Open access peer-reviewed chapter

Functionalized Carbon Nanomaterials in Drug Delivery: Emergent Perspectives from Application

Written By

Nabanita Saikia

Submitted: 25 June 2017 Reviewed: 23 October 2017 Published: 20 December 2017

DOI: 10.5772/intechopen.71889

From the Edited Volume

Novel Nanomaterials - Synthesis and Applications

Edited by George Z. Kyzas and Athanasios C. Mitropoulos

Chapter metrics overview

1,686 Chapter Downloads

View Full Metrics

Abstract

Carbon nanotubes (CNTs) have attracted substantial research interest in biomedical sciences and bionanotechnology, rendered from its unique structure, electronic, mechanical, and optical properties. Despite the diverse potential applications, the integration of CNTs in biomedical research is one of the most challenging areas where nanotubes fall under much scrutiny. Pristine nanotubes are highly hydrophobic, and non-dispersible in most of the common aqueous and organic solvents and to render nanotubes biocompatible, functionalization is one of the key prerequisites. In this regard, covalent and noncovalent functionalization are the two widely adopted approaches for co-tethering biologically active molecules on the CNTs. Likewise, the hollow cavity of the nanotube facilitates in the endohedral encapsulation of biomolecules, peptides, DNA oligonucleotides, and proteins, thereby retaining the physiological attributes of the biological molecules. The chapter focuses on the emerging approaches to the functionalization of single-wall CNTs (SWCNTs) and the potential application of functionalized SWCNTs in tuberculosis and cancer chemotherapy using state-of-the-art density functional theory, molecular docking and molecular dynamics simulation methods.

Keywords

  • carbon nanotubes
  • drug delivery
  • molecular dynamics
  • density functional theory

1. Introduction

1.1. Carbon: The fundamental building block of life

Carbon is the most versatile element in the periodic table that forms the basis of all kinds of life on earth. Elemental carbon displays a complex allotropy depending on the nature of hybridization; diamond (sp3 hybridized), graphite, graphene, fullerenes, and carbon nanotubes (sp2 hybridized). Graphite is the most common allotrope of carbon and the word graphite in Greek means ‘to write’. Graphene an acronym for the 2D layered graphite, is the mother of all carbon materials [1], as a graphene sheet can be wrapped to form 0D fullerenes, rolled to form 1D nanotubes, or stacked to form 3D graphite as depicted in Figure 1. The unearthing of “ground-breaking experiments regarding the two-dimensional material graphene” by Geim and Novoselov in 2010, heralded graphene as the next generation carbon material [2].

Figure 1.

Formation of SWCNT, fullerene and graphite from a single graphene monolayer.

CNTs are hexagonally arranged, honey-combed lattice of carbon atoms formed by the rolling of graphene into seamless cylindrical structures (see Figure 2a). Nanotubes like graphene have a high diameter to length ratio (aspect ratio) [3] and demonstrate high electrical, mechanical, and thermal conductivity along with structural stability [4, 5, 6, 7]. CNTs are broadly classified as single-wall CNTs (SWCNTs) and multi-wall CNTs (MWCNTs). The SWCNT comprise of a single graphene sheet, with diameter ~ 0.5–1.5 nm and length of ~100 μm [8], while MWCNT is formed from the co-axial stacking of SWCNTs, with diameter ~1.4–100 nm, length between 1 nm-μm, and internuclear distance of 0.3–0.4 nm between the co-axial tubes. The representation of a zigzag (m = 0), armchair (n = m), and chiral (n ≠ m) nanotube is depicted in Figure 2b-d. The (n, m) indices render remarkable electronic properties to the CNTs [9] and the sp2 hybridization along the tubular axis makes it chemically inert by nature.

Figure 2.

(a) A graphene sheet depicting the (b) zigzag and (c) armchair and (d) chiral CNT based on rolling of carbon atoms along chiral vectors through the circumference (OA) of nanotube.

The unique electronic properties exhibited by CNT are governed by the quantum confinement of electrons where the periodic boundary conditions come into interplay. Because of the quantum confinement, electrons can propagate along the tube axis: forward and backward, along with the conservation of energy and momentum. Unlike metals which have a smooth density of states (DOS), CNTs are characterized by many van Hove singularities [10], and the DOS depends on diameter and chirality of the nanotube [11]. The conducting properties of CNT is an inverse function of its diameter, that is, with increase in diameter, band gap between the valence and conduction bands decreases and at a certain point both the bands overlap to give rise to metallic nanotubes. Semiconducting nanotubes on the other hand (with similar diameter as metallic nanotubes) possess similar van Hove singularities near the Fermi level [12].

These unprecedented properties have largely contributed to the extensive biomedical research, especially as nanocapsules for therapeutic drugs, proteins, and gene delivery [13]. CNTs find application in bionanotechnology and pharmaceutical sciences, and current drug delivery modules have been incorporating CNTs for improved target specific detection and treatment of diseases. Although the potential application of carbon nanomaterials, particularly CNTs are farfetched, the concerns raised over the effect of large-scale synthesis of CNTs to the environment, biocompatibility, toxicity, biodegradation and remediation cannot be undermined and needs to be addressed thoroughly. Hence, a detailed in vivo and in vitro toxicity analyses is mandatory in understanding CNT-based therapeutic regimes for sustained drug delivery applications.

Some of the questions that underlie the importance of the study are (i) understanding the mechanism of CNT uptake followed by the subsequent release of therapeutic molecules, (ii) in vivo biocompatibility, and (iii) long-term practical implication to direct exposure to the physiological environment. Although theoretical and/or experimental studies have attempted to address the main questions like mechanism of drug-nanotube interaction, the preferable binding sites of drugs onto nanotubes, drug activity under confinement, and change in redox properties of drugs under the physiological conditions, these studies are rather limited in predicting the likelihood of using CNT as carrier vehicles for the long-term storage and release of therapeutic and biologically active molecules in vivo.

The chapter in a very comprehensive yet succinct way addresses the potential applications of SWCNTs in drug delivery, managing to draw a fine line between the scopes of application and practical viability of integrating carbon nanomaterials in biomedical research. Herein, we report the theoretical aspects of modeling novel SWCNT-based drug delivery systems using the covalent and noncovalent functionalization schemes. Nanotubes of varying chirality and length are considered for functionalization, drug loading, and targeting onto the active binding sites of receptor proteins. With the successful incorporation of CNTs in cancer therapy, we propose a novel approach toward integrating CNTs in Tuberculosis (TB) therapy. To the best of our knowledge, theoretical studies on the potential application of CNTs in pharmaceutical sciences pertaining to TB and other bacterial diseases have not been discussed extensively. We address the recent theoretical advancements using the state-of-the-art density functional theory (DFT), molecular docking, and molecular dynamics (MD) simulation methods. Molecular docking serves as an instrumental tool in computer-aided drug design for predicting the preferred binding mode of a ligand to a receptor (protein). Docking studies help characterize the protein binding cavity, understand the orientation of ligand with respect to the receptor protein, and the nature of interaction between the protein with functionalized nanomaterial, which can aid in the structure-based design of novel drug delivery systems for future experimental studies.

Advertisement

2. Functionalization of CNTs

Traditional approaches to drug delivery function over a broad spectrum, resulting which, specificity toward drug administration and delivery are rarely accomplished. Development of polymer-based nanocomposite materials has enabled the successful engineering of drug delivery modules via the incorporation of nanomaterials and nanoparticles as nanocapsules for sustained release of therapeutics in a dosage-dependent manner. Nanotechnology, on the other hand, has revolutionized the pharmaceutical sector with the assimilation of functionalized nanomaterials like CNTs in drug, gene delivery, and tissue engineering [14, 15]. CNTs play dual role by rendering directionality in targeting the tumor (malignant) cells and facilitating the controlled mediated release of therapeutic molecules. The application of CNTs as carrier payloads for anticancer drugs cisplatin [16], carboplatin [17], doxorubicin (DOX) [18, 19, 20], mechlorethamine [21], paclitaxel [22] and antitubercular drugs like isoniazid (INH) [23], rifampicin, pyrazinamide (PZA) [24] have been reported.

