You are on page 1of 12

Advanced Drug Delivery Reviews 63 (2011) 1340–1351

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / a d d r

Polymeric nanohybrids and functionalized carbon nanotubes as drug delivery


carriers for cancer therapy☆
Satya Prakash ⁎, Meenakshi Malhotra, Wei Shao, Catherine Tomaro-Duchesneau, Sana Abbasi
Biomedical Technology and Cell Therapy Research Laboratory, Departments of Biomedical Engineering and Physiology, Artificial Cells and Organs Research Center, Faculty of Medicine,
McGill University, 3775 University Street, Montreal, Quebec, Canada H3A 2B4

a r t i c l e i n f o a b s t r a c t

Article history: The scope of nanotechnology to develop target specific carriers to achieve higher therapeutic efficacy is
Received 25 September 2010 gaining importance in the pharmaceutical and other industries. Specifically, the emergence of nanohybrid
Accepted 27 June 2011 materials is posed to edge over chemotherapy and radiation therapy as cancer therapeutics. This is primarily
Available online 3 July 2011
because nanohybrid materials engage controlled production parameters in the making of engineered particles
with specific size, shape, and other essential properties. It is widely expressed that these materials will
Keywords:
Nanohybrids
significantly contribute to the next generation of medical care technology and pharmaceuticals in areas of
Polymeric nanoparticles disease diagnosis, disease prevention and many other treatment procedures. This review focuses on the
Carbon nanotubes currently used nanohybrid materials, polymeric nanoparticles and nanotubes, which show great potential as
Drug delivery effective drug delivery systems for cancer therapy, as they can be grafted with cell-specific receptors and
Microcapsule intracellular targeting molecules for the targeted delivery of therapeutics. Specifically, this article focuses on
Cancer therapy the current status, recent advancements, potentials and limitations of polymeric nanohybrids and
functionalized carbon nanotubes as drug delivery carriers.
Crown Copyright © 2011 Published by Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1341
2. Nanohybrid drug delivery carriers for cancer therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1341
2.1. Polymeric nanohybrid materials as drug delivery systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1342
2.1.1. Pharmacokinetics and biodistribution of polymeric nanohybrid materials . . . . . . . . . . . . . . . . . . . . . . . . . 1342
2.1.2. Long circulating polymeric nanohybrids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1343
2.2. Synthesis of polymeric nanohybrid materials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1343
2.2.1. PACA nanohybrids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1343
2.2.2. PLGA/PLA nanohybrids. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1344
2.2.3. Polymeric nano micelles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1344
3. Carbon nanotubes as new drug delivery carriers for use in cancer therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1345
3.1. Functionalization of carbon nanotubes for drug delivery and other applications . . . . . . . . . . . . . . . . . . . . . . . . . . 1345
3.2. Applications of carbon nanotubes in cancer therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1346
3.2.1. Small hydrophobic cancer drug delivery system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1346
3.2.2. Microencapsulated carbon nanotube delivery devices for cancer therapy . . . . . . . . . . . . . . . . . . . . . . . . . 1347
3.2.3. Selective tumor destruction via Near-Infrared radiation of carbon nanotubes . . . . . . . . . . . . . . . . . . . . . . . 1347
3.2.4. In vivo toxicity of carbon nanotubes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1347
4. Conclusions: toward future perspective, benefits and concerns . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1347
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1348
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1348

☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “Hybrid Nanostructures for Diagnostics and Therapeutics”.
⁎ Corresponding author. Tel.: + 1 514 398 3676; fax: + 1 514 398 7461.
E-mail address: satya.prakash@mcgill.ca (S. Prakash).

0169-409X/$ – see front matter. Crown Copyright © 2011 Published by Elsevier B.V. All rights reserved.
doi:10.1016/j.addr.2011.06.013
S. Prakash et al. / Advanced Drug Delivery Reviews 63 (2011) 1340–1351 1341

1. Introduction compounds, due to their inability to completely discriminate between


tumor and healthy tissues, have a high toxicity [37].
Currently available technologies have made enormous advance- It can be said that, as of yet, for a number of reasons, an adequate
ment in cancer research, but an adequate therapy remains elusive. therapy for cancer has not been developed. Some of these reasons
Cancer is characterized as a disorder which arises following a number include: late stage diagnosis, inadequate methods for controlling
of mutagenesis steps, allowing cancerous cells to grow and divide aggressive metastasis, and a lack of methods to overcome multi-drug
uncontrollably. These processes allow cancer cells to acquire resistant (MDR) cancer. The current medical anticancer treatments
properties of unlimited proliferation potential, self-sufficiency in involve applications where therapies are provided by removing the
growth signals and resistance to both anti-proliferative and apoptotic diseased cells (surgery), burning them out (radiation therapy), or
cues which would, otherwise, contain their growth [1]. Tumors have poisoning the diseased cells faster than the healthy cells (chemo-
also evolved methods to gain further support through interactions therapy), all of which do cause damage to healthy cells [1,38–40].
with surrounding stromal cells, promoting their angiogenesis, their Nanotechnology, involving the development of an appropriate
evasion of immune detection and their metastasis to distal organs [2]. delivery system, is an emerging field of research that attempts to
The Global Cancer Report issued by the World Health Organization overcome the challenges associated with designing an anti-cancer
(WHO) estimates that there are over 10 million new cases of cancer therapy. A number of compounds have been formulated using
every year with over 6 million annual deaths caused by the disease nanotechnology methods and combinations of polymers with anti-
[3]. The most frequent types of cancers causing death are, as listed by cancer drugs to allow for the development of a delivery system.
the WHO: lung, stomach, liver, colon and breast cancers [4]. Table 2 lists some of these anticancer therapeutic formulations and
Current cancer therapies can be broadly categorized into two their current clinical status. The benefits of a nanoscale drug delivery
groups: cytotoxic therapies and molecular targeted drugs [5]. system are numerous, and can involve, but are not limited to: 1) a
Examples of traditional cytotoxic therapies include radiation and targeted delivery, allowing for an increased drug concentration at the
chemotherapeutic compounds such as platinum-based drugs [5,6]. desired site, reducing systemic exposure to a potentially toxic
Radiation, utilizing photons, damages the DNA of cancer cells by compound 2) a constant rate of drug delivery to allow for the
creating free radicals, leading to the inhibition of cell division and, maintenance of a constant therapeutic dose at the site of delivery and
eventually, causing cell death. Cancer cells are most abundantly found 3) an increase in drug stability due to protection from degradation and
in the mitotic phase, in a state of continuous DNA replication, allowing loss of drug [37,41].
radiation to be more toxic to tumor cells than normal cells, providing a Recently, nanohybrids and carbon nanotubes (CNTs) have been
therapeutic window [7]. Platinum-based drugs, such as cisplatin, act proposed as drug delivery carriers. Nanohybrids combine biological or
through the formation of DNA-platinum adducts [8]. These adducts bio-functionalized molecules giving rise to a system capable of drug
lead to DNA deformations, such as unwinding and bending, while also delivery. CNTs, on the other hand, are synthetic nanomaterials, made
being recognized by a number of cellular proteins involved in DNA- from carbon atoms, which can be functionalized to act as a drug
repair pathways [9]. The formation of these adducts ultimately results delivery system. This review focuses on polymeric nanohybrid
in apoptosis, although the mechanism of action has not been fully materials and carbon nanotubes as drug delivery systems, specifically
defined [8]. focusing on their synthesis, their functionalization and their applica-
The compounds listed in Table 1 rely mainly on one important tions, with an emphasis toward cancer therapy.
characteristic of cancer cells: their rapid and uncontrolled proliferation
and division. Hence, the compounds do show a level of toxicity to non- 2. Nanohybrid drug delivery carriers for cancer therapy
cancerous cells since these are still proliferating, albeit at a slower rate
than cancerous cells. There are also a number of compounds currently The tumor microenvironment comprises fast-growing, hyperpro-
undergoing clinical trials, but most of these still rely on the proliferation liferative cancer cells having a high metabolic rate and demand for the
properties of cancerous cells, attacking the cell division and apoptosis recruitment of new vessels (neovascularization) in order to supply
pathways [36]. It is, hence, evident that the newly developed them with oxygen and nutrients [54]. The pathophysiologic condition
compounds, which still rely on these properties, will also give rise to of a tumor is its utilization of the glycolysis pathway to obtain
serious side effects such as nausea, hair loss, neuropathies, neutropenia additional energy, resulting in the generation of an acidic environ-
and kidney failure [37]. This is simply due to the fact that these ment [55]. Tumor cells also release growth factors and enzymes such

Table 1
Anti-cancer therapeutics and their mechanism of action.

Compound Mechanism of action Ref.

Amethopterin/methotrexate Inhibits dihydrofolate reductase, blocking thymidine synthesis and, in turn, DNA [10,11]
and protein synthesis
Cisplatin/carboplatin/tetraplatin/oxaliplatin Platinum-based compounds, adduct formation leading to DNA cross linking, [8,12,13]
inducing apoptosis
Daunorubicin/adriamycin/doxorubicin/mitoxanthrone/actinomycin/idarubicin DNA intercalating compounds, blocking polymerase activity, inhibiting DNA [14–16]
replication, inducing apoptosis
Etoposide Forms a ternary complex with topoisomerase II and DNA, leading to errors in [17,18]
synthesis, inducing apoptosis
Fluorouracil Pyrimidine analog, inhibiting DNA synthesis, induces p53-dependent apoptosis [19–21]
Hydroxyurea Inhibits ribonucleoside reductase, inhibiting DNA synthesis, inducing apoptosis [22–24]
Mercaptopurine Purine analog, inhibiting DNA synthesis, inducing apoptosis [25,26]
Paclitaxel/ Taxol/ Docetaxel Binds β-tubulin, forming highly stable microtubules that resist depolymerization, [27–29]
preventing cell division and inducing apoptosis
Tamoxifen Selective estrogen response modifier (SERM), used against estrogen-sensitive [30,31]
breast tumors, competitively inhibits estrogen binding, slowing cell proliferation
Vinblastine/vincristine/vindesine Binds tubulin, inducing self-association and depolymerizes pre-existing [32,33]
microtubules, inducing apoptosis
Camptothecin inhibits the catalytic activity of mammalian DNA topoisomerase I, inhibiting DNA [34,35]
synthesis and inducing DNA strand breaks, leading to apoptosis
1342 S. Prakash et al. / Advanced Drug Delivery Reviews 63 (2011) 1340–1351

as matrix metalloproteinases, which lead to an imbalance of immunological barriers of the body. The use of nanovector drug delivery
angiogenic regulators dilating the tumor vessels, resulting in large vehicles has gained importance in biomedical applications, as they
gap junctions, ranging from 200 to 2000 nm in size, between enable the encapsulation and the successful delivery of drugs with poor
endothelial cells [54]. Moreover, the higher interstitial pressure aqueous solubility profiles such as paclitaxel, an antitumor agent [69–
generated by a compromised lymphatic drainage and a lower intra- 71]. Paclitaxel bound to albumin nanoparticles is an FDA approved
vascular pressure limit the movement of macromolecules/particulate nanoparticle formulation, under the market name Abraxane, for
materials out of the tumor blood vessels into the extra-vascular delivery to metastatic breast cancer patients [72,73]. Another advantage
compartment [56]. This enhanced vascular permeability and the lack of utilizing polymeric nanovectors is the potential for non-invasive
of adequate lymphatic drainage at tumor sites facilitate passive targeting to the tumor. Nanohybrid materials exhibit multifunctional
targeting using polymeric nanovectors. The phenomenon is termed features that facilitate imaging, targeting and drug delivery. Other
the Enhanced Permeability and Retention effect (EPR) [57–59](Fig. 1). polymeric nanovector composites that have received much attention in
This effect is also attributed to the molecular weight, surface charge cancer drug delivery are polylactide–polyglycolide copolymers entrap-
and nature of the polymer. The conjugation of drugs to polymers ping leutinizing hormone releasing hormone (LHRH), marketed as
avoids the random bioavailability of the low molecular weight drugs goserelin (Zoladex) and leuprolide (Leuprone Depot) [74,75] and
and enables target specificity. However, the molecular weight of the liposomes encapsulating daunorubicin and doxorubicin, marketed as
nanovectors also plays an equally important role in the delivery, as DaunoXome and Doxil/Caelyx respectively [76,77]. Other polymers
nanovectors of a molecular weight of less than 50 kDa or of a size less include N-(2-hydroxyl propyl)methacrylamide (HPMA) copolymers,
than 6 nm are rapidly cleared by the kidney following systemic polyglycolic acid (PGA) with paclitaxel, marketed as Xyotax™ [78,79]
administration [60–63]. On the other hand, the size is crucial for non- and polycaprolactones and natural polymers like albumin, gelatin,
biodegradable polymers, in order to be eliminated by the renal alginate, collagen and chitosan. A detailed overview on clinically
system, following drug delivery. In general, cationic nanovectors are evaluated polymer-drug conjugates has been well documented else-
more susceptible to glomerular filtration than anionic nanovectors, as where [67,80].
the negative charge on the glomerular capillary results in a charge-
selective barrier and the nanovectors of larger molecular weight
accumulate in the liver, kidneys, and lungs [64–66]. Thus, the ideal 2.1.1. Pharmacokinetics and biodistribution of polymeric nanohybrid
molecular weight of the polymer depends on the particular materials
application, but ranges from 30 kDa to 100 kDa. Nanoparticles that The drug polymer conjugates appear to improve a drug's half-life in
abide the size and surface characteristics mentioned above, escape the plasma due to the larger hydrodynamic volume of polymers which, in
Reticulo-Endothelial System (RES) and are able to circulate longer in turn, increases the drug's accumulation at the tumor site. For example,
the bloodstream with a greater chance of reaching the targeted tumor PHPMA copolymer conjugated with doxorubicin has shown a drug
tissues. Drugs conjugated to polymers through peptidyl and ester stability of 5 min in plasma when administered alone, compared to 1 h
linkages allow for a controlled drug release. In some cases, the drug is when administered with polymer which also shows an increased drug
released through thiol dependent cleavage by protease cathepsin B accumulation in solid tumors [81–83]. Antibodies [84,85], peptides [86]
[67]. Other drug release methods involve pH sensitive cis-aconityl, and saccharides [48] have also been attached to PHPMA copolymer.
hydrazone and acetyl linkages [68]. The hydrolysis cleaves the linkage Galactosamine has been attached as a targeting moiety to the HPMA-
within the compartment of the endosomes and lysosomes, which doxorubicin, targeting the asialoglycoprotein receptor present in
have an acidic pH ranging from 4.0 to 6.5. hepatocytes [48]. Another example is the styrene maleic anhydride
conjugated with the anti-tumor protein, neocarzinostatin (SMANCS)
2.1. Polymeric nanohybrid materials as drug delivery systems used for the treatment of hepatocellular carcinoma [87]. Surface
modifications by various ligands and antibodies such as transferrin,
Polymeric nanohybrid materials comprise a core material, a folate, epidermal growth factor, and arginine–glycine–aspartic acid
therapeutic “payload” and a biological surface modification that aids (RGD) tripeptide conjugated to the polymer backbone, allow for the
in the biodistribution and selective cell targeting moieties, Fig. 2. The active targeting of the therapeutic to tumor cells, via receptor-mediated
nanohybrid materials/nanovectors in conjunction with drugs are mostly endocytosis [88–95]. Usually it is favorable to use non-cationic or
delivered intravenously, as they bear the key characteristic of their surface-modified polymers to prevent the adsorption of plasma
ability to be tailored to bypass the biological/physiological and proteins onto the surface of the nanoparticles, in order to avoid uptake

Table 2
List of polymer–drug conjugate compounds and their clinical status.

Compound Clinical name Clinical Polymer Drug Ref.


status

Albumin-Taxol Abraxane® Market Albumin Paclitaxel [42,43]


CNT-MTX N/A Preclinical Carbon nanotube Methotrexate [44]
HPMA copolymer doxorubicin PK1, FCE28068 Phase III N-(2-hydroxypropyl) methacrylamide (HPMA) Doxorubicin [45]
copolymer
HPMA copolymer-carboplatin AP5280 Phase II N-(2-hydroxypropyl) methacrylamide (HPMA) Platinum [46]
platinate copolymer
HPMA copolymer-DACH- AP5346, ProLindac™ Phase II N-(2-hydroxypropyl) methacrylamide (HPMA) Platinum [47]
platinate copolymer
HPMA copolymer-doxorubicin- PK2, FCE28069 Phase II N-(2-hydroxypropyl) methacrylamide (HPMA) Doxorubicin and [48]
galactosamine copolymer Galactosamine
PEG-PAA-DOX NK911 Phase III Poly(ethylene glycol) Doxorubicin [49,50]
PEG-PLA-Taxol Genexol-PM Phase III Poly(ethylene glycol) Paclitaxel [51]
Polyglutamate-camptothecin CT-2106 Phase I/II Poly-L-glutamic acid (PGA) Camptothecin [52]
Polyglutamate-paclitaxel CT-2103, Xyotax™, Phase III Poly-L-glutamic acid (PGA) Paclitaxel [53]
OPAXIO™
S. Prakash et al. / Advanced Drug Delivery Reviews 63 (2011) 1340–1351 1343

by the RES when administering nanoparticles via a systemic route of nonsolvent-containing surfactant, leading to the formation of mono-
administration. mer-swollen micelles and stabilized monomer droplets. Similar to DP,
the polymerization is performed in the presence of an initiator,
generating radicals or ions, nucleating the monomeric units to initiate
2.1.2. Long circulating polymeric nanohybrids
the polymerization process. This process can be performed either in
Hydrophilic polymers with neutral charge like polyethylene glycol
organic or aqueous media as the continuous phase [105]. The
(PEG) or polyethylene oxide (PEO) have been extensively used for
following sections elaborate on some of the nanohybrid materials
surface modification to increase the systemic retention and circulation
developed using the above two (EP or DP) methods of synthesis.
time of nanovectors [96]. Incorporation of PEG imparts a stealth
characteristic to the polymers, reducing the hydrophobic interactions
with the RES. The hydrophilicity is attributed to the hydrogen bonding
2.2.1. PACA nanohybrids
of water molecules with the ether oxygen of PEG, creating a hydrating
The synthesis of Poly(alkyl cyanoacrylate) (PACA) nanoparticles
shell around the nanovector [97]. This further increases the hydrody-
involves EP in the continuous organic phase and results in the
namic size of the polymeric nanovector, decreasing its clearance via the
formation of nanocapsules (shell-like structure, encapsulating a liquid
RES [98,99]. The molecular weight and molar ratio of PEG used impart a
cavity) [106]. In this case, the drug is dissolved in an aqueous phase
steric character to the nanovector. A recent review on PEG conjugates
such as iso-octane, cyclohexane–chloroform, isopropyl myristate–
with drugs for anti-cancer therapy is described elsewhere [100]. PEO has
butanol, and hexane, along with surfactants, resulting in a
been used to modify a Poly(3-caprolactone) (PCL) nanovector to deliver
microemulsion with water-swollen micelles containing the drug. On
therapeutic agents, such as paclitaxel. The modification resulted in the
the other hand, EP in an aqueous continuous phase is used for the
prolonged circulation of the nanovector to 25 h and exhibited a greater
polymerization of monomers like alkylcyanoacrylates but involves
accumulation in the tumor region, as compared to the administration of
low quantities of surfactants. It is only used to stabilize the newly
free drug [101]. Other hydrophilic polymers used for surface modifica-
formed polymer particles. Poly(ethyl cyanoacrylate) (PEC) and poly
tion include polyvinyl alcohol, polyacryl amide, polyvinyl pyrrolidone,
(isobutyl cyanoacrylate) (PIBC) nanospheres containing metaclopra-
polysorbate-80, and block co-polymers such as poloxomer (Pluronicw)
mide have been prepared using this technique. However, it does not
and poloxamine (Tetronicw) [102–104].
facilitate efficient drug loading as much as the former method. The use
of stabilizers/emulsifiers has a significant role in controlling the size of
2.2. Synthesis of polymeric nanohybrid materials the polymer particles. It has been observed that high concentrations of
poloxamer 188, i.e. N2%, reduced the particle size of poly(isobutyl
Synthetic polymers are developed by two processes; dispersion cyanoacrylate) (PIBC) nanoparticles from around 200 nm to 31–
polymerization (DP) and emulsion polymerization (EP). In the 56 nm [107]. Poly(butyl cyanoacrylate) (PBCA) nanovectors of n-
process of DP, chains of monomer are formed in an aqueous butyl cyanoacrylate containing methotrexate were prepared by DP
continuous phase with the help of an initiator, and precipitate in an and EP methods using dextran and polaxamer 188 as stabilizers
insoluble phase, where stabilization occurs with a polymeric respectively [108]. Nanovectors prepared by DP showed a higher zeta
stabilizer. If the initiator generates ions in inducing the polymeriza- potential and a higher rate of drug release when compared to those
tion process from a monomer, the mechanism is termed “ionic prepared by EP. The contrast in the drug release profile is attributed to
polymerization”, which can result in the formation of cationic or the channelizing effect of dextran as a stabilizer.
anionic polymers. If a radical is involved in nucleating the monomer, Drug loading to PACA nanoparticles is primarily performed during
the mechanism is termed “radical polymerization.” Another method the polymerization process or by absorption onto preformed particles.
of synthesis is the EP process wherein the monomer is emulsified in a In the former case, this can lead to the covalent coupling of the drug to

Fig. 1. Schematic of tumor targeting by nanohybrids via Enhanced Permeation and Retention (EPR) effect.
1344 S. Prakash et al. / Advanced Drug Delivery Reviews 63 (2011) 1340–1351

the polymer. The type of drug loading determines the carrier capacity, regression and higher survival rates in the treated animals [126]. It was
i.e. the percentage of the drug associated with a given amount of observed that the IC50 for paclitaxel with transferrin conjugated
nanoparticles, when compared to the initial concentration of the drug nanovector particles was 5 fold lower than that obtained with
[109,110] Some examples of drugs adsorbed on the surface of PACA unconjugated nanovectors. Another study demonstrated the enhanced
nanoparticles are betaxololchlorhydrate, [111] hematoporphyrin intracellular delivery of PLGA nanovectors, surface-coated with cationic
[112] and primaquine [113], and the ones that were covalently di-block copolymer, poly(L-lysine)–poly(ethylene glycol)–folate
bound during the synthesis process are doxorubicin [114] and (PLL–PEG–FOL) in KB cells, overexpressing folate receptors [127]. In
actinomycin D. It has been observed that doxorubicin-loaded poly another similar study, paclitaxel-loaded PLGA nanoparticles were
(alkyl cyanoacrylate) (PACA) nanovectors can overcome multidrug- conjugated to wheat germ agglutinin (WGA) and demonstrated a
resistance-1-type efflux pumps expressed by tumor cells [115]. greater anti-proliferation activity in A549 and H1299 cells due to a
The drug release kinetics greatly depends on the degradation rate targeted receptor-mediated endocytosis [128]. Another therapeutic
of the polymer and the type of drug binding to the polymer [109,116]. agent encapsulated in PLGA nanovectors was cystatin which inhibits
The interaction between PACA-doxorubicin conjugates occurs via tumor-associated activity of intracellular cysteine protease cathepsins B
lipophilic, oligomeric units of PACAs that encapsulate the amphiphilic and L. The cystatin-loaded PLGA nanovector formulation exhibited a
doxorubicin during the polymerization process. This conjugation is 160-fold greater cytotoxic effect in MCF-10A neoT cells than the free
driven by hydrogen bonds formed between the N and H functions of drug [129]. The anticancer agent doxorubicin has been delivered via
doxorubicin and the cyano groups of alkylcyanoacrylate. The collective PLGA nanovectors by a chemical conjugation through an ester linkage to
force of dipole–charge interaction, H-bonds, and hydrophobic forces PLGA polymer, and showed a sustained drug release over a month, an
leads to the cohesion of the drug–polymer conjugate. Another drug increased drug uptake in the HepG2 cell line and exhibited a slightly
that integrates while in the polymerization process is 5-fluorouracil lower IC50 value compared to that of free doxorubicin [130].
(5-FU) [117]. However, it may interfere with the initiation process of A recent study reported lipid–polymer nanohybrids, comprising
the polymerization, as the amino groups of 5-FU form zwitterions at an PLGA as a hydrophobic polymeric core encapsulating the hydrophobic
acidic pH of 2–3. Another concept of targeting tumor cells was drug docetaxel (Dtxl). This polymer–drug assembly was encapsulated
developed in which a copolymer poly[aminopoly(ethylene glycol) in a lipid layer of lecithin, which was further covalently conjugated to
cyanoacrylate-co-hexadecyl cyanoacrylate] [poly(H2NPEGCA-co- 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE)-PEG as a
HDCA)] was synthesized via nanoprecipitation and conjugated to hydrophilic polymer [131]. The lipid–polymer nanohybrids yielded
folic acid via the amino terminal groups on PEG [118]. high drug loading and sustained drug release profiles. Another similar
study demonstrated a paclitaxel carrying lipid–polymer nanohybrid
2.2.2. PLGA/PLA nanohybrids conjugated to anti-carcinoma embryonic antigen (CEA) half antibody
Polylactides (PLA) and poly (D,L-lactide-coglycolide) (PLGA) have to target pancreatic cancer [132].
been extensively investigated for drug delivery applications because of
their biodegradability via hydrolytic cleavage of the ester linkage 2.2.3. Polymeric nano micelles
[119,120], because of their biocompatibility and the already present FDA Polymeric micelles are another class of nanovectors and have
approval for use in humans as resorbable sutures, bone implants and gained much attention for encapsulating and delivering hydrophobic
screws, contraceptive implants [121,122], as scaffolds in tissue engi- drugs. The driving forces of polymeric chains to form micelles are the
neering and as graft materials [123–125]. PLGA/PLA hybrid nanovectors hydrophobic, electrostatic, π–π and hydrogen bonding interactions.
have been utilized for the delivery of anti-cancer agents and other Polymeric micelles composed of poly(ethylene oxide–aspartate)
therapeutic agents [119]. Paclitaxel-loaded PLGA nanovectors conjugat- block copolymer conjugated to an anti cancer drug, doxorubicin,
ed to transferrin were administered to animal models of prostate exhibited a sustained systemic circulation [133], reduced uptake by
carcinoma via intratumoral injection. An enhanced cellular uptake was the RES and a higher accumulation in a tumor bearing mouse model of
observed by these targeted nanoparticles, resulting in complete tumor Colon-26 [134]. Polymeric micelles have also been employed in active

Fig. 2. Schematic of multifunctional polymeric nanohybrid devices for targeted drug delivery.
S. Prakash et al. / Advanced Drug Delivery Reviews 63 (2011) 1340–1351 1345

nanovectors were used to deliver ceramide with paclitaxel, in


combination to overcome MDR, especially in cases of breast and
ovarian cancers. The co-treatment resulted in enhanced cell death
when compared to the drugs alone and exhibited a 100-fold increase
in chemosensitivity and overcoming of MDR via combination therapy.

3. Carbon nanotubes as new drug delivery carriers for use in


cancer therapy

Carbon nanotubes (CNTs) are synthetic nanomaterials made from


carbon and belong to the family of fullerene. Structurally, carbon
nanotubes can be pictured as rolled sheets of graphene rings built
Fig. 3. Functionalization of carbon nanotubes via 1,3-dipolar cycloadditions. The 1,3-
dipolar cycloaddition of azomethine ylides on CNT is generated by in-situ thermal from sp 2 hybridized carbon atoms into hollow tubes. There are two
condensation of aldehydes and α-amino acids. Azomethine ylides are very reactive categories of carbon nanotubes, single-walled carbon nanotubes
intermediates and attack efficiently the p-system of the CNT causing large number of (SWNTs) and multi-walled carbon nanotubes (MWNTs). SWNTs
pyrrolidine rings fused to the CNT side wall, which helps in solubilisation of CNTs [135].
contain one layer of graphene sheet with a diameter of 1–2 nm with a
length ranging from 50 to several hundred nanometers, whereas
targeting applications, wherein the polymeric micelles were prepared MWNTs are co-axially arranged multiple layers of SWNTs, positioned
from thermo- [135,136] or pH-sensitive [137] block co-polymers. An within one another with a diameter ranging from 5 to 100 nm [144].
example of a polymeric micelle with pH sensitive solubility properties There are various techniques to produce CNTs. The three main
was synthesized by conjugating 4,40-trimethylenedipiperidine with techniques are 1) electric arc discharge [145], 2) laser ablation [146]
1,4-butanediol diacrylate to form a biodegradable, hydrophobic poly and 3) chemical vapor deposition [147]. These methods involve
(b-amino ester) (PBAE) polymer. The nanovectors prepared from this synthesis at high temperature, pressure and the use of reaction
polymer were loaded with paclitaxel (150–200 nm) and were catalysts, leading to fine structures of CNTs along with some synthesis
modified with Pluronic F-108 (poloxamer 407), a triblock copolymer induced impurities like graphitic debris and catalytic particles.
of polyethylene oxide/polypropylene oxide/polyethylene oxide Various methods such as oxidation, acid treatment, chromatography,
(PEO/PPO/PEO). The PPO attached to the hydrophobic surface of the filtration and functionalization are involved for purification [148]. A
nanovectors, and the hydrophilic PEO contributed to the stealth recent review on CNTs and their applications describes various other
properties of the polymeric nanovectors. processing and purification techniques [149]. Compared to other
The pH-sensitive nature of the PBAE nanovectors has been tested nanomaterials, carbon nanotubes have an ultrahigh surface area ratio
at various pH conditions ranging from 5.0 to 7.4 and was found to and distinct optical properties, such as, high absorption in the near-
degrade at a pH below 6.5, thereby releasing their contents on infrared (NIR) range and a strong Raman shift, which make them
encountering the acidic tumor microenvironment and in the endo- advantageous for biomedical applications linked to detection and
somes and lysosomes of the cells following internalization; a imaging [150]. CNTs are hydrophobic in nature and thus insoluble in
condition ideal in anti-cancer drug delivery. The in vitro studies water, which limits their application in biomedical and medicinal
with PEO–PBAE nanovectors loaded with paclitaxel were performed chemistry. Therefore, various functionalization methods like adsorp-
on human breast adenocarcinoma cells (MDAMB231) [138,139]. The tion, electrostatic interaction and covalent bonding are being utilized
surface modification using a triblock copolymer (PEO/PPO/PEO) with a number of compounds and polymers to render a hydrophilic
enhanced the half life of the nanovectors with a residence circulation character to CNTs so as to avoid their aggregation and to facilitate
time of 21 h compared to 10 min for the unmodified nanovectors. It their use in biomedical applications. Recent developments with CNTs
also improved the drug concentration from 5.2 to 23 fold in solid span the areas of gene therapy, drug delivery, thermotherapy, imaging
tumors, monitored 5 h post-administration [140]. Polymeric micelles, and anticancer treatments, as explained in further sections.
like Pluronics composed of ethylene oxide and propylene oxide have
been shown to overcome MDR but may, however induce complement 3.1. Functionalization of carbon nanotubes for drug delivery and other
activation [141]. applications
Another example of biodegradable polymeric nanovectors is Poly
(3-caprolactone) (PCL), used to encapsulate hydrophobic drugs like The functionalization of carbon nanotubes for biomedical applica-
tamoxifen. The nanovectors were synthesized via solvent displace- tions involves covalent or non-covalent modifications. Covalent
ment by utilizing an acetone–water system, facilitating the instant modifications are carried out by reacting carbon atoms on the
adsorption of PPO–PEO groups when the organic solution of the sidewall of carbon nanotubes to a therapeutic molecule. In biological
polymer is introduced into aqueous solution in the presence of a applications, oxidation and grafting polymers on the sidewalls of
stabilizer, Pluronic [142]. These nanovectors were taken up by the carbon nanotubes are widely adopted. In the process of oxidation, the
MCF-7 estrogen receptor-positive breast cancer cells and MDA- raw (pristine) carbon nanotubes are refluxed in nitric acid, which
MB231 human breast adenocarcinoma cells within 30 min of results in open tubes and the tips that bear oxygenated functions,
incubation [143]. In another similar study, PEO-modified PCL mainly carboxyl acids [151,152]. Another important covalent

Fig. 4. Synthetic scheme of SWNT-PEG graft copolymers. The carboxylic acid functionalized Single Walled Carbon Nanotubes (SWNT–COOH) react with oxalyl chloride to form the
acyl chloride intermediate, which is further reacted with PEG to form SWNT–PEG [131].
1346 S. Prakash et al. / Advanced Drug Delivery Reviews 63 (2011) 1340–1351

modification of carbon nanotubes involves 1,3-dipolar cycloaddition emerging nanomaterial, the use of CNTs as drug delivery carriers has
reactions [153]. Using this method, azomethineylides are added on brought great attention in recent years. Several small molecules of
the graphitic surface of CNTs, forming pyrrolidine-fused rings, Fig. 3. anticancer drug were carried to tumor cells by functionalized carbon
These covalent modifications lead to the generation of various nanotubes. Doxorubicin (DOX) is a chemotherapeutic agent and has
functional moieties on the ends and side wall of CNTs and enable been used for the treatment of many human cancers. DOX has been
the conjugation of various fluorescent dyes, drugs, peptides etc. loaded to branched PL-PEG functionalized SWNTs by π–π stacking to
[154,155]. the sidewall of carbon nanotubes [164]. In this study, the loading ratio
A detailed review on attaching various compounds and drugs of DOX was evaluated as 2.5 grams per gram of SWNTs. In vivo studies
covalently to CNTs, following the two above mentioned methods have with an injection of the SWNT formulated DOX in a mouse model of
been described elsewhere [156]. To further improve the solubility of breast cancer showed significant, enhanced therapeutic efficacy and a
carbon nanotubes, other biocompatible polymers like polyethylene marked reduction in toxicity compared with free DOX and DOXIL. The
glycol (PEG) can be grafted that enhance the solubility of the same group conducted another study using paclitaxel loaded onto
nanotubes [157] and has recently been shown to overcome the SWNTs by conjugating the drug to the amino group of the branched
problem of multidrug resistance [158]. The grafting of a biocompatible phospholipid-PEG chains via cleavable ester linkages, hydrolysed in
polymer can be achieved via an acyl chloride intermediate, which the cellular environment [165]. This resulted in a water soluble
reacts with the hydrophilic polymer PEG to form SWNT-PEG graft compound: SWNT-PEG–paclitaxel. The obtained SWNT–PTX conju-
copolymers, Fig. 4. The SWNT-PEG hybrid has been tested for neuron gate displayed higher efficacy in suppressing tumor growth than did
growth [159,160] and drug delivery [161]. Apart from covalent clinical Taxol in a murine 4T1 breast cancer model [166]. Furthermore,
modifications, non-covalent functionalization of CNTs has also been the SWNT–PTX showed prolonged blood stability and decreased
explored with different materials, including surfactants, natural toxicity. A similar scheme was developed for the conjugation of the
polymers, synthetic polyelectrolytes and amphiphilic block polymers antitumor prodrug cisplatin to PL-PEG-SWNT [167]. The platinum(IV)
or polymeric micelles. Among them, functionalization of carbon with complex, c,c,t-[Pt(NH3)2Cl2(OEt)(O2CCH2CH2CO2H)], was conjugated
some amphiphilic polymers provides good serum stability and has to the amino end group of PL-PEG on the carbon nanotubes through
been utilized in many biomedical applications. The hydrophobic peptide linkages. It was observed that the SWNTs were taken up by
moiety binds to the carbon nanotube sidewalls via π–π interactions the testicular cancer cells by endocytosis, and the pH drop in the
which, in turn, help the CNTs to disperse in aqueous solution. A recent compartment facilitated the drug release. As well, the SWNT
study demonstrated the non-covalent pyrene conjugation of glyco- formulated platinum(IV) complex showed much less toxicity when
dendrimers via π–π stacking on the sidewalls of CNTs, enhancing their compared with the free drug treatment. The targeted delivery of
water solubility [162]. Amphiphilic polymer phospholipid linked PEG anticancer agents like cisplatin has also been accomplished using
(PL-PEG) was used to non-covalently bind to the sidewall of SWNTs, SWNTs in conjugation with either quantum dots (QDs) or targeting
which gives rise to a large number of biomedical applications ranging ligand epidermal growth factor (EGF) to target squamous cancer cells.
from gene, peptide, drug delivery and biodistribution studies as well The QD luminescence facilitates the tracking of the SWNT formulation
as biological sensing and imaging [163]. and the conjugation of EGF proves that the formulation was readily
taken up due to the presence of EGF receptors on squamous
3.2. Applications of carbon nanotubes in cancer therapy carcinoma cells [168]. SWNTs have also been explored to deliver
small interfering RNAs (siRNAs) via non-covalent interactions [169]. A
3.2.1. Small hydrophobic cancer drug delivery system recent study involved cationic SWNTs and their capability to form
The administration of chemotherapeutic agents often poses a stable complexes with siRNAs with the goal to silence the expression
limitation of solubility and cell-penetration ability. In addition, the of telomerase reverse transcriptase (TERT) and inhibit cell prolifer-
systemic toxicity caused by a lack of selective drug targeting to cancer ation both in vitro and in vivo in tumor models [170].
cells, limits the clinical applications. The development of an effective The covalent functionalization of CNTs with methotrexate (MTX)
drug delivery system becomes an active area of research. As a new and was achieved via a 1,3-dipolar cycloaddition approach. In this study, a

Fig. 5. Alginate-poly-L-lysine-alginate (APA) microcapsules encapsulating carbon nanotubes. The calcium ions are responsible for cross-linking of the alginate monomeric units,
trapping the carbon nanotubes into the core of microcapsule.
S. Prakash et al. / Advanced Drug Delivery Reviews 63 (2011) 1340–1351 1347

methodology was developed for the introduction of two linking sites minimum toxicity. As for the concern of the intrinsic toxicity of CNTs,
on the sidewalls of carbon nanotubes for the fluorescent agents FITC it has been proposed that CNTs covalently functionalized with phenyl-
and MTX respectively [44]. The conjugation of MTX to carbon SO3H or phenyl (COOH)2 groups pose less toxicity to cells than raw
nanotubes enhanced its cellular internalization. However, the drug CNTs stabilized with surfactants, such as, Pluronic F108 [174]. A
release profiles and their in vitro and in vivo efficacy were not detailed account of toxicological studies performed with CNTs has
evaluated. Antitumor agent 10-hydroxycamptothecin (HCPT) was been well reviewed elsewhere [175]. Functionalized SWNTs have
delivered by a covalently modified MWNT [171]. Diaminotriethylene been used as drug carriers with paclitaxel [161,165]. Paclitaxel, being
glycol was used as a spacer linking the carboxylic groups on the insoluble in water, is currently clinically administered as a highly toxic
oxidized nanotube to drug molecules via amidation. The MWNT formulation with Cremophor EL. However, using SWNTs as a carrier
formulated HCPT showed an improved antitumor activity both in vitro allowed for a prolonged circulation of the drug and a 10-fold higher
and in vivo compared with the clinical HCPT formulation and the uptake by tumor cells [165]. The high uptake of SWNT complexes by
MWNT–HCPT conjugates have a longer blood circulation and a higher the RES may endanger the liver and spleen, as they may suffer toxic
drug accumulation at the tumor site. effects from SWNT deposition and accumulation. One of the main
problems encountered with intravenous administrations is the rapid
3.2.2. Microencapsulated carbon nanotube delivery devices for cancer clearance through the blood and another is the toxic effects to other
therapy organs and tissues. Another study tested the toxicity of intravenously
Our research focuses on microencapsulating carbon nanotubes using administered functionalized SWNTs in mice and demonstrated no
biocompatible polymeric membranes, such as alginate-poly-L-lysine- evidence of toxicity [176]. PEG-SWNTs were shown to be gradually
alginate (APA) for target specific delivery [172]. The purpose of removed from the body following an intravenous administration in
microencapsulation is to protect the payload from the harsh external mouse models with no evidence of toxicity during the excretory
environments while allowing the specific solutes to pass through the process [177].
polymeric membrane. It enables the optimum delivery of bioactive In many cases, the functionalization of nanotubes is detrimental
molecules through controlled, sustained and prolonged release of the for their serum stability and toxicity. SWNTs were made soluble in
therapeutics at the targeted site. The CNT encapsulation was achieved physiological solution by first suspending them in Polysorbate-80 and
by mixing a suspension of functionalized SWNTs with a sodium alginate experiments were performed to investigate their solubility and
solution, which was then gelled as beads formed by a microencapsulator toxicity [178]. A low-level of toxicity was observed with high dosages
in a calcium chloride solution. The beads were subsequently coated with of SWNT-Polysorbate-80 formulation via intravenous administration.
a sterile solution of poly-L-lysine which was followed by a sodium The dose accumulation was observed in the liver and the lungs [179].
alginate coating, giving rise to SWNT-APA microcapsules, Fig. 5. Our In order to understand the toxic impact of the systemic delivery of
preliminary studies evaluated the distribution of SWNTs inside the carbon nanotubes in greater detail, additional studies using a variety
polymeric core. Based on the optimization and characterization data of animal models over a longer time frame need to be performed.
obtained with the microencapsulated SWNT formulation, we are However, one way of decreasing the toxicity and the side-effects of
currently focussing on targeting the therapeutic to desired gastrointes- carbon nanotube based drug delivery would be to have targeted
tinal sites of rodents as a treatment for colon cancer. In a preliminary in delivery. Zhuang et al. exemplified this idea by conjugating PEGylated
vitro study performed by our group on a colon cancer cell line, SWNTs SWNTs to the RGD peptide that specifically binds to integrin αvβ3,
were conjugated to pEGFP in order to optimize transfection with expressed on the surface of numerous tumor cells. The formulation
minimal cellular toxicity [173]. consisted of RGD-bound SWNTs, which were intravenously injected
into U87MG tumor bearing mice. The RGD-bound SWNTs showed a
3.2.3. Selective tumor destruction via Near-Infrared radiation of carbon higher tumor uptake as compared to SWNTs alone [179].
nanotubes
It is known that SWNTs have a very strong optical absorbance in 4. Conclusions: toward future perspective, benefits and concerns
the NIR (800–2000 nm). This intrinsic property of SWNT has been
exploited for the optical stimulation of nanotubes inside the living Biomedical applications of nanotechnology have shown a clear
cells, since biological systems remain unharmed and are transparent edge over conventional methods. It promises effective treatment in a
to NIR radiation. The photoluminescent property of CNTs in the NIR cost-effective way, wherein the diseases will be combated with
region was first discovered in 2002 [171]. This property of CNTs maximum therapeutic effect and minimum intervention. It will lead
established a unique sensitive and selective imaging and detection of to a decrease in both the time and money invested into developing
biological samples and an application in laser heating cancer therapy, new medicines where modeling, targeting and safety assessment of
both in vitro and in vivo. In account of this, a formulation based on the drug require a large amount of resources. The use of particles of
FA-PL-PEG functionalized SWNTs was used for targeting cancer cells, the scale of biomolecules will lead to a better interaction and
which over-express folate receptors. Selective cancer cell destruction monitoring of cells, intercellular interactions and intracellular
was achieved by continuous NIR radiation of SWNTs. The NIR- pathways, thus providing us with an important tool for targeted
triggered cell death has been shown to remain nontoxic for drug delivery, in vivo imaging, in vitro diagnostics and regenerative
neighboring normal cells [172]. This research presented a novel medicine. It opens an opportunity for “personalized medicine”
application of carbon nanotubes in cancer therapy. Another study through which a therapy/medicine could be suggested according to
utilizing the NIR characteristic of CNTs involved the destruction of the molecular characteristics of the patient.
breast cancer cells by non-covalently attaching monoclonal antibodies Current research attempts to fuse all of the desired characteristics
against membrane markers: insulin like growth factor 1 receptor of nanomedicine into one particle, which would perform targeted
(IGF1R) and human endothelial receptor 2 (HER2). The antibodies drug delivery and monitoring of the induced repair. As with any new
were attached using π–π interactions of the pyrene rings onto the side technology, there are concerns over the application of nanomedicine,
walls of SWNTs [173]. stemming from the lack of proper knowledge and the need for the
establishment of regulatory measures. The primary concern with
3.2.4. In vivo toxicity of carbon nanotubes medical nanodevices is their potential toxicity. For example, the
Nanohybrid materials made of drug–carbon nanotube conjugants exposure of human keratinocytes to insoluble SWNTs was associated
as a potential biotherapeutic candidate for drug delivery would only with oxidative stress and apoptosis. Further alarming is the fact that
be a success if it can provide maximum bioavailability while having nanoparticles are extremely stable in the body and have been
1348 S. Prakash et al. / Advanced Drug Delivery Reviews 63 (2011) 1340–1351

designed so as to protect them from the immune system. Further- nanotechnologies is required for future clinical studies. Efficient studies,
more, no information is currently available as to how and when they using preclinical and clinical studies, as those highlighted in Table 2,
are excreted. Minimal excretion of nanoparticles could potentially be must continue to ensure efficiency and safety of any developed
harmful due to overdosing. Another concern of nanoparticles is that formulation. One also must consider that drug delivery systems are
they are active and have a large surface area capable of interacting inherently limited by the payload they deliver. Anti-cancer drug
with unintended sites if not tagged properly with targeting molecules. research must progress to allow any delivery system formulation,
Additionally, they are of a size range similar to that of proteins and can nanoparticles or CNT, to be effective in the future.
thus interfere with normal and cancerous cell signaling pathways.
Thus, care has to be taken to implement this technology in a way as to
Acknowledgments
avoid environmental, socio-economic and toxic side-effects. It should
be seen that new regulatory mechanisms are in place prior to the
The authors would like to acknowledge Canadian Institute of
avalanche of data that is forthcoming.
Health Research (CIHR) grant to Dr. S. Prakash, the support of McGill
The efficacy of nanomedicine needs to be analyzed on the basis of
Majors scholarship to Meenakshi Malhotra, McGill Provost's Graduate
pharmacogenetics due to known population variability in drug
Fellowship to Wei Shao, Industrial Innovation Scholarship (IIS) BMP
sensitivity. There is a possibility of “varying” specific and non-specific
Innovation-NSERC, FQRNT and Micropharma Limited Scholarship to
interactions due to nanoparticle contact based on cell and tissue type.
Catherine Tomaro-Duchesneau and McGill Internal scholarship to
Currently, there are no established methodologies or model system(s)
Sana Abbasi. We also acknowledge Dr. V.R. Swamy for assistance with
to study such intricacies to rule out the possibility of any hazardous
Fig. 2.
effects of nanoparticles delivered in vivo. In this scenario, vital
questions need to be addressed further: how do these nanomaterials
interact with other types of drugs, foods and secondary metabolites of References
a tissue/organ, will there be any induction or interference mediated
[1] J. Luo, N.L. Solimini, S.J. Elledge, Principles of cancer therapy: oncogene and non-
by these nanomaterials, and do they influence macromolecular oncogene addiction, Cell 136 (2009) 823–837.
interactions happening in a cell in response to different signals? An [2] D. Hanahan, R.A. Weinberg, The hallmarks of cancer, Cell 100 (2000) 57–70.
emphasis on metabolism, degradation, clearance and bioaccumula- [3] B.W. Stewart, P. Kleihues, World Cancer Report, World Health Organization, 2003.
[4] World Health Organization., Cancer, Fact Sheet #297, 2009.
tion of nanomaterials, especially CNTs remains to be addressed.
[5] S. Aggarwal, Targeted cancer therapies, Nat. Rev. Drug Discov. 9 (2010) 427–428.
Moreover, the pharmacokinetic behavior, the production reproduc- [6] H.A. Shih, J.S. Loeffler, N.J. Tarbell, Late Effects of CNS Radiation Therapy, 2009,
ibility of CNT formulations and their size distribution need to be pp. 23–41.
[7] T. Helleday, E. Petermann, C. Lundin, B. Hodgson, R.A. Sharma, DNA repair
validated both in vitro and in vivo before their use is implemented. For
pathways as targets for cancer therapy, Nat. Rev. Cancer 8 (2008) 193–204.
example, proper disposal and use of CNT formulations need to be [8] L. Kelland, The resurgence of platinum-based cancer chemotherapy, Nat. Rev.
determined to prevent environmental and health effects. A multi Cancer 7 (2007) 573–584.
dimensional investigation is needed to rule out all of the possible [9] S.G. Chaney, S.L. Campbell, B. Temple, E. Bassett, Y. Wu, M. Faldu, Protein
interactions with platinum-DNA adducts: from structure to function, J. Inorg.
short-term and long-term toxicological effects. Biochem. 98 (2004) 1551–1559.
Efforts to ensure the safety of nanomaterials require manufac- [10] M.J. Chen, T. Shimada, A.D. Moulton, A. Cline, R.K. Humphries, J. Maizel, A.W.
turers to fully assess how the “nanosizing” of drugs affects dissolution Nienhuis, The functional human dihydrofolate reductase gene, J. Biol. Chem. 259
(1984) 3933–3943.
rate, solubility and therapeutic action. Safety has to be given higher [11] C. Morales, M.J. Garcia, M. Ribas, R. Miro, M. Munoz, C. Caldas, M.A. Peinado,
priority for the in vivo use of nanoparticles, particularly those in the Dihydrofolate reductase amplification and sensitization to methotrexate of
size of b50 nm, which can enter cells via adsorptive endocytosis. methotrexate-resistant colon cancer cells, Mol. Cancer Ther. 8 (2009) 424–432.
[12] S.G. Chaney, S.L. Campbell, E. Bassett, Y. Wu, Recognition and processing of
Validated assays are mandatory for detecting and quantifying cisplatin- and oxaliplatin-DNA adducts, Crit. Rev. Oncol. Hematol. 53 (2005) 3–11.
nanoparticles in tissues and medical products. Additional standard [13] J. Graham, M. Muhsin, P. Kirkpatrick, Oxaliplatin, Nat. Rev. Drug Discov. 3 (2004)
test methods may be needed to ensure the appropriate formulation of 11–12.
[14] W.D. Meriwether, N.R. Bachur, Inhibition of DNA and RNA metabolism by
nanoparticles for drugs and biologics. However, a major challenge is
daunorubicin and adriamycin in L1210 mouse leukemia, Cancer Res. 32 (1972)
to find a reliable and affordable method of connecting the steering 1137–1142.
(delivery) system to the payload (drug). Having described both [15] C.A. Frederick, L.D. Williams, G. Ughetto, G.A. Van der Marel, J.H. Van Boom, A.
Rich, A.H.J. Wang, Structural comparison of anticancer drug–DNA complexes:
nanoparticles and CNTs, each of these should be efficiently used and
adriamycin and daunomycin, Biochemistry 29 (1990) 2538–2549.
functionalized based on the desired applications. The characteristics, [16] A. Mukherjee, R. Lavery, B. Bagchi, J.T. Hynes, Simulation Study of the Molecular
both physical and biochemical, of each delivery system should be Mechanism of Intercalation of the Anti-Cancer Drug Daunomycin into DNA,
considered in the design and drug functionalization processes. 2009, pp. 165–180.
[17] J.M.S. van Maanen, J. Retel, J. de Vries, H.M. Pinedo, Mechanism of action of
Furthermore, current preclinical studies focus on a number of antitumor drug etoposide: a review, J. Natl Cancer Inst. 80 (1988) 1526–1533.
different animal models, including tumor-induced and genetically [18] B.K. Sinha, N. Haim, L. Dusre, D. Kerrigan, Y. Pommier, DNA strand breaks
encoded cancers. This makes it difficult to compare and contrast the produced by etoposide (VP-16,213) in sensitive and resistant human breast
tumor cells: implications for the mechanism of action, Cancer Res. 48 (1988)
efficiency of each delivery system and drug combination. In addition, 5096–5100.
the route of administration, the targeted site, the stage of the cancer [19] J.C. Wurzer, R.J. Tallarida, M.A. Sirover, New mechanism of action of the cancer
and the physiological stability of the formulation are all aspects that chemotherapeutic agent 5-fluorouracil in human cells, J. Pharmacol. Exp. Ther.
269 (1994) 39–43.
need to be addressed to conclude on the efficacy of any developed [20] W.H. Eaglstein, G.D. Weinstein, P. Frost, Fluorouracil: mechanism of action in
therapeutic. For this analysis, established clinical trial methods would human skin and actinic keratoses: I. Effect on DNA synthesis in vivo, Arch.
need to be employed. Dermatol. 101 (1970) 132–139.
[21] W.B. Parker, Y.C. Cheng, Metabolism and mechanism of action of 5-fluorouracil,
There are still several questions that remain unanswered concerning
Pharmacol. Ther. 48 (1990) 381–395.
the fate of these nanomaterials in vivo and their long term effects on [22] J.W. Yarbro, Further studies on the mechanism of action of hydroxyurea, Cancer
gene expression, interaction/interference with the normal physiology, Res. 28 (1968) 1082–1087.
[23] I.H. Krakoff, N.C. Brown, P. Reichard, Inhibition of ribonucleoside diphosphate
dose effect and drug–drug interactions. These properties have to be
reductase by hydroxyurea, Cancer Res. 28 (1968) 1559–1565.
validated in addition to the conventional methods of ADMETox. The [24] J.W. Yarbro, Mechanism of action of hydroxyurea, Semin. Oncol. 19 (1992) 1–10.
answers may start coming from the clinical research and trials in cancer [25] J.S. Salser, M.E. Balis, The mechanism of action of 6-mercaptopurine, Cancer Res.
and infectious diseases, provided that the drug manufacturers can 25 (1965) 539–543.
[26] J.C. Panetta, W.E. Evans, M.H. Cheok, Mechanistic mathematical modelling of
resolve technical challenges, unwanted cellular/immunological reac- mercaptopurine effects on cell cycle of human acute lymphoblastic leukaemia
tions, in vivo stability and limitations in delivery. Industrial scale-up of cells, Br. J. Cancer 94 (2005) 93–100.
S. Prakash et al. / Advanced Drug Delivery Reviews 63 (2011) 1340–1351 1349

[27] B.P. Schiff, J. Fant, S.B. Horwitz, Promotion of microtubule assembly in vitro by [57] H. Maeda, SMANCS and polymer-conjugated macromolecular drugs: advantages
taxol, Nature 277 (1979) 665–667. in cancer chemotherapy, Adv. Drug Deliv. Rev. 46 (2001) 169–185.
[28] D.L. Morse, H. Gray, C.M. Payne, R.J. Gillies, Docetaxel induces cell death through [58] H. Maeda, Y. Matsumura, Tumoritropic and lymphotropic principles of
mitotic catastrophe in human breast cancer cells, Mol. Cancer Ther. 4 (2005) macromolecular drugs, Crit. Rev. Ther. Drug Carrier Syst. 6 (1989) 193–210.
1495–1504. [59] Y. Matsumura, H. Maeda, A new concept for macromolecular therapeutics in
[29] R.S. Herbst, F.R. Khuri, Mode of action of docetaxel — a basis for combination with cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and
novel anticancer agents, Cancer Treat. Rev. 29 (2003) 407–415. the anti-tumor agent smancs, Cancer Res. 46 (1986) 6387–6392.
[30] C.A. Sawka, K.I. Pritchard, A.H.G. Paterson, D.J.A. Sutherland, D.B. Thomson, W.E. [60] B.M. Brenner, T.H. Hostetter, H.D. Humes, Glomerular permselectivity: barrier
Shelley, R.E. Myers, B.G. Mobbs, A. Malkin, J.W. Meakin, Role and mechanism of function based on discrimination of molecular size and charge, Am. J. Physiol.
action of tamoxifen in premenopausal women with metastatic breast carcinoma, 234 (1978) F455–F460.
Cancer Res. 46 (1986) 3152–3156. [61] T. Maack, V. Johnson, S.T. Kau, J. Figueiredo, D. Sigulem, Renal filtration,
[31] A.J.J. Wood, C.K. Osborne, Tamoxifen in the treatment of breast cancer, N Engl J. transport, and metabolism of low-molecular-weight proteins: a review, Kidney
Med. 339 (2009) 1609–1618. Int. 16 (1979) 251–270.
[32] B. Gigant, C. Wang, R.B.G. Ravelli, F. Roussi, M.O. Steinmetz, P.A. Curmi, A. Sobel, [62] K. Mihara, T. Hojo, M. Fujikawa, Y. Takakura, H. Sezaki, M. Hashida, Disposition
M. Knossow, Structural basis for the regulation of tubulin by vinblastine, Nature characteristics of protein drugs in the perfused rat kidney, Pharm. Res. 10 (1993)
435 (2005) 519–522. 823–827.
[33] R.H. Himes, Interactions of the catharanthus (Vinca) alkaloids with tubulin and [63] K. Mihara, M. Mori, T. Hojo, Y. Takakura, H. Sezaki, M. Hashida, Disposition
microtubules, Pharmacol. Ther. 51 (1991) 257–267. characteristics of model macromolecules in the perfused rat kidney, Biol. Pharm.
[34] Y.H. Hsiang, R. Hertzberg, S. Hecht, L.F. Liu, Camptothecin induces protein-linked Bull. 16 (1993) 158–162.
DNA breaks via mammalian DNA topoisomerase I, J. Biol. Chem. 260 (1985) [64] T. Fujita, M. Nishikawa, C. Tamaki, Y. Takakura, M. Hashida, H. Sezaki, Targeted delivery
14873–14878. of human recombinant superoxide dismutase by chemical modification with mono-
[35] F. Leroy Liu, S.H. Desai, T.-K. Li, Y. Mao, M. Sun, S.-P. Sim, Mechanism of action of and polysaccharide derivatives, J. Pharmacol. Exp. Ther. 263 (1992) 971–978.
camptothecin, Ann. N. Y. Acad. Sci. 922 (2006) 1–10 (The Camptothecins: [65] R.I. Mahato, K. Kawabata, Y. Takakura, M. Hashida, In vivo disposition
Unfolding their Anticancer Potential). characteristics of plasmid DNA complexed with cationic liposomes, J. Drug
[36] C. Le Tourneau, S. Faivre, E. Raymond, New developments in multitargeted Target. 3 (1995) 149–157.
therapy for patients with solid tumours, Cancer Treat. Rev. 34 (2008) 37–48. [66] S.F. Ma, M. Nishikawa, H. Katsumi, F. Yamashita, M. Hashida, Cationic charge-
[37] Y. Malam, M. Loizidou, A.M. Seifalian, Liposomes and nanoparticles: nanosized dependent hepatic delivery of amidated serum albumin, J. Control Release 102
vehicles for drug delivery in cancer, Trends Pharmacol. Sci. 30 (2009) 592–599. (2005) 583–594.
[38] R. Sinha, G.J. Kim, S. Nie, D.M. Shin, Nanotechnology in cancer therapeutics: [67] R. Duncan, Polymer conjugates as anticancer nanomedicines, Nat. Rev. Cancer 6
bioconjugated nanoparticles for drug delivery, Mol. Cancer Ther. 5 (2006) (2006) 688–701.
1909–1917. [68] K. Ulbrich, V. Subr, Polymeric anticancer drugs with pH controlled activation,
[39] T. Tsuruo, M. Naito, A. Tomida, N. Fujita, T. Mashima, H. Sakamoto, N. Haga, Adv. Drug Deliv. Rev. 23 (2004) 1023–1050.
Molecular targeting therapy of cancer: drug resistance, apoptosis and survival [69] V.P. Torchilin, Lipid-core micelles for targeted drug delivery, Curr. Drug Deliv. 2
signal, Cancer Sci. 94 (2005) 15–21. (2005) 319–327.
[40] L.S. Jabr-Milane, L.E. van Vlerken, S. Yadav, M.M. Amiji, Multi-functional nanocarriers [70] F. Zeng, J. Liu, C. Allen, Synthesis and characterization of biodegradable poly
to overcome tumor drug resistance, Cancer Treat. Rev. 34 (2008) 592–602. (ethylene glycol)-block-poly(5-benzyloxy-trimethylene carbonate) copolymers
[41] A.S. Hoffman, The origins and evolution of “controlled” drug delivery systems, J. for drug delivery, Biomacromolecules 5 (2004) 1810–1817.
Control. Release 132 (2008) 153–163. [71] A. Roby, S. Erdogan, V.P. Torchilin, Solubilization of poorly soluble PDT agent,
[42] Abraxane., Abraxis BioScience, LLC, www.abraxane.com 2010. mesotetraphenylporphin, in plain or immunotargeted PEG-PE micelles results in
[43] W.J. Gradishar, S. Tjulandin, N. Davidson, H. Shaw, N. Desai, P. Bhar, M. Hawkins, dramatically improved cancer cell killing in vitro, Eur. J. Pharm. Biopharm. 62
J. O'Shaughnessy, Phase III trial of nanoparticle albumin-bound paclitaxel (2006) 235–240.
compared with polyethylated castor oil-based paclitaxel in women with breast [72] C.L. Bartels, A.F. Wilson, How does a novel formulation of paclitaxel affect drug
cancer, J. Clin. Oncol. 23 (2005) 7794–7803. delivery in metastatic breast cancer? US Pharm. 29 (2004) HS18.
[44] G. Pastorin, W. Wu, S. Wieckowski, J.P. Briand, K. Kostarelos, M. Prato, A. Bianco, [73] K. Garber, Improved paclitaxel formulation hints at new chemotherapy
Double functionalisation of carbon nanotubes for multimodal drug delivery, approach, J. Natl Cancer Inst. 96 (2004) 90–91.
Chem. Commun. 11 (2006) 1182–1184. [74] S. Tsukagoshi, A new LH-RH agonists for treatment of prostate cancer, 3 month
[45] P.A. Vasey, S.B. Kaye, R. Morrison, C. Twelves, P. Wilson, R. Duncan, A.H. Thomson, L. controlled release formulation of goserelin acetate (Zoladex LA 10.8 mg depot).
S. Murray, T.E. Hilditch, T. Murray, S. Burtles, D. Fraier, E. Frigerio, J. Cassidy, Phase I Outline of preclinical and clinical studies, Gan To Kagaku Ryoho 29 (2002) 1675–1687.
clinical and pharmacokinetic study of PK1 [N-(2-hydroxypropyl)methacrylamide [75] C.F. Heyns, M.P. Simonin, P. Grosgurin, R. Schall, H.C. Porchet, Comparative
copolymer doxorubicin]: first member of a new class of chemotherapeutic agents efficacy of triptorelin pamoate and leuprolide acetate in men with advanced
drug–polymer conjugates, Clin. Cancer Res. 5 (1999) 83–94. prostate cancer, BJU Int. 92 (2003) 226–231.
[46] J.M. Rademaker-Lakhai, C. Terret, S.B. Howell, C.M. Baud, R.F. Boer, D. Pluim, J.H. [76] V.P. Torchilin, Recent advances with liposomes as pharmaceutical carriers, Nat.
Beijnen, J.H.M. Schellens, J.P. Droz, A phase I and pharmacological study of the Rev. Drug Discov. 4 (2005) 145–160.
platinum polymer AP5280 given as an intravenous infusion once every 3 weeks [77] I. Brigger, C. Dubernet, P. Couvreur, Nanoparticles in cancer therapy and
in patients with solid tumors, Clin. Cancer Res. 10 (2004) 3386–3395. diagnosis, Adv. Drug Deliv. Rev. 54 (2002) 631–651.
[47] D.P. Nowotnik, E. Cvitkovic, ProLindac(TM) (AP5346): a review of the [78] J.W. Singer, S. Shaffer, B. Baker, A. Bernareggi, S. Stromatt, D. Nienstedt, M.
development of an HPMA DACH platinum polymer therapeutic. polymer Besman, Paclitaxel polyglumex (XYOTAX; CT-2103) [XYOTAX TM]: an intracel-
therapeutics: clinical applications and challenges for development, Adv. Drug lularly targeted taxane, Anti-cancer Drug 16 (2005) 243–254.
Deliv. Rev. 61 (2009) 1214–1219. [79] G.M. Springett, C. Takimoto, M. McNamara, J.H. Doroshow, S. Syed, E. Eastham, D.
[48] L.W. Seymour, D.R. Ferry, D. Anderson, S. Hesslewood, P.J. Julyan, R. Poyner, J. Spriggs, S. Pezzulli, G. Michelson, J. Dupont, Phase 1 study of CT2106
Doran, A.M. Young, S. Burtles, D.J. Kerr, Hepatic drug targeting: phase I (polyglutamate camptothecin) in patients with advanced malignancies, J. Clin.
evaluation of polymer-bound doxorubicin, J. Clin. Oncol. 20 (2002) 1668–1676. Oncol. 22 (2004) 3127 Suppl. S.
[49] T. Nakanishi, S. Fukushima, K. Okamoto, M. Suzuki, Y. Matsumura, M. Yokoyama, [80] V.J. Maria, R. Duncan, Polymer conjugates: nanosized medicines for treating
T. Okano, Y. Sakurai, K. Kataoka, Development of the polymer micelle carrier cancer, Trends Biotechnol. 24 (2006) 39–47.
system for doxorubicin, J. Control. Release 74 (2001) 295–302. [81] P. Rejmanova, J. Kopecek, R. Duncan, J.B. Lloyd, Stability in rat plasma and serum
[50] Y. Matsumura, T. Hamaguchi, T. Ura, K. Muro, Y. Yamada, Y. Shimada, K. Shirao, T. of lysosomally degradable oligopeptide sequences in N-(2-hydroxypropyl)
Okusaka, H. Ueno, M. Ikeda, N. Watanabe, Phase I clinical trial and pharmaco- methacrylamide) copolymers, Biomaterials 6 (1985) 45–48.
kinetic evaluation of NK911, a micelle-encapsulated doxorubicin, Br. J. Cancer 91 [82] L.W. Seymour, K. Ulbrich, P.S. Steyger, M. Brereton, V. Subr, J. Strohalm, R.
(2004) 1775–1781. Duncan, Tumour tropism and anti-cancer efficacy of polymer-based doxorubicin
[51] T.Y. Kim, D.W. Kim, J.Y. Chung, S.G. Shin, S.C. Kim, D.S. Heo, N.K. Kim, Y.J. Bang, prodrugs in the treatment of subcutaneous murine B16F10 melanoma, Br. J.
Phase I and pharmacokinetic study of genexol-PM, a cremophor-free, polymeric Cancer 70 (1994) 636–641.
micelle-formulated paclitaxel, in patients with advanced malignancies, Clin. [83] L.W. Seymour, K. Ulbrich, J. Strohalm, J. Kopecek, R. Duncan, Pharmacokinetics of
Cancer Res. 10 (2004) 3708–3716. polymer-bound adriamycin, Biochem. Pharmacol. 39 (1990) 1125–1131.
[52] R. Bhatt, P. Vries, J. Tulinsky, G. Bellamy, B. Baker, J.W. Singer, P. Klein, Synthesis [84] K. Ulbrich, T. Etrych, P. Chytil, M. Jelínková, B. Ríhová, Antibody targeted
and in vivo antitumor activity of Poly(L-glutamic acid) conjugates of 20(S)- polymer-doxorubicin conjugates with pH controlled activation, J Drug Target. 12
camptothecin, J. Med. Chem. 46 (2003) 190–193. (2004) 477–489.
[53] P. Sabbatini, C. Aghajanian, D. Dizon, S. Anderson, J. Dupont, J.V. Brown, W.A. [85] V. Omelyanenko, P. Kopecková, C. Gentry, J.G. Shiah, J. Kopecek, HPMA
Peters, A. Jacobs, A. Mehdi, S. Rivkin, A.J. Eisenfeld, D. Spriggs, Phase II study of copolymer-anticancer drug-OV-TL-TL16 antibody conjugates. 1. Influence of
CT-2103 in patients with recurrent epithelial ovarian, fallopian tube, or primary the method of the synthesis on the binding affinity to OVCAR-3 ovarian
peritoneal carcinoma, J. Clin. Oncol. 22 (2004) 4523–4531. carcinoma cells in-vitro, J Drug Target. 3 (1996) 357–373.
[54] P. Carmeliet, R.K. Jain, Angiogenesis in cancer and other diseases, Nature 407 [86] K.W. Wan, M. Vicent, R. Duncan, Targeting endothelial cells using HPMA
(2000) 249–257. copolymer–doxorubicin–RGD conjugates, Proc. Int'l Symp. Control. Rel. Bioact.
[55] H. Pelicano, D.S. Martin, R.H. Xu, P. Huang, Glycolysis inhibition for anticancer Mater. 30 (2003) 491–492.
treatment, Oncogene 25 (2006) 4633–4646. [87] H. Maeda, T. Sawa, T. Konno, Mechanism of tumor-targeted delivery of macromolec-
[56] R.K. Jain, Delivery of molecular medicine to solid tumors: lessons from in vivo ular drugs, including the EPR effect in solid tumor and clinical overview of the
imaging of gene expression and function, J. Control. Release 74 (2001) 7–25. prototype polymeric drug SMANCS, J. Control Release 74 (2001) 47–61.
1350 S. Prakash et al. / Advanced Drug Delivery Reviews 63 (2011) 1340–1351

[88] R.M. Schiffelers, A. Ansari, J. Xu, Q. Zhou, Q. Tang, G. Storm, G. Molema, P.Y. Lu, P.V. [119] J. Panyam, V. Labhasetwar, Biodegradable nanoparticles for drug and gene
Scaria, M.C. Woodle, Cancer siRNA therapy by tumor selective delivery with ligand- delivery to cells and tissue, Adv. Drug Deliv. Rev. 55 (2003) 329–347.
targeted sterically stabilized nanoparticle, Nucleic Acids Res. 32 (2004) e149. [120] R.A. Jain, The manufacturing techniques of various drug loaded biodegradable
[89] R. Kircheis, T. Blessing, S. Brunner, L. Wightman, E. Wagner, Tumor targeting poly(lactide–coglycolide) (PLGA) devices, Biomaterials 21 (2000) 2475–2490.
with surface shielded ligand–polycation DNA complexes, J. Control. Release 72 [121] S. Hanafusa, Y. Matsusue, T. Yasunaga, T. Yamamuro, M. Oka, Y. Shikinami, Y.
(2001) 165–170. Ikada, Biodegradable plate fixation of rabbit femoral shaft osteotomies. A
[90] S.H. Kim, J.H. Jeong, K.C. Cho, S.W. Kim, T.G. Park, Target-specific gene silencing comparative study, Clin. Orthop. Relat. Res. 315 (1995) 262–271.
by siRNA plasmid DNA complexed with folate-modified poly(ethylenimine), J. [122] Y. Matsusue, S. Hanafusa, T. Yamamuro, Y. Shikinami, Y. Ikada, Tissue reaction of
Control. Release 104 (2005) 223–232. bioabsorbable ultra high strength poly(L-lactide) rod. A long-term study in
[91] W. Suh, S.O. Han, L. Yu, S.W. Kim, An angiogenic, endothelial-cell-targeted rabbits, Clin. Orthop. Relat. Res. 317 (1995) 246–253.
polymeric gene carrier, Mol. Ther. 6 (2002) 664–672. [123] R. Langer, Tissue engineering: a new field and its challenges, Pharm. Res. 14
[92] M. Kursa, G.F. Walker, V. Roessler, M. Ogris, W. Roedl, R. Kircheis, E. Wagner, Novel (1997) 840–841.
shielded transferrin–polyethylene glycol–polyethylenimine/DNA complexes for [124] D.J. Mooney, K. Sano, P.M. Kaufmann, K. Majahod, B. Schloo, J.P. Vacanti, R.
systemic tumortargeted gene transfer, Bioconjug. Chem. 14 (2003) 222–231. Langer, Long-term engraftment of hepatocytes transplanted on biodegradable
[93] H. Lee, T.H. Kim, T.G. Park, A receptor-mediated gene delivery system using polymer sponges, J. Biomed. Mater. Res. 37 (1997) 413–420.
streptavidin and biotin-derivatized, pegylated epidermal growth factor, J. [125] S.K. Sahoo, W. Ma, V. Labhasetwar, Efficacy of transferrin-conjugated paclitaxel-
Control. Release 83 (2002) 109–119. loaded nanoparticles in a murine model of prostate cancer, Int. J. Cancer 112
[94] T. Merdan, J. Callahan, H. Petersen, K. Kunath, U. Bakowsky, P. Kopeckova, T. (2004) 335–340.
Kissel, J. Kopecek, Pegylated polyethylenimine-Fab' antibody fragment conju- [126] S.H. Kim, J.H. Jeong, K.W. Chun, T.G. Park, Target-specific cellular uptake of PLGA
gates for targeted gene delivery to human ovarian carcinoma cells, Bioconjug. nanoparticles coated with poly(L-lysine)–poly(ethylene glycol)–folate conju-
Chem. 14 (2003) 989–996. gate, Langmuir 21 (2005) 8852–8857.
[95] A. Natarajan, C.Y. Xiong, H. Albrecht, G.L. DeNardo, S.J. DeNardo, Characterization [127] Y. Mo, L.Y. Lim, Paclitaxel-loaded PLGA nanoparticles: potentiation of anticancer
of site-specific ScFv PEGylation for tumor-targeting pharmaceuticals, Bioconjug. activity by surface conjugation with wheat germ agglutinin, J. Control. Release
Chem. 16 (2005) 113–121. 108 (2005) 244–262.
[96] V.P. Torchilin, Drug targeting, Eur. J. Pharm. Sci. 11 (2000) S81–S91. [128] M. Cegnar, A. Premzl, V. Zavasnik-Bergant, J. Kristl, J. Kos, Poly(lactide-co-
[97] R. Gref, A.J. Domb, P. Quellec, T. Blunk, R.H. Muller, J.M. Verbavatz, R. Langer, The glycolide) nanoparticles as a carrier system for delivering cysteine protease
controlled intravenous delivery of drugs using PEG-coated sterically stabilized inhibitor cystatin into tumor cells, Exp. Cell Res. 301 (2004) 223–231.
nanospheres, Adv. Drug Deliv. Rev. 16 (1995) 215–233. [129] H.S. Yoo, K.H. Lee, J.E. Oh, T.G. Park, In vitro and in vivo anti-tumor activities of
[98] C. Delgado, G.E. Francis, D. Fisher, The uses and properties of PEG-linked nanoparticles based on doxorubicin–PLGA conjugates, J. Control. Release 68
proteins, Crit. Rev. Ther. Drug Carrier Syst. 9 (1992) 249–304. (2000) 419–431.
[99] R. Mehvar, Dextrans for targeted and sustained delivery of therapeutic and [130] L. Zhang, J.M. Chan, F.X. Gu, J.-W. Rhee, A.Z. Wang, A.F. Radovic-Moreno, F. Alexis,
imaging agents, J. Control. Release 69 (2000) 1–25. R. Langer, O.C. Farokhzad, Self-assembled lipid polymer hybrid nanoparticles: a
[100] R.B. Greenwald, C.D. Conover, Y.H. Choe, Poly(ethylene glycol) conjugated drugs robust drug delivery platform, Am. Chem. Soc. 2 (2008) 1696–1702.
and prodrugs: a comprehensive review, Crit. Rev. Ther. Drug Carrier Syst. 17 [131] C.-M.J. Hu, S. Kaushal, H.S.T. Cao, S. Aryal, M. Sartor, S. Esener, M. Bouvet, L.
(2000) 101–161. Zhang, Half-antibody functionalized lipid–polymer hybrid nanoparticles for
[101] S. Kommareddy, D.B. Shenoy, M. Amiji, Nanoparticulate carriers of gelatin and targeted drug delivery to carcinoembryonic antigen presenting pancreatic
gelatin derivatives, in: C. Kumar (Ed.), Biological and Pharmaceutical Nanoma- cancer cells, Mol. Pharm. 7 (2010) 914–920.
terials, Wiley-VCH, Weinheim, Germany, 2005, pp. 330–347. [132] G. Kwon, M. Yokoyama, T. Okano, Y. Sakurai, K. Kataoka, Biodistribution of
[102] V.P. Torchilin, How do polymers prolong circulation time of liposomes? J. micelle-forming polymer–drug conjugates, Pharm. Res. 10 (1993) 970–974.
Liposome Res. 6 (1996) 99–116. [133] G. Kwon, S. Suwa, M. Yokoyama, T. Okano, Y. Sakurai, K. Kataoka, Enhanced
[103] D. Oupicky, K.A. Howard, C. Konak, P.R. Dash, K. Ulbrich, L.W. Seymour, Steric tumor accumulation and prolonged circulation times of micelle-forming poly
stabilization of poly-L-lysine/DNA complexes by the covalent attachment of (ethylene oxide–aspartate) block copolymer–adriamycin conjugates, J. Control
semitelechelic poly[N-(2-hydroxypropyl)methacrylamide], Bioconjug. Chem. 11 Release 29 (1994) 17–23.
(2000) 492–501. [134] F. Kohori, K. Sakai, T. Aoyagi, M. Yokoyama, Y. Sakurai, T. Okano, Preparation and
[104] D.B. Fenske, I. MacLachlan, P.R. Cullis, Long-circulating vectors for the systemic characterization of thermally responsive block copolymer micelles comprising
delivery of genes, Curr. Opin. Mol. Ther. 3 (2001) 153–158. poly(N-isopropylacrylamide-b-DL-lactide), J. Control Release 55 (1998) 87–98.
[105] H.J. Krause, A. Schwarz, P. Rohdewald, Interfacial polymerization, a useful [135] J.E. Chung, M. Yokoyama, T. Aoyagi, Y. Sakurai, T. Okano, Effect of molecular
method for the preparation of polymethylcyanoacrylate nanoparticles, Drug architecture of hydrophobically modified poly(N-isopropylacrylamide) on the
Dev. Ind. Pharm. 12 (1986) 527–552. formation of thermoresponsive core-shell micellar drug carriers, J. Control
[106] R.S.R. Murthy, L. Harivardhan Reddy, Poly(alkyl cyanoacrylate) nanoparticles for Release 53 (1998) 119–130.
delivery of anti-cancer drugs, in: M.M. Amiji (Ed.), Nanotechnology for cancer [136] D. Le Garrec, J. Taillefer, J.E. Van Lier, V. Lenaerts, J.C. Leroux, Optimizing pH-
therapy, Taylor & Francis Group, LLC., FL, 2007, pp. 251–288. responsive polymeric micelles for drug delivery in a cancer photodynamic
[107] B. Seijo, Design of nanoparticles of less than 50 nm diameter: preparation, therapy model, J. Drug Target. 10 (2002) 429–437.
characterization and drug loading, Int. J. Pharm. 62 (1990) 1–7. [137] G. Ashwell, J. Harford, Carbohydrate-specific receptors of the liver, Annu. Rev.
[108] L.H. Reddy, R.S.R. Murthy, Influence of polymerization technique and experi- Biochem. 51 (1982) 531–554.
mental variables on the particle properties and release kinetics of methotrexate [138] J. Kuiper, The Liver: Biology and Pathobiology, 3rd Raven Press, New York, 1994.
from poly(butylcyanoacrylate) nanoparticles, Acta Pharm. 54 (2004) 103–118. [139] G.R. Newkome, Z. Yao, G.R. Baker, V.K. Gupta, Cascade molecules: a new
[109] P. Couvreur, B. Kante, M. Roland, P. Speiser, Adsorption of antineoplastic drugs to approach to micelles, A[27]-arborol, J. Org. Chem. 50 (1985) 2003–2004.
polyalkyl cyanoacrylate nanoparticles and their release in calf serum, J. Pharm. [140] J.C. Roberts, M.K. Bhalgat, R.T. Zera, Preliminary biological evaluation of
Sci. 68 (1979) 1521–1524. polyamidoamine (PAMAM) Starburst dendrimers, J. Biomed. Mater. Res. 30
[110] J. Kreuter, H.R. Hartman, Comparative study on the cytostatic effects and tissue (1996) 53–65.
distribution of 5-fluorouracil in a free from and bound to polybutylcyanocrylate [141] P.D. Scholes, A.G. Coombes, L. Illum, S.S. Davis, J.F. Watts, C. Ustariz, M. Vert, M.C.
nanoparticles in sarcoma 180-bearing mice, Oncology 40 (1983) 363. Davies, Detection and determination of surface levels of poloxamer and PVA
[111] L. Marchal-Heussler, P. Maincent, H. Hoffman, J. Spittler, P. Couvereur, Antiglau- surfactant on biodegradable nanospheres using SSIMS and XPS, J. Control.
comatous activity of betaxolol chlorhydrate sorbed onto different isobutyl Release 59 (1999) 261–278.
cyanoacrylate nanoparticle preparations, Int. J. Pharm. 58 (1990) 115–122. [142] J.S. Chawla, M.M. Amiji, Cellular uptake and concentrations of tamoxifen upon
[112] N. Brasseur, D. Brault, P. Couvreur, Adsorption of hematoporphyrin onto administration in poly(epsilon-caprolactone) nanoparticles, AAPS PharmSci 5
polyalkyl cyanoacrylate nanoparticles: carrier capacity and drug release, Int. J. (2003) E3.
Pharm. 70 (1991) 129–135. [143] H. Hu, B. Zhao, M.E. Itkis, R.C. Haddo, Nitric acid purification of single-walled
[113] R. Gaspar, V. Preat, M. Roland, Nanoparticles of polyisohexyl cyanoacrylate carbon nanotubes, J. Phys. Chem. B 107 (2003) 13838–13842.
(PIHCA) as carriers of primaquine: formulation, physico-chemical characteriza- [144] F. Liang, B. Chen, A review on biomedical applications on single-walled carbon
tion and acute toxicity, Int. J. Pharm. 68 (1991) 111–119. nanotubes, Curr. Med. Chem. 17 (2010) 10–24.
[114] C. Verdun, P. Couvreur, H. Vranckx, V. Lenaerts, M. Roland, Development of a [145] D.S. Bethune, C.H. Klang, M.S. de Vries, G. Gorman, R. Savoy, J. Vazquez, R. Beyers,
nanoparticle controlled-release formulation for human use, J. Control. Release 3 Cobalt-catalysed growth of carbon nanotubes with single-atomic-layer walls,
(1986) 205–210. Nature 363 (1993) 605–607.
[115] L. Illum, M.A. Khan, E. Mak, S.S. Davis, Evaluation of carrier capacity and release [146] A. Thess, R. Lee, P. Nikolaev, H. Diah, P. Petit, J. Robert, C. Xu, J.E. Fischer, R.E.
characteristic for poly(butyl-2-cyanoacrylate) nanoparticles, Int. J. Pharm. 30 Samalley, Crystalline ropes of metallic nanotubes, Science 273 (1996) 483–487.
(1986) 17–28. [147] A.M. Cassel, J.A. Raymakers, J. Kong, H. Dia, Large scale CVD synthesis of single-
[116] T. Harmia, P. Speiser, J. Kreuter, Optimization of pilocarpine loading onto walled carbon nanotubes, J. Phys. Chem. B 103 (1999) 6484–6492.
nanoparticles by sorption procedures, Int. J. Pharm. 33 (1986) 45–54. [148] T. Park, S. Banerjee, T. Hemraj-Bennya, S.S. Wong, Purification strategies and
[117] M. Simeonova, R. Velichkova, G. Ivanova, V. Enchev, I. Abrahams, Study on the purity visualization techniques for single-walled carbon nanotubes, J. Mater.
role of 5-fluorouracil in the polymerization of butylcyanoacrylate during the Chem. 16 (2006) 141–154.
formation of nanoparticles, J. Drug Target. 12 (2004) 49–56. [149] R. Hirlekar, M. Yamagar, H. Garse, M. Vij, V. Kadam, Carbon nanotubes and its
[118] B. Stella, S. Arpicco, M.T. Peracchia, D. Desmaële, J. Hoebeke, M. Renoir, J. applications: a review, Asian J. Pharm.Clin. Res. 2 (2009) 17–27.
D'Angelo, L. Cattel, P. Couvreur, Design of folic acid-conjugated nanoparticles for [150] A. Bianco, K. Kostarelos, C.D. Partidos, M. Prato, Biomedical applications of
drug targeting, J. Pharm. Sci. 89 (2000) 1452–1464. functionalised carbon nanotubes, Chem. Commun. (2005) 571–577.
S. Prakash et al. / Advanced Drug Delivery Reviews 63 (2011) 1340–1351 1351

[151] D. Bonifazi, C. Nacci, R. Marega, S. Campidelli, G. Ceballos, S. Modesti, M. [167] A. Kulamarva, J. Bhathena, M. Malhotra, S. Sebak, O. Nalamasu, P. Ajayan, S.
Meneghetti, M. Prato, Microscopic and spectroscopic characterization of paint Prakash, In vitro cytotoxicity of functionalized single walled carbon nanotubes
brush-like single-walled carbon nanotubes, Nano Lett. 6 (2006) 1408–1414. for targeted gene delivery applications, Nanotoxicology 2 (2008) 184–188.
[152] B. Zhao, H. Hu, A. Yu, D. Perea, R.C. Haddon, Synthesis and characterization of [168] S.A. Bhirde, V. Pate, J. Gavard, G. Zhang, A.A. Sousa, A. Masedunskas, R.D.
water soluble single-walled carbon nanotube graft copolymers, J. Am. Chem. Soc. Leapman, R. Weigert, G.S. Gutkind, J.F. Rusling, Targeted killing of cancer cells
127 (2005) 8197–8203. in vivo and in vitro with EGF directed carbon nanotube based drug delivery, ACS
[153] E.B. Malarkey, R.C. Reyes, B. Zhao, R.C. Haddon, V. Parpura, Water soluble single- Nano 3 (2009) 307–316.
walled carbon nanotubes inhibit stimulated endocytosis in neurons, Nano Lett [169] R. Krajcik, A. Jung, A. Hirsch, W. Neuhuber, O. Zolk, Functionalization of carbon
8 (2008) 3538–3542. nanotubes enables non-covalent binding and intracellular delivery of small
[154] E.B. Malarkey, K.A. Fisher, E. Bekyarova, W. Liu, R.C. Haddon, V. Parpura, interfering RNA for efficient knockdown of genes, Biochem. Biophys. Res.
Conductive single-walled carbon nanotube substrates modulate neuronal Commun. 369 (2008) 595–602.
growth, Nano Lett 9 (2009) 264–268. [170] Z. Zhang, X. Yang, Y. Zhang, B. Zeng, S. Wang, T. