  80. Ramos-Soriano, J.; Reina, J.J.; Illescas, B.M.; de la Cruz, N.; Rodríguez-Pérez, L.; Lasala, F.; Rojo, J.; Delgado, R.; Martín, N. Synthesis of Highly Efficient Multivalent Disaccharide/[60]Fullerene Nanoballs for Emergent Viruses. J. Am. Chem. Soc. 2019, 141, 15403–15412. [Google Scholar] [CrossRef]
  81. Muñoz, A.; Illescas, B.M.; Luczkowiak, J.; Lasala, F.; Ribeiro-Viana, R.; Rojo, J.; Delgado, R.; Martín, N. Antiviral activity of self-assembled glycodendro [60]fullerene monoadducts. J. Mater. Chem. B 2017, 5, 6566–6571. [Google Scholar] [CrossRef] [Green Version]
  82. Mattei, S.; Glass, B.; Hagen, W.J.H.; Kräusslich, H.-G.; Briggs, J.A.G. The structure and flexibility of conical HIV-1 capsids determined within intact virions. Science 2016, 354, 1434–1437. [Google Scholar] [CrossRef]
  83. Rodríguez-Pérez, L.; Ramos-Soriano, J.; Pérez-Sánchez, A.; Illescas, B.M.; Muñoz, A.; Luczkowiak, J.; Lasala, F.; Rojo, J.; Delgado, R.; Martín, N. Nanocarbon-Based Glycoconjugates as Multivalent Inhibitors of Ebola Virus Infection. J. Am. Chem. Soc. 2018, 140, 9891–9898. [Google Scholar] [CrossRef] [PubMed]
  84. Zhu, X.; Xiao, S.; Zhou, D.; Sollogoub, M.; Zhang, Y. Design, synthesis and biological evaluation of water-soluble per-O-methylated cyclodextrin-C60 conjugates as anti-influenza virus agents. Eur. J. Med. Chem. 2018, 146, 194–205. [Google Scholar] [CrossRef] [PubMed]
  85. Xiao, S.-L.; Wang, Q.; Yu, F.; Peng, Y.-Y.; Yang, M.; Sollogoub, M.; Sinaÿ, P.; Zhang, Y.-M.; Zhang, L.-H.; Zhou, D.-M. Conjugation of cyclodextrin with fullerene as a new class of HCV entry inhibitors. Bioorg. Med. Chem. 2012, 20, 5616–5622. [Google Scholar] [CrossRef] [PubMed]
  86. Sirotkin, A.K.; Zarubaev, V.V.; Poznyiakova, L.N.; Dumpis, M.A.; Muravieva, T.D.; Krisko, T.K.; Belousova, I.M.; Kiselev, O.I.; Piotrovsky, L.B. Pristine Fullerene C60: Different Water Soluble Forms—Different Mechanisms of Biological Action. Fuller. Nanotub. Carbon Nanostructures 2006, 14, 327–333. [Google Scholar] [CrossRef]
  87. Du, C.X. The antiviral effect of fullerene-liposome complex against influenza virus (H1N1) in vivo. Sci. Res. Essays 2012, 7, 705–711. [Google Scholar]
Figure 1. Possible interaction between fullerene molecules and coronavirus, in which fullerene molecules inhibit virus replication.
Figure 1. Possible interaction between fullerene molecules and coronavirus, in which fullerene molecules inhibit virus replication.
Nanomaterials 12 02547 g001
Figure 2. The first example of a water-soluble fullerene derivative (1) used as a virus inhibitor.
Figure 2. The first example of a water-soluble fullerene derivative (1) used as a virus inhibitor.
Nanomaterials 12 02547 g002
Figure 3. PM3-minimized structures of compounds 2 and 3. Adapted with permission from Ref. [52]. Copyright 2000 American Chemical Society.
Figure 3. PM3-minimized structures of compounds 2 and 3. Adapted with permission from Ref. [52]. Copyright 2000 American Chemical Society.
Nanomaterials 12 02547 g003
Figure 4. Computer-simulated interactions between compound 3 and HIVP. (a) Accommodation of compound 3 in the cavity of HIVP. (b) Closer view of the complex of compound 3 and HIVP. Adapted with permission from Ref. [52]. Copyright 2000 American Chemical Society.
Figure 4. Computer-simulated interactions between compound 3 and HIVP. (a) Accommodation of compound 3 in the cavity of HIVP. (b) Closer view of the complex of compound 3 and HIVP. Adapted with permission from Ref. [52]. Copyright 2000 American Chemical Society.
Nanomaterials 12 02547 g004
Figure 5. Fullerene amino acid derivatives (4) and (5) as potential inhibitors of HIV-1 cell replication.
Figure 5. Fullerene amino acid derivatives (4) and (5) as potential inhibitors of HIV-1 cell replication.
Nanomaterials 12 02547 g005
Figure 6. Synthesis route of fullerene amino ester derivative (8).
Figure 6. Synthesis route of fullerene amino ester derivative (8).
Nanomaterials 12 02547 g006
Figure 7. Synthesis route for fullerene penta-N-methyl piperazine salt (10).
Figure 7. Synthesis route for fullerene penta-N-methyl piperazine salt (10).
Nanomaterials 12 02547 g007
Figure 8. Structure diagram of fullerene pyrrolidine derivatives (1123).
Figure 8. Structure diagram of fullerene pyrrolidine derivatives (1123).
Nanomaterials 12 02547 g008
Figure 9. Structure diagram of fullerene pyrrolidine derivatives (2428).
Figure 9. Structure diagram of fullerene pyrrolidine derivatives (2428).
Nanomaterials 12 02547 g009
Figure 10. Structure diagram of fullerene pyrrolidine derivatives (2941).
Figure 10. Structure diagram of fullerene pyrrolidine derivatives (2941).
Nanomaterials 12 02547 g010
Figure 11. Chemical structures of the cationic N,N-dimethyl [70]Fullerene pyrrolidine iodide derivatives (4244).
Figure 11. Chemical structures of the cationic N,N-dimethyl [70]Fullerene pyrrolidine iodide derivatives (4244).
Nanomaterials 12 02547 g011
Figure 12. Structural diagram of carboxylic fullerene derivatives (4546).
Figure 12. Structural diagram of carboxylic fullerene derivatives (4546).
Nanomaterials 12 02547 g012
Figure 13. Schematic diagram of synthetic route of carboxylic fullerene derivative (48).
Figure 13. Schematic diagram of synthetic route of carboxylic fullerene derivative (48).
Nanomaterials 12 02547 g013
Figure 14. Schematic diagram of synthetic routes of carboxylic fullerene derivatives 49 and 50.
Figure 14. Schematic diagram of synthetic routes of carboxylic fullerene derivatives 49 and 50.
Nanomaterials 12 02547 g014
Figure 15. Structure diagram of polycarboxylic fullerene acid derivatives (5153).
Figure 15. Structure diagram of polycarboxylic fullerene acid derivatives (5153).
Nanomaterials 12 02547 g015
Figure 16. Structure diagram of fullerenol. Oxygen, carbon, and hydrogen atoms are marked in red, gray, and white, respectively.
Figure 16. Structure diagram of fullerenol. Oxygen, carbon, and hydrogen atoms are marked in red, gray, and white, respectively.
Nanomaterials 12 02547 g016
Figure 17. Structure diagram of glycofullerene (54). Adapted with permission from Ref. [78]. Copyright 2013 American Chemical Society.
Figure 17. Structure diagram of glycofullerene (54). Adapted with permission from Ref. [78]. Copyright 2013 American Chemical Society.
Nanomaterials 12 02547 g017
Figure 18. Synthesis of glycofullerenes 56–58 [45]. Adapted with permission from Ref [45]. Copyright 2015 Springer Nature.
Figure 18. Synthesis of glycofullerenes 56–58 [45]. Adapted with permission from Ref [45]. Copyright 2015 Springer Nature.
Nanomaterials 12 02547 g018
Figure 19. Structure diagram of nanoglycofullerene conjugate 61 [83]. Adapted with permission from Ref. [83]. Copyright 2018 American Chemical Society.
Figure 19. Structure diagram of nanoglycofullerene conjugate 61 [83]. Adapted with permission from Ref. [83]. Copyright 2018 American Chemical Society.
Nanomaterials 12 02547 g019
Figure 20. α-Cyclodextrin-C60 conjugate (62) [84]. Adapted with permission from Ref. [84]. Copyright 2012 Elsevier.
Figure 20. α-Cyclodextrin-C60 conjugate (62) [84]. Adapted with permission from Ref. [84]. Copyright 2012 Elsevier.
Nanomaterials 12 02547 g020
Figure 21. The structure of the fullerene liposome complex [87].
Figure 21. The structure of the fullerene liposome complex [87].
Nanomaterials 12 02547 g021
Table 1. Anti-HIV activity and cytostatic toxicity of compounds (1123) in CEM cell cultures.
Table 1. Anti-HIV activity and cytostatic toxicity of compounds (1123) in CEM cell cultures.
CompoundEC50 (μM)CC50 (μM)
HIV-1HIV-2
11>4>444.3
12 (trans-2)>4>47.2
13 (trans-3)>4>47.63
14 (trans-4)>4>47.4
15 (equatorial)>4>49.6
16 (trans-2)0.40 ± 0.0>44.79
17 (trans-3)0.96 ± 0.39>43.02
18 (trans-4)2.60 ± 0.88>413.2
19 (equatorial)1.60 ± 0.0>46.59
20 (trans-2)>4>4-
21 (equatorial)>4>4-
22 (trans-2)2.01 ± 0.0>4-
23 (equatorial)>4>4-
Table 2. Anti-HIV activity and cytostatic activity of compounds (2428) in CEM cell cultures.
Table 2. Anti-HIV activity and cytostatic activity of compounds (2428) in CEM cell cultures.
CompoundEC50 (μM)CC50 (μM)
HIV-1HIV-2
240.21 (±0.07)0.2 to 1.02.93 (±1.20)
250.35 (±0.07)0.70 (±0.42)9.04 (±0.18)
261.08 (±0.57)2.50 (±1.90)12.5 (±7.54)
27>25>25>125
282.50 (±0.71)>1028.7 (±1.27)
Table 3. HIV-RT inhibitory activity and cytotoxicity of fullerene pyrrolidine derivatives (2941).
Table 3. HIV-RT inhibitory activity and cytotoxicity of fullerene pyrrolidine derivatives (2941).
CompoundR1R2R3IC50 *CC50 *
292-(N-Methylpyridinium)–CH2COOC2H5–C2H50.30>50
302-(N-Methylpyridinium)–H–C2H50.33>50
312-Pyridine–CH2COOH–H0.20>50
322-Pyridine–H–H0.46>50
333-(N-Methylpyridinium)–CH2COOC2H5–C2H50.25>50
343-(N-Methylpyridinium)–H–C2H50.094>50
353-Pyridine–CH2COOH–H0.4139.4
363-Pyridine–H–H0.080>50
374-(N-Methylpyridinium)–CH2COOC2H5–C2H50.74>50
384-(N-Methylpyridinium)–H–C2H50.37>50
394-Pyridine–CH2COOH–H1.60>50
404-Pyridine–H–H0.80>50
41--->500>50
Nevirapine---3.52-
* These values (~µM) are based on the average of three test results for each test compound.
Table 4. IC50 and RIP values of different glycofullerene derivatives.
Table 4. IC50 and RIP values of different glycofullerene derivatives.
CompoundIC50/(nM)Mannoses (No.)RIP *
56 (120 mannoses)0.6671201.58 × 104
58 (120 mannoses)20.3751205.2 × 102
54 (36 mannoses)300361.17 × 102
* Relative inhibitory effect, calculated as (IC50)mono/IC50 * valency ((IC50)mono, IC50 of the monovalent compound; IC50 * valency, IC50 of the multivalent compound multiplied by the number of ligands present in the multivalent compound).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Xu, P.-Y.; Li, X.-Q.; Chen, W.-G.; Deng, L.-L.; Tan, Y.-Z.; Zhang, Q.; Xie, S.-Y.; Zheng, L.-S. Progress in Antiviral Fullerene Research. Nanomaterials 2022, 12, 2547. https://doi.org/10.3390/nano12152547

AMA Style

Xu P-Y, Li X-Q, Chen W-G, Deng L-L, Tan Y-Z, Zhang Q, Xie S-Y, Zheng L-S. Progress in Antiviral Fullerene Research. Nanomaterials. 2022; 12(15):2547. https://doi.org/10.3390/nano12152547

Chicago/Turabian Style

Xu, Piao-Yang, Xiao-Qing Li, Wei-Guang Chen, Lin-Long Deng, Yuan-Zhi Tan, Qianyan Zhang, Su-Yuan Xie, and Lan-Sun Zheng. 2022. "Progress in Antiviral Fullerene Research" Nanomaterials 12, no. 15: 2547. https://doi.org/10.3390/nano12152547

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