With the inherent limitations in application of pristine, unmodified nanotubes, functionalization is the collective approach toward tailoring nanotubes electronic properties. Pristine CNTs are generally hydrophobic with low solubility in most of the common aqueous and organic solvents and the hydrophobicity is accounted to the size, structure, and bundling effect which restricts the uptake and assimilation within the biological environment [25]. Functionalization assists in reducing the bundling effect which arises due to the van der Waals (vdW) attractive forces between adjacent nanotube surfaces and is efficient in increasing the biocompatibility thereby facilitating cellular internalization and trafficking. It has been reported that the functionalized nanotubes (fCNT) exhibit better biocompatibility with reduced in vivo and in vitro toxicity [26, 27, 28]. The extent of functionalization depends on the nature and reactivity of sidewall (curvature), number of functional groups that can be co-tethered along the sidewall, and steric hindrance between functional groups and nanotube sidewall. The subsequent sections discuss some of the adopted approaches in the functionalization of SWCNTs at the level of experiment and theory.

2.1. Solubilization of CNT through covalent functionalization

Some of the alternative schemes to functionalization of CNT is through covalent method using 1,3-dipolar cycloaddition [29], [2 + 1] cycloaddition of dichlorocarbene, silylene, germylene [30], hydroboration [31], arylation, hydrogenation by Birch reduction [32], carboxylic acid groups [33], Diels-Alder reaction, esterification of carboxylated nanotubes [34] and fluorination reactions [35]. Experimental and theoretical studies have shown that the extent of covalent functionalization depends on the curvature of nanotube as an increase in curvature decreases the reactivity toward sidewall functionalization [36].

2.1.1. 1,3-dipolar Cycloaddition (DC)

The solubility of CNT can be enhanced by the covalent functionalization using 1,3-DC reactions. Azomethine ylide (CH2NHCH2), ozone (O3), nitrone (CH2N(H)O), nitrile ylide (CHNCH2), nitrile imine (CHNNH) are the commonly used functional groups for 1,3-DC reaction. The intrinsic physical properties of CNTs such as photoluminescence and Raman scattering decreases upon covalent functionalization, due to chemical bond formation between the functional group and carbon atoms. Theoretical studies by Lu et al. [30] reported the reaction energies (Er), barrier heights (Ea) and retro barrier height values of a series of 1,3-dipolar molecules on (5, 5) SWCNT using two-layered ONIOM (B3LYP/6-31G*:AM1) approach. Likewise, experimental studies by Prato and co-workers [37] substantiated the theoretical findings on the 1,3-DC functionalization of CNTs. 1,3-DC functionalization was also achieved through the addition of ozone wherein ozone adds onto the end caps and kink regions rather than nanotube sidewall due to increased strain and loss in conjugation. Lu et al. [38] reported the 1,3-DC reaction of ozone onto nanotube sidewalls using two-layered ONIOM (B3LYP/6-31G*:AM1) method.

Prato and co-workers [39] investigated the 1,3-DC reaction of azomethine ylide on both SWCNT and MWCNT. The water-soluble amine functionalized CNT was highly suitable for immobilization of biomolecules, and purification of pristine nanotubes during syntheses. Attachment of peptide molecules onto covalently functionalized SWCNT was reported by Prato and co-workers [40]. The C-terminal group of peptide chain was attached to N-terminal side group to form a supramolecular complex of peptide wrapped nanotubes. Gallo et al. [23], incorporated fSWCNT and fullerenes as nanovectors for the functionalization of INH drug. Armchair (5, 5) SWCNT was functionalized via 1,3-DC reaction of azomethine ylide with the polyethylene glycol (PEG) oligomer tailored to the INH drug. Increasing the number of functionalized units leads to an increase in HOMO-LUMO energy gap and global hardness, and decrease in binding (−3.52 to −6.65 eV) and solvation energy (−31.60 to −49.99 eV) values. An increase in global hardness with increase in functionalization suggests a net stabilization of the complex. It is noteworthy to mention that the optimum length of PEG oligomer used as a linker for the 1,3-DC functionalization is essential as longer PEG chains can interfere with drug administration, block the interaction between the nanotube and cell lines of the body, cellular uptake of drug, and degrade the therapeutic activity of drug molecules [41]. The PEG units with superior hydrophilicity, biocompatibility, and low immunogenicity can resist the opsonisation and increase the retention time of the nanotube-drug conjugate system in vivo [42, 43].

The structure, electronic properties, and reactivity of a series of 1,3-DC functionalized armchair (n, m) and zigzag (n, 0) SWCNTs with antitubercular drugs 2-methyl heptyl isonicotinate (MHI) and PZA via. PEG linker was investigated using first-principles DFT calculations [44, 45, 46]. With increase in sidewall functionalization, the global hardness and HOMO-LUMO energy gap decreases suggesting an overall decrease in stability of the complex, which is indicative of the localized induced deformation in the nanotube at the site of covalent attachment. On the other hand, the solubility of bare INH and PZA drugs was enhanced in presence of nanotube support. We showed that the optimum length and chirality of the nanotube is central to understand the electronic properties of functionalized nanotubes, particularly from a drug delivery perspective.

2.1.2. Functionalization using organic acids

Covalent functionalization of CNTs using carboxylic (−COOH) group was realized through oxidation with strong organic acids like H2SO4/HNO3 [47], phosphates, and sulfur-containing units. Acid functionalized CNTs are highly soluble in water under a wide range of pH and exhibit a significant reduction in the aggregation of nanotube bundles. The dispersibility facilitates in the sidewall, endohedral and end tip functionalization of CNT with organic acids and different functional groups. The sidewall functionalization of CNTs via cycloaddition reaction with azide, ozone, transition metal oxides, and carbenes [48] is illustrated in Figure 3.

Figure 3.

Schemes for sidewall functionalization of SWCNT using covalent bonds. Adopted from Ref. [48].

Lu et al. [31] using two-layered ONIOM (B3LYP/6-31G*:AM1) approach reported the reaction pathway and site selectivity for [2 + 1] cycloaddition of dichlorocarbene, silylene, germylene, and oxycarbonylnitrene onto (5, 5) SWCNT. Dichlorocarbene addition occurs preferentially at the 1,2-pair site. The silylene addition at 1,2-pair site was predicted to be exothermic (−20.7 kcal mol−1) and follows a barrier less reaction pathway. Germylene addition was exothermic by 8.5 kcal mol−1, lower than dichlorocarbene and silylene and proceeds in absence of a transition state pathway. Oxycarbonylnitrene addition onto 1,2-pair site of SWCNT was exothermic by 66.2 kcal mol−1, higher than the other three cycloaddition groups. The transition state had an activation barrier of 7.2 kcal mol−1, which suggested that the cycloaddition reaction was facile in nature.

2.2. Noncovalent functionalization of CNTs

Although covalent functionalization improves the solubility of CNT, it modifies the intrinsic electronic properties by deforming the C-C bond length, perturbing the π-delocalization, and shortening the length of the nanotube. Noncovalent functionalization provides the alternative approach to improving the solubility of nanotubes without deforming the π-delocalization. For example, exohedral wrapping with polymeric molecules like PEG [49], polymers [50], ss-DNA, and endohedral filling can help in increasing the solubility. Likewise, the polymer molecules can form a surface coating via π-stacking interactions, mediated by weak vdW forces, and hydrophobic interactions. The following subsection discusses some of the widely adopted approaches to the noncovalent functionalization of CNTs.

2.2.1. Functionalization via π-π stacking

The π-π stacking of organic molecules namely pyrene, anthracene, and porphyrin increases the solubility of pristine nanotubes and facilitates in the binding of proteins, polysaccharides, and peptides. Dai and co-workers [51] investigated the noncovalent functionalization of CNT, wherein succinimidyl ester group was co-tethered onto pyrene rings via butanoic acid side chains, facilitating the immobilization of proteins. The amide group of the protein replaces the N-hydroxysuccinimide group that propagates the transportation of biomolecules. Falvo and co-workers [52] reported the functionalization of MWCNT with streptavidin protein, wherein the MWCNT pre-functionalized with 1-pyrene butanoic acid succidymidyl ester (1-pbase) was co-tethered on the nanotube sidewall. The pyrenes formed π-π bonds with the MWCNT sidewall under the influence of which MWCNT undergoes a phase transfer with 1-base acting as a phase transfer catalyst.

The noncovalent functionalization of pyrene on SWCNT was investigated by Cosnier and co-workers [53] for application as modified electrodes in biosensing devices. Calomel electrode was taken as the reference and Pt electrode (5 mm diameter) modified by casting 20 μl THF solution of pristine SWCNT and B-doped SWCNT polished with 20 μm diamond paste as the counter electrode. The SWCNT/pyrene-biotin and B-SWCNT/pyrene-biotin was incubated in 20 μl avidin solutions for 20 min, and the response time for glucose sensing was measured using amperometric response technique. The enzyme-modified SWCNT electrodes were incorporated as electrodes for glucose sensing. In-situ polymerization of MWCNT with polyimide (PI) results in the π-stacking interactions between the imide and aromatic benzene rings of CNT with subsequent wrapping of PI along the nanotube circumferential axis [54]. Polymer wrapping improves the thermal stability and renders the conjugated complex suitable for nanoelectronics devices with improved electronic, thermal and optical properties.

Noncovalent functionalization of porphyrin molecules with SWCNTs have been extensively studied as high yield light-harvesting systems with tunable electronic properties for biological and optoelectronic applications [55, 56]. Roquelet et al. [57], reported an efficient method for the synthesis of porphyrin/SWCNT complex utilizing a micelle-swelling technique in presence of organic solvent. The organic solvent leads to swelling of the micelle facilitating the interaction of porphyrin molecules to the micelle core and SWCNT. Dispersion corrected DFT calculations on the structure, electronic and optical properties of SWCNT functionalized tetraphenylporphyrin (TPP) molecule showed that diameter rather than chirality of the nanotube stabilizes the π-π stacking of TPP molecule [58]. The optical absorption of TPP was not affected by the diameter or chirality of CNT and the optical spectra showed the absorption of π-stacked TPP at almost the same position as the isolated TPP, indicating that the TPP absorption properties were preserved in the complex.

2.2.2. Functionalization using biomolecules and nucleobases

Functionalization of CNTs with biomolecules is useful in the development of nanobio composite devices. Immobilization of DNA in DNA-based biosensors is possible with the incorporation of CNTs in nucleic acid sensing, gene therapy, and biosensor fabrication [59, 60, 61]. DNA because of the base pairing sequence facilitates in the alignment of nanotube assembly [62]. Rodger and co-workers [63] investigated the interaction of CNT with DNA using linear dichroism (LD) method. DNA/CNT hybrid exhibited higher LD signals, higher than the sum of the LD spectrum of individual DNA and SWCNT. Jung et al. [64] developed methods for covalent linking of DNA oligonucleotides onto SWCNT films which were later immobilized onto solid surfaces. The carboxylated/aminated DNA oligonucleotides were covalently attached to functionalized SWCNT, the length of which was controlled via oxidation with strong organic acids, leading to the formation of carboxylated SWCNT.

Li et al. [65] investigated the self-assembly of CNT and gold nanoparticles into multicomponent structures using DNA oligonucleotides. The CNT pre-functionalized with -COOH facilitated in the grafting of ssDNA strands and multiple assemblies of nanotubes were thus possible using this technique. In another combined theoretical and experimental study by Sood and co-workers [66], interaction of DNA nucleobases namely adenine (A), guanine (G), cytosine (C), thymine (T) with (5, 5) SWCNT was reported. The ab initio studies showed that binding energies of nucleobases onto SWCNT was governed mainly by vdW forces and follow the order: C > G > A > T, respectively. Likewise, the binding energies of A, G, C, T, U nucleobases on (7, 0) SWCNT was predicted to follow the order: G > C > A > T > U, and bears a monotonic dependence on nanotube diameter; that is, nanotubes with small diameter due to low curvature exhibits low interaction energy whereas for nanotubes with high diameter the interaction energy tend to be on the higher side [67].

2.2.3. Noncovalent functionalization using polymers

Polymer wrapping of CNTs mediated via noncovalent functionalization toward the synthesis of highly dispersed, stable and reinforced functional dispersants in aqueous and organic solvents was reviewed by Fujigaya and Nakashima [68]. The polymer wrapping forms a thermodynamically stable coating and any unbound polymer could be removed via dialysis, ultra-centrifugation or chromatographic separation techniques. Similarly, noncovalent functionalization of CNTs using polyethylene glycol (PEG) PEGylated-phospholipid chains forms an effective means of high loading of the drug and biomolecules at the free end of PEG chain and onto nanotube sidewall. PEGylated-phospholipid chain facilitates the high loading (about ~400%) of drug molecules onto CNT and characterizes as a potent carrier vehicle in drug delivery applications. PEG tailored SWCNT exhibit no toxicity for over several months, which was further substantiated from time-dependent assays performed onto mice. Drugs which normally remained insoluble within the biological systems, upon conjugation with PEG modified CNT revealed high solubility as well as retention time within the body. The noncovalent functionalization retained the physical properties of nanotubes without drastically perturbing the overall electronic properties.

First-principles studies on the interaction of conjugated polymers with (8, 0) SWCNT and (10 × 10) graphene sheet was investigated by Jilili et al. [69], to confirm the experimental observation that polymers are suitable for noncovalent functionalization. The GGA approximation was predicted to be inadequate in describing the physisorbed systems, whereas LDA and vdW corrected GGA yield conclusive results. The electronic structure of SWCNT/graphene was maintained around the Fermi energy with negligible charge transfer between the conjugates. The interaction of polymer-SWCNT/graphene was of weak vdW type with minimal effects on the physical and electronic properties of SWCNT/graphene, important for an effective noncovalent functionalization.

Advertisement

3. Functionalized carbon Nanomaterials in drug delivery

Drug delivery is a process of administering drugs in a controlled, sustained manner to achieve maximal therapeutic efficacy upon transdermal administration. The foremost objectives in developing novel drug delivery systems are to improve the therapeutic competence by [1] increasing bioavailability, [2] preventing toxicity, harmful side-effects by increasing the persistence of a drug, [3] reducing drug exposure toward non-target cells, and [4] minimizing drug degradation and loss [70, 71, 72, 73]. Drug delivery systems are designed to improve the pharmacological and therapeutic profile of drug molecules with an ability to cross the cell membrane upon administration [74, 75]. The most important characteristic of SWCNT as a drug delivery system is its ability to penetrate the cell membrane [76], and facilitate the intracellular internalization and trafficking within the cell cytoplasm [77]. A major breakthrough in nanoscience was the advent of CNTs as one of the most sought-after materials for designing novel drug delivery carrier modules to comply with the biotechnological and pharmaceutical objectives. CNT due to its needle-like cylindrical structure can easily penetrate the cell membrane and enter the cell nuclei, while the cell does not recognize it as an intruder.

Functionalized CNTs can act as carriers for antimicrobial agents like amphotericin B [78, 79] and transport it within the mammalian cells. This reduces the antifungal toxicity as compared to the toxicity of free drug (40% of the cells being killed by CNTs-free formulation compared to no cell death by CNTs formulation). The surface-engineered CNTs can capture the pathogenic bacteria in liquid media [80, 81]. In addition, SWCNTs exhibit unique optical properties such as near-infrared region (NIR) fluorescence, and Raman scattering. The fluorescence range spans the entire biological tissue transparent window and is, therefore, promising for drug detection and biological imaging [82, 83, 84].

3.1. SWCNTs in tuberculosis therapy

The science of bacteriology is credited to the contributions of Louis Pasteur and Robert Koch. It was the discovery of Mycobacterium tuberculosis by Koch that revolutionized medical history [85]. TB is a chronic disease caused by the infection of Mycobacterium tuberculosis [86] and is a leading cause of mortality worldwide. The World Health Organization (WHO) 2017 annual report prompted to 10.4 million new TB cases, of which, India and Indonesia alone accounted for a third of the world’s TB-burden [87]. In 2016, a total of 9287 new TB cases were reported in the United States [88]. The drastic widespread of TB is mainly accounted to poverty, homelessness, synergy with HIV/AIDS pandemic, multi-drug resistant (MDR), and extensively drug resistant (XDR) stains of M. tuberculosis [89].

Streptomycin was the first antitubercular drug discovered in 1943 [90] and since then several therapeutic drugs like para-amino salicylic acid (1946), isoniazid (INH) (1952), pyrazinamide (PZA) (1952), cycloserine (1952), ethionamide (1956), rifampicin (1957) and ethambutol (1962) have been discovered. The TB therapy involves a combination of four first-line drugs namely; INH, PZA, rifampicin, and ethambutol administered for a period of 2 months followed by minimum 4 months’ treatment regimen of INH and rifampicin [91]. PZA (pyrazine-2-carboxamide) is one of the first-line drugs used in TB treatment recommended by the WHO. PZA is metabolized into its active form (pyrazoic acid) by the amidase activity of M. tuberculosis nicotinamidase/pyrazinamidase (MtPncA) encoded by the pncA gene [92]. The administration of PZA in high dosage can cause minor to detrimental health problems and the antibiotic resistance of bacteria under prolonged exposure triggers the need for better drug delivery methods to directly bind with the TB bacteria.

We performed DFT, molecular docking and MD studies on the SWCNT-mediated PZA delivery onto the active site of M. Tuberculosis pncA enzyme [93]. The DFT calculations predict that the covalent functionalization was thermodynamically favored with negative binding energy values. The decrease in binding energy of PZA/SWCNT with increase in nanotube diameter illustrates that the curvature of nanotube plays an important role in determining the reactivity, and nanotubes with narrow diameter are thermodynamically favorable compared to tubes with larger diameter. The molecular docking studies supported the DFT results thereby establishing that, incorporation of SWCNT facilitates in target specific delivery of PZA within the binding site of pncA as shown in Figure 4. The narrow diameter nanotubes were better docked compared to the larger diameter nanotubes and length of PEG chain was predicted to be reasonably adequate for the delivery of PZA within the binding site of pncA. The presence of nanotube did not result in any structural deformation in pncA, rather the incorporation of SWCNT facilitated in the stabilization of PZA conjugated complex.

Figure 4.

The docked conformation and hydrophobic surface of sidewall functionalized PZA/SWCNT within the active binding site of pncA protein, (a–b) PZA/(9,0) SWCNT (3 unit cells), (c–d) PZA/(9,0) SWCNT (5 unit cells), (e–f) edge functionalized PZA/(9,0) SWCNT (5 unit cells). Reprinted with permission from Ref. [93]. Copyright 2017, Elsevier.

Noncovalent functionalization of SWCNT and boron nitride nanotubes (BNNTs) with PZA was investigated using DFT and MD simulation (see Figure 5) to comprehend the role of nanotube chirality on the electronic properties of the complexes [94]. BNNTs are structural analogs of CNTs with a wide band gap of ~5.5 eV, high chemical, and thermal stability. The potential application of BNNTs is rather limited in terms of its high chemical stability and poor dispersibility. The theoretical results predict the modification in electronic structure of both SWCNTs and BNNTs with the enhancement of electronic states, significant lowering in HOMO-LUMO energy gap and the presence of new dispersionless states within the band gap. Depending on the nanotube chirality, PZA exhibits a preferential selectivity for adsorption, which is further confirmed from the band structure, DOS, total projected DOS, and frontier orbital analysis.

Figure 5.

(a) Adsorption sites in the model SWCNT, (b) Adsorption sites in a model (5, 5) BNNT; optimized geometries of (c) PZA, (d) (5, 5) CNT, (e) (8, 0) CNT and (f) (5, 5) BNNT. Adopted from Ref. [94].

The functionalized nanotube facilitates in the loading and delivery of a PZA onto the active entering pathway of pncA without the nanotube affecting the structural conformation of pncA as shown in Figure 6. The incorporation of nanotube yields better docking scores for PZA then the drug being administered in bare form. Although covalent functionalization aids in achieving target specific delivery of PZA within the active site of pncA, noncovalent functionalization was predicted to be effective for engineering nanotube structure and electronic properties for successful drug delivery applications.

Figure 6.

(a) Docked PZA/pncA, (b) electrostatic surface (c) PZA/(5, 5) SWCNT docked onto pncA, (d) electrostatic surface, (e) PZA/(8, 0) SWCNT docked onto pncA, (f) electrostatic surface, (g) docked PZA/(5, 5) BNNT with pncA, (h) electrostatic surface. Adopted from Ref. [94].

Zanella et al. [95] performed theoretical studies on the interaction of non-steroid anti-inflammatory drug nimesulide with pristine and Si-doped capped SWCNT. The adsorption of nimesulide on Si-doped capped SWCNT exhibit a higher binding energy of 1.8 eV compared to pristine capped SWCNT (0.32 eV) which was due to the high reactive bonding sites on Si atom. The strong interaction of nimesulide with Si-doped SWCNT served as better drug delivery carriers in comparison to pristine capped SWCNT.

Wang and co-workers [96] performed MD studies to investigate the mechanism of encapsulation of nifedipine drug within (10, 10) SWCNT. Their studies showed that the internal adsorption of nifedipine was more stable than external adsorption by 5.3 to 7.8 kcal/mol. In solvent phase, the encapsulation of nifedipine was impeded due to competitive vdW and hydrophobic interactions in SWCNT-nifedipine-water complex. Encapsulation of nifedipine orients the distribution of water molecules inside SWCNT accompanied by the H-bond formation between water molecules and oxygen atom of nifedipine. During the encapsulation process, SWNT undergoes weak fluctuations due to the oscillatory behavior of nifedipine encapsulated within the CNT.

Advertisement

4. SWCNTs in cancer therapy

Platinum-based Phase II and Phase III anticancer drugs hold promise in the treatment of cancer with new drugs being discovered, some of which are still under clinical trials. The two main limitations in use of Pt-based anticancer drugs are [1] the anticancer drugs undergo poor circulation in tissue cells and its activity is reduced with time due to the complex formation with plasma and tissue cells, and [2] tumor cells demonstrate resistance toward Pt-based drugs under prolonged exposure, rendering them ineffective as potent anti-tumor agents. Lippard and co-workers [16, 17] incorporated capped fCNT as longboat delivery vehicles for cisplatin anticancer drug through clathrin-dependent endocytosis and measured the changes in redox potential before and after release of the drug. The substituted c,c,t [Pt(NH3)2Cl2(OEt)(O2CCH2CH2COOH)] pro-drug was attached to SWCNT functionalized with phospholipid tethered amine with PEG to solubilize the nanotube. Burger et al. [97] investigated the encapsulation of cisplatin in a phospholipid formulation. The lipid-coated cisplatin nanocapsules exhibit drug-lipid ratio and in vitro cytotoxicity 1000 times higher than free cisplatin. This method thus formed an effective approach in drug delivery and the means of producing lipid-based nanocapsules for encapsulating different bio- and therapeutic molecules. Hilter and Hill [98] suggested three preferred orientations of cisplatin toward the entry into CNT and probable interactions using mechanical principles and mathematical modeling. The atomic interaction between nanotubes and cisplatin was calculated using hybrid-discrete-continuum approximation. In this approximation, cisplatin was taken as a collection of discrete atoms and the CNT was treated as a continuum body of repeating carbon atoms. Non-bonded interaction, suction, and acceptance energies were calculated using the Lennard-Jones (LJ) potential. For nanotube radius of 5.3 Å, cisplatin exhibited maximum suction energy, depending on the orientation of nanotubes as a function of radii.

We performed density functional studies on the noncovalent functionalization of non-Pt-based anticancer drug camptothecin (CPT) on graphene-based nanomaterials and its prototypes, including graphene oxide (GO) [99]. The noncovalent adsorption of CPT induces a significant strain within the nanosheets and the interaction was thermodynamically favored from energetics perspective. In case of GO, surface incorporation of functional groups resulted in significant crumpling along the basal plane and the interaction was mediated by H-bonding rather than π-π stacking. The molecular docking studies of CPT onto Top1 (Figure 7a) showed CPT to be stacked between the Watson Crick AT and GC base pairs and the interaction was mediated via π-π stacking (Figure 7b). For the binding of CPT functionalized graphene and GO with topoisomerase I (top 1) CPT interacts through π stacking with AT and GC base pairs of DNA. The optimum interacting distance of CPT from AT and GC bases was calculated at 3.87 and 3.38 Å, from the central aromatic rings (Figure 7b). The re-rank score of bare CPT drug was calculated as −89.01 a.u. with an H-bond score of −2.53 a.u. as shown in Table 1.

Figure 7.

(a) Secondary structure of Top1 protein with the CPT drug docked within the DNA, (b) interacting distance between CPT and the DNA base pairs of top 1. Reprinted with permission from Ref. [99]. Copyright 2017, Elsevier.

SystemRe-rank score CPTRe-rank score nanosheetH-bond score CPTH-bond score nanosheet
CPT_Top1−89.01−2.53
8 × 8 graphene/CPT docked onto Top1−89.1095.87−2.570.00
8 × 8 GO/CPT docked onto Top1−90.21126.21−2.29−4.44

Table 1.

The re-rank scores and H-bond scores for the best docked conformations of CPT and CPT/8 × 8 graphene, and CPT/8 × 8 GO sheets, respectively.

Likewise, for the docking of CPT/8 × 8 graphene with Top1 (Figure 8a) CPT gets docked between the AT and GC base pairs. However, graphene gets docked along the phosphate backbone of the ds-DNA helix as shown from Figure 8b indicating a strong interaction between the polar phosphate groups of the DNA helix. Compared to the docking of bare CPT drug, presence of graphene stabilizes the intercalation of CPT between the AT and GC base pairs, as observed from the increase in re-rank score values.

Figure 8.

(a) Secondary structure of Top1 with the CPT/8 × 8 graphene sheet docked within the DNA, (b) binding of CPT/8 × 8 graphene sheet with DNA base pairs of top 1. Reprinted with permission from Ref. [99]. Copyright 2017, Elsevier.

The docking of CPT/GO with Top1 as illustrated in Figure 9a, depicts CPT to get docked between AT and GC base pairs of DNA, mediated by π-π stacking interaction similar to that observed for bare CPT and CPT/8 × 8 graphene. However, in the presence of GO, GO undergoes strong interactions with DNA bases and gets docked between the DNA helix and the interaction is stabilized by intermolecular H-bond between polar functional units on the basal plane of GO and DNA nucleobases (Figure 9b). The molecular docking studies on bare CPT and CPT functionalized graphene and GO systems showed that the interaction of CPT with Top1 is mediated by π-stacking interaction between the aromatic rings of CPT and the A and C bases of DNA. In presence of graphene and GO, CPT undergoes a similar trend in adsorption while the graphene and GO nanomaterial gets docked along the phosphate backbone indicating a strong preferential interaction with DNA.

Figure 9.

(a) Secondary structure of Top1 protein with the CPT/8 × 8 GO sheet docked within the DNA, (b) the binding of CPT/8 × 8 GO sheet with DNA base pairs of top 1. Reprinted with permission from Ref. [99]. Copyright 2017, Elsevier.

Boucetta et al. [20] investigated the supramolecular MWCNT-DOX-copolymer complex for anticancer activities. Since DOX, a clinically acclaimed anticancer drug belonging to the family of anthracyclines exhibit fluorescence properties, its uptake and interaction with nanotubes upon administration can be monitored using fluorescent spectrophotometry. The copolymer coated MWCNT formed supramolecular complexes with DOX via π-stacking and revealed enhanced cytotoxicity leading to highly efficient cell killing efficiency. Likewise, Liu et al. reported the use of DOX loaded PEG functionalized CNT for targeted delivery of anticancer drugs in tumor cells [22]. The SWCNT was pre-functionalized with PEG and DOX and a fluorescence probe (fluorescein) was loaded onto the nanotube via π stacking. The loading and subsequent release of DOX were found to be pH dependent; decrease in pH from 9 to 5 showed a decrease in DOX loading. Under acidic conditions (pH 5.5), DOX exhibited increased hydrophilicity and solubility with lysosomes and endosomes, facilitating the release of drug molecules from the nanotube. On decreasing the pH, surface loading of DOX onto nanotube surface lowered and at lower acidic pH, amine group of DOX underwent protonation resulting in increased solubility of DOX molecules.

Advertisement

5. Summary

Carbon nanotubes have proffered as one of the novel functional materials of the 21st century, broadening the theoretical and experimental perspectives in research to explore its novel and intriguing properties. Due to the conjugated π-electron backbone and curvature (properties very similar to fullerene and graphene) they are highly reactive and depending on the size, length and (n, m) indices, the electronic properties can be tuned to fit the desired functionality. Since the synthesis of CNT yields a mixture of both metallic and semiconducting tubes of varying diameter and chirality, separation and purification of nanotubes pose a major problem which restricts the applicability. Secondly, nanotubes are highly hydrophobic and non-dispersible in most of the common aqueous and organic solvents and tend to aggregate in bundles. To improve nanotube dispersibility, surface modification via functionalization is thus a sought-after approach and covalent and noncovalent functionalization methods can reduce the bundling effect and hydrophobicity. Covalent functionalization although renders high stability to the nanotubes, it tends to distort the structural and inherent electronic properties. Noncovalent functionalization, on the other hand, retains the intrinsic properties of the nanotube, as it forms a surface coating on the nanotube sidewall, and facilitates the uptake of drugs, biomolecules, peptides, proteins, DNA, RNA, and genes within the biological systems.

Although CNTs demonstrate practical applicability in all facets of science, be it biology, physics, medicine, nanotechnology, catalysis, or materials science, its long-term implications need to be assessed from the perspective of human health to environmental risks. The long-term fate of CNTs released into the environment depends on the structural, morphological and synthetic treatments [100]. Methods of reducing toxicity in vivo and in vitro can be envisaged via functionalization of CNT. Proper assessment and in-depth study are essential to render nanotubes useful for diverse and environmentally benign applications.

We investigated the potential application of SWCNTs, graphene-based nanomaterials and its prototypes in TB and cancer chemotherapy using conventional DFT methods supported by molecular docking and MD simulation on the nature of interaction of therapeutic drug functionalized SWCNTs/graphene with the binding site of the protein. The functionalization of SWCNTs with therapeutic drugs using covalent and noncovalent schemes were adopted to investigate the drug binding with the nanotube and the stability of the conjugated complexes. DFT results supported by molecular docking and MD simulation helps in contemplating the feasibility of SWCNT-based novel drug delivery in cancer and TB therapy.

References

  1. 1. Geim AK, Novoselov KS. The rise of graphene. Nature Materials. 2007;6:183-191
  2. 2. Novoselov KS, Geim AK, Morozov SV, Jiang D, Zhang Y, Dubonos SV, Grigorieva IV, Firsov AA. Electric field effect in atomically thin carbon films. Science. 2004;306:666-669
  3. 3. Liu L, Guo GY, Jayanthi CS, Wudate SY. Colossal paramagnetic moments in metallic carbon nanotori. Physical Review Letters. 2002;88:217206
  4. 4. Dresselhaus MS, Dresselhaus G, Jorio A. Unusual properties and structures of carbon nanotubes. Annual Review of Materials Research. 2004;34:247
  5. 5. Hersam MC. Progress towards monodisperse single-walled carbon nanotubes. Nature Nanotechnology. 2008;3:387
  6. 6. Meyyappan M. Carbon Nanotubes Science and Applications. Boca Raton: CRC Press, LLC; 2005. p. 104
  7. 7. Ando T. The electronic properties of graphene and carbon nanotubes. NPG Asia Materials. 2009;1:17-21
  8. 8. Tran PA, Zhang L, Webster T. Carbon nanofibers and carbon nanotubes in regenerative medicine. Advanced Drug Delivery Reviews. 2009;61:1097-1114
  9. 9. Reich S et al. Carbon Nanotubes Basic Concepts and Physical Properties. Weinheim: WILEY-VCH, Verleg GmbH & Co. KGaA; 2004
  10. 10. Kim P, Odom TW, Huang JL, Lieber CM. Electronic density of states of atomically resolved single-walled carbon nanotubes: Van Hove singularities and end states. Physical Review Letters. 1999;82:1225
  11. 11. Charlier JC, Lambin P. Electronic structure of carbon nanotubes with chiral symmetry. Physical Review B. 1998;57:R15037-R15039
  12. 12. White CT, Mintmire JW. Density of states reflects diameter in nanotubes. Nature. 1998;394:29-30
  13. 13. Kostarelos K, Lacerda L, Pastorin G, Wu W, Wieckowski S, Luangsivilay J, Godefroy S, Pantarotto D, Briand JP, Muller S, Prato M, Bianco A. Cellular uptake of functionalized carbon nanotubes is independent of functional group and cell type. Nature Nanotechnology. 2007;2:108-113
  14. 14. Veetil JV, Ye K. Tailored carbon nanotubes for tissue engineering applications. Biotechnology Progress. 2009;25:709-721
  15. 15. Harrison BS, Atala A. Carbon nanotube applications for tissue engineering. Biomaterials. 2007;28:344-353
  16. 16. Feazell RP, Nakayama-Ratchford N, Dai H, Lippard SJ. Soluble single-walled carbon nanotubes as longboat delivery systems for platinum(IV) anticancer drug design. Journal of the American Chemical Society. 2007;129:8438-8439
  17. 17. Dhar S, Liu Z, Thomale J, Dai H, Lippard SJ. Targeted Single-Wall carbon nanotube-mediated Pt(IV) Prodrug delivery using Folate as a homing device. Journal of the American Chemical Society. 2008;130:11467-11476
  18. 18. Pastorin G, Wu W, Wieckowski S, Briand J-P, Kostarelos K, Prato M, Bianco A. Double functionalisation of carbon nanotubes for multimodal drug delivery. Chemical Communications. 2006;(11):1182-1184
  19. 19. Liu Z, Sun X, Nakayama RN, Dai H. Supramolecular chemistry on water-soluble carbon nanotubes for drug loading and delivery. ACS Nano. 2007;1:50-56
  20. 20. Ali-Boucetta H, Al-Jamal KT, McCarthy D, Prato M, Bianco A, Kostarelos K. Multiwalled carbon nanotube-doxorubicin supramolecular complexes for cancer therapeutics. Chemical Communications. 2008;(4):459-461
  21. 21. Shukla PK, Mishra PC, Suhai S. Reactions of DNA bases with the anti-cancer nitrogen mustard mechlorethamine: A quantum chemical study. Chemical Physics Letters. 2007;449:323-328
  22. 22. Liu Z, Chen K, Davis C, Sherlock S, Cao Q, Chen X, Dai H. Drug delivery with carbon nanotubes for in vivo cancer treatment. Cancer Research. 2008;68:6652-6660
  23. 23. Gallo M, Favila A, Mitnik DG. DFT studies of functionalized carbon nanotubes and fullerenes as nanovectors for drug delivery of Antitubercular compounds. Chemical Physics Letters. 2007;447:105-109
  24. 24. Saikia N, Deka RC. Theoretical study on pyrazinamide adsorption onto covalently functionalized (5,5) metallic single-walled carbon nanotube. Chemical Physics Letters. 2010;500:65-70
  25. 25. Upadhyayula VKK, Gadhamshetty V. Appreciating the role of carbon nanotube composites in preventing biofouling and promoting biofilms on material surfaces in environmental engineering: A review. Biotechnology Advances. 2010;28:802-816
  26. 26. Schipper ML, Nakayama-Ratchford N, Davis CR, Kam NWS, Chu P, Liu Z, Sun X, Dai H, Gambhir SS. A pilot toxicology study of single-walled carbon nanotubes in a small sample of mice. Nature Nanotechnology. 2008;3:216-221
  27. 27. Cherukuri P, Bachilo SM, Litovsky SH, Weisman RB. Near-infrared fluorescence microscopy of single-walled carbon nanotubes in phagocytic cells. Journal of the American Chemical Society. 2004;126:15638-15639
  28. 28. Yang ST, Guo W, Lin Y, Deng XY, Wang HF, Sun HF, Liu YF, Wang X, Wang W, Chen M, Huang YP, Sun YP. Biodistribution of pristine single-walled carbon nanotubes in vivo. Journal of Physical Chemistry C. 2007;111:17761-17764
  29. 29. Lu X, Tian F, Xu X, Wang N, Zhang Q. A theoretical exploration of the 1,3-dipolar Cycloadditions onto the sidewalls of (n,n) Armchair single-wall carbon nanotubes. Journal of the American Chemical Society. 2003;125:10459-10464
  30. 30. Lu X, Tian F, Zhang Q. The [2+1] cycloadditions of dichlorocarbene, silylene, germylene, and oxycarbonylnitrene onto the sidewall of armchair (5,5) single-wall carbon nanotube. The Journal of Physical Chemistry. B. 2003;107:8388-8391
  31. 31. Long L, Lu X, Tian F, Zhang Q. Hydroboration of C(100) surface, fullerene, and the sidewalls of single-wall carbon nanotubes with borane. The Journal of Organic Chemistry. 2003;68:4495-4498
  32. 32. Pekker S, Salvetat PP, Jakab E, Bonard JM, Forro L. Hydrogenation of carbon nanotubes and graphite in liquid ammonia. The Journal of Physical Chemistry. B. 2001;105:7938-7943
  33. 33. Zhao J, Park H, Han J, LU JP. Electronic properties of carbon nanotubes with covalent sidewall functionalization. The Journal of Physical Chemistry. B. 2004;108:4227-4230
  34. 34. Qin YJ, Shi JH, Wu W, Li XG, Guo ZX, Zhu DB. Concise route to functionalized carbon nanotubes. The Journal of Physical Chemistry. B. 2003;107:12899
  35. 35. Hamwi A, Alvergnat H, Bonnamy S, Béguin F. Fluorination of carbon nanotubes. Carbon. 1997;35:723-728
  36. 36. Niyogi S, Hamon MA, Hu H, Zhao B, Bhowmik P, Sen R, Itkis ME, Haddon RC. Chemistry of single-walled carbon nanotubes. Accounts of Chemical Research. 2002;35:1105-1113
  37. 37. Bianco A, Kostarelos K, Partidos CD, Prato M. Biomedical applications of functionalised carbon nanotubes. Chemical Communications. 2005;(5):571-577
  38. 38. Lu X, Zhang L, Xu X, Wang N, Zhang Q. Can the sidewalls of single-wall carbon nanotubes be ozonized? The Journal of Physical Chemistry. B. 2002;106:2136-2139
  39. 39. Georgakilas V, Kordatos K, Prato M, Guldi DM, Holzinger M, Hirsch A. Organic functionalization of carbon nanotubes. Journal of the American Chemical Society. 2002;124:760-761
  40. 40. Pantarotto D, Partidos CD, Graff R, Hoebeke J, Briand JP, Prato M, Bianco A. Synthesis, structural characterization, and immunological properties of carbon nanotubes functionalized with peptides. Journal of the American Chemical Society. 2003;125:6160-6164
  41. 41. Liu Z, Winters M, Holodniy M, Dai H. siRNA delivery into human T cells and primary cells with carbon-nanotube transporters. Angewandte Chemie, International Edition. 2007;46:2023-2027
  42. 42. Zerda ADL, Zavaleta C, Keren S, Vaithilingam S, Bodapati S, Liu Z, Levi J, Smith BR, Ma TJ, Oralkan O, Cheng Z, Chen X, Dai H, K-Yakub BT, Gambhir SS. Carbon nanotubes as photoacoustic molecular imaging agents in living mice. Nature Nanotechnology. 2008;3:557-562
  43. 43. Liu Z, Cai W, He L, Nakayama N, Chen K, Sun X, Chen X, Dai H. In vivo biodistribution and highly efficient tumour targeting of carbon nanotubes in mice. Nature Nanotechnology. 2007;2:47-52
  44. 44. Saikia N, Deka RC. Theoretical study on pyrazinamide adsorption onto covalently functionalized (5,5) metallic single-walled carbon nanotube. Chemical Physics Letters. 2010;500:65-70
  45. 45. Saikia N, Deka RC. Density functional calculations on adsorption of 2-methyl heptyl isonicotinate antitubercular drug onto functionalized carbon nanotube. Computational & Theoretical Chemistry. 2011;964:257-261
  46. 46. Saikia N, Deka RC. A comparison of the effect of nanotube chirality and electronic properties on the π-π interaction of single-wall carbon nanotubes with pyrazinamide antitubercular drug. International Journal of Quantum Chemistry. 2013;113:1272-1284
  47. 47. Rosca ID, Watari F, Uo M, Akaska T. Oxidation of multiwalled carbon nanotubes by nitric acid. Carbon. 2005;43:3124-3131
  48. 48. Karousis N, Tagmatarchis N, Tasis D. Current progress on the chemical modification of carbon nanotubes. Chemical Reviews. 2010;110:5366-5397
  49. 49. Huang WJ, Fernando S, Lin Y, Zhou B, Allard LF, Sun YP. Preferential solubilization of smaller single-walled carbon nanotubes in sequential functionalization reactions. Langmuir. 2003;19:7084-7088
  50. 50. Carrillo A, Swartz JA, Gamba JM, Kane RS. Noncovalent functionalization of graphite and carbon nanotubes with polymer multilayers and gold nanoparticles. Nano Letters. 2003;3:1437-1440
  51. 51. Chen RJ, Zhang Y, Wang D, Dai H. Noncovalent sidewall functionalization of single-walled carbon nanotubes for protein immobilization. Journal of the American Chemical Society. 2001;123:3838-3839
  52. 52. Prakash R, Superfine R, Washburn S, Falvo MR. Functionalization of carbon nanotubes with proteins and quantum dots in aqueous buffer solutions. Applied Physics Letters. 2006;88:063102-063101
  53. 53. Haddad R, Holzinger M, Maaref A, Cosnier S. Pyrene functionalized single-walled carbon nanotubes as precursors for high performance biosensors. Electrochimica Acta. 2010;55:7800-7803
  54. 54. Yang Z, Chen X, Chen C, Li W, Zhang H, Xu L, Yi B. Noncovalent-wrapped sidewall functionalization of multiwalled carbon nanotubes with polyimide. Polymer Composites. 2007;28:36-41
  55. 55. Nel AE, Mdler L, Velegol D, Xia T, Hoek EMV, Somasundaran P, Klaessig F, Castranova V, Thompson M. Understanding biophysicochemical interactions at the nano-bio interface. Nature Materials. 2009;8:543-547
  56. 56. Ehli C, Oelsner C, Guldi DM, Mateo-Alonso A, Prato M, Schmidt C, Backes C, Hauke F, Hirsch A. Manipulating single-wall carbon nanotubes by chemical doping and charge transfer with perylene dyes. Nature Chemistry. 2009;1:243-249
  57. 57. Roquelet C, Lauret JS, Alain-Rizzo V, Voisin C, Fleurier R, Delarue M, Garrot D, Loiseau A, Roussignol P, Delaire JA, Deleporte E. Π-stacking functionalization of carbon nanotubes through micelle swelling. ChemPhysChem. 2010;11:1667-1672
  58. 58. Orellana W, Correa JD. Noncovalent functionalization of carbon nanotubes and graphene with tetraphenylporphyrins: Stability and optical properties from ab-initio calculations. Journal of Materials Science. 2015;50:898-905
  59. 59. Wei F, Lillehoj PB, Ho C-M. Self-assembled monolayer based electrochemical nucleic acid sensor for vibrio cholerae detection. Pediatric Research. 2010;67:458-468
  60. 60. Abu-Salah KM, Zourob MM, Mouffouk F, Alrokayan SA, Alaamery MA, Ansari AA. DNA-based nanobiosensors as an emerging platform for detection of disease. Sensors. 2015;15:14539-14568
  61. 61. Balasubramanian K, Burghard M. Biosensors based on carbon nanotubes. Analytical and Bioanalytical Chemistry. 2006;385:452-468
  62. 62. Douglas SM, Chou JJ, Shi WM. DNA-nanotube-induced alignment of membrane proteins for NMR structure determination. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:6644-6688
  63. 63. Rajendra J, Baxendale M, Rap LGD, Rodger A. Flow linear Dichroism to probe binding of aromatic molecules and DNA to single-walled carbon nanotubes. Journal of the American Chemical Society. 2004;126:11182
  64. 64. Jung DH, Kim BH, Ko YK, Jung MS, Jung S, Lee SY, Jung HT. Covalent attachment and hybridization of DNA oligonucleotides on patterned single-walled carbon nanotube films. Langmuir. 2004;20:8886-8891
  65. 65. Li S, He P, Dong J, Guo Z, Dai L. DNA-directed self-assembling of carbon nanotubes. Journal of the American Chemical Society. 2005;127:14-15
  66. 66. Das A, Sood AK, Maiti PK, Das M, Varadarajan R, Rao CNR. Binding of nucleobases with single-walled carbon nanotubes: Theory and experiment. Chemical Physics Letters. 2008;453:266
  67. 67. Shukla MK, Dubey M, Zakar E, Namburu R, Czyznikowska Z, Leszcznski J. Interaction of nucleic acid bases with single-walled carbon nanotube. Chemical Physics Letters. 2009;480:269
  68. 68. Fujigaya T, Nakashima N. Non-covalent polymer wrapping of carbon nanotubes and the role of wrapped polymers as functional dispersants. Science and Technology of Advanced Materials. 2015;16:024802
  69. 69. Jilili J, Abdurahman A, Gülseren O, Schwingenschlögl U. Non-covalent functionalization of single wall carbon nanotubes and graphene by a conjugated polymer. Applied Physics Letters. 2014;105:013103-013105
  70. 70. Prasad PV, Kumar A, Pal PC, Sharma RS, Rao DN, Shrivastav TG, Ge R-S. Bird’s eye view on the recent advances in drug delivery systems. Journal of Biomaterials and Nanobiotechnology. 2011;2:544-556
  71. 71. Torchilin VP. Lipid-Core micelles for targeted drug delivery. Current Drug Delivery. 2005;2:319-327
  72. 72. Torchilin VP. Micellar Nanocarriers: Pharmaceutical perspectives. Pharmaceutical Research. 2007;24:1-16
  73. 73. Sawant RR, Torchilin VP. Polymeric micelles: Polyethylene glycol-Phosphatidylethanolamine (PEG-PE) – Based micelles as an example. Methods in Molecular Biology. 2010;624:131-149
  74. 74. Kostarelos K. Rational design and engineering of delivery systems for therapeutics: Biomedical exercises in colloid and surface science. Advances in Colloid and Interface Science. 2003;106:147-168
  75. 75. Pantarotto D, Briand J-P, Prato M, Bianco A. Translocation of bioactive peptides across cell membranes by carbon nanotubes. Chemical Communications. 2004;(1):16-17
  76. 76. Kam NWS, Jessop TC, Wender PA, Dai H. Nanotube molecular transporters: Internalization of carbon nanotube-protein conjugates into mammalian cells. Journal of the American Chemical Society. 2004;126:6850-6851
  77. 77. Pantarotto D, Singh R, McCarthy D, Erhardt M, Briand JP, Prato M, Kostarelos K, Bianco A. Functionalized carbon nanotubes for plasmid DNA gene delivery. Angewandte Chemie (International Ed. in English). 2004;43:5242
  78. 78. Wu W, Wieckowski S, Pastorin G, Benincasa M, Klumpp C, Briand J-P, Gennaro R, Prato M, Bianco A. Targeted delivery of amphotericin B to cells by using functionalized carbon nanotubes. Angewandte Chemie, International Edition. 2005;44:6358-6362
  79. 79. Benincasa M, Pacor S, Wu W, Prato M, Bianco A, Gennaro R. Antifungal activity of amphotericin B conjugated to carbon nanotubes. ACS Nano. 2011;5:199-208
  80. 80. Gu L, Elkin T, Jiang X, Li H, Lin Y, Qu L, Tzeng T-RJ, Joseph R, Sun Y-P. Single-walled carbon nanotubes displaying multivalent ligands for capturing pathogens. Chemical Communications. 2005;(7):874-876
  81. 81. Brady-Estévez AS, Kang S, Elimelech M. A single-walled-carbon-nanotube filter for removal of viral and bacterial pathogens. Small. 2008;4:481-484
  82. 82. Welsher K, Liu Z, Daranciang D, Dai H. Selective probing and imaging of cells with single walled carbonnanotubes as near-infrared fluorescent molecules. Nano Letters. 2008;8:586-590
  83. 83. Liu ZA, Li X, Tabakman SM, Jiang K, Fan S, Dai H. Multiplexed multicolor Raman imaging of live cells with isotopically modified single walled carbonnanotubes. Journal of the American Chemical Society. 2008;130:13540-13541
  84. 84. Hadjiev VG, Arepalli S, Nikolaev P, Jandl S, Yowell L. Enhanced Raman microprobe imaging of single-wall carbonnanotubes. Nanotechnology. 2004;15:562-567
  85. 85. Available from: http://nobelprize.org/nobel_prizes/medicine/laureates/1905/koch.html
  86. 86. Glaziou P, Floyd K, Raviglione M. Global burden and epidemiology of tuberculosis. Clinics in Chest Medicine. 2009;30:621-636
  87. 87. Available from: http://www.searo.who.int/tb/documents/annual_tb_repot_2017/en/
  88. 88. Schmit KM, Wansaula Z, Pratt R, Price SF, Langer AJ. Tuberculosis-United States, 2016. MMWR. Morbidity and Mortality Weekly Report. 2017;66:289-294
  89. 89. Cegielski JP. Extensively drug-resistant tuberculosis: There must be some kind of way out of here. Clinical Infectious Diseases. 2010;50:S195-S200
  90. 90. WHO. Anti-tuberculosis Drug Resistance in the World: The WHO/IUATLD Global Project on Anti-tuberculosis Drug Resistance Surveillance 1994-1997, WHO/TB/97.229; 1997
  91. 91. Cole ST, Riccardi G. New tuberculosis drugs on the horizon. Current Opinion in Microbiology. 2011;14:570
  92. 92. Petrella S, Gelus-Ziental N, Maudry A, Laurans C, Boudjelloul R, Sougakoff W. Crystal structure of the Pyrazinamidase of mycobacterium tuberculosis: Insights into natural and acquired resistance to pyrazinamide. PLoS One. 2011;6:e15785
  93. 93. Saikia N, Rajkhowa S, Deka RC. Density functional and molecular docking studies towards investigating the role of single-wall carbon nanotubes as nanocarrier for loading and delivery of pyrazinamide antitubercular drug onto pncA protein. Journal of Computer-Aided Molecular Design. 2013;27:257-276
  94. 94. Saikia N, Jha AN, Deka RC. Interaction of pyrazinamide drug functionalized carbon and boron nitride nanotubes with pncA protein: A molecular dynamics and density functional approach. RSC Advances. 2013;3:15102-15107
  95. 95. Zanella I, Fagan SB, Mota R, Fazzio A. Ab initio study of pristine and Si-doped capped carbon nanotubes interacting with nimesulide molecules. Chemical Physics Letters. 2007;439:348-353
  96. 96. Liu H, Bu Y, Mi Y, Wang Y. Interaction site preference between carbon nanotube and nifedipine: A combined density functional theory and classical molecular dynamics study. Journal of Molecular Structure (THEOCHEM). 2009;901:163
  97. 97. Burger KNJ, Staffhorst RWHM, De Vijlder HC, Velinova MJ, Bomans PH, Frederir PM, De Kruijff B. Nanocapsules: Lipid-coated aggregates of cisplatin with high cytotoxicity. Nature Medicine. 2002;8:81-84
  98. 98. Hilder TA, Hill JM. Modeling the encapsulation of the anticancer drug cisplatin into carbon nanotubes. Nanotechnology. 2007;18:1
  99. 99. Saikia N, Deka RC. Ab initio study on the noncovalent adsorption of Camptothecin anticancer drug onto graphene, defect modified graphene and graphene oxide. Journal of Computer-Aided Molecular Design. 2013;27:807-821
  100. 100. Helland A, Wick P, Koehler A, Schmid K, Som C. Reviewing the environmental and human health Knowledge Base of carbon nanotubes. Environmental Health Perspectives. 2007;115:1125

Written By

Nabanita Saikia

Submitted: 25 June 2017 Reviewed: 23 October 2017 Published: 20 December 2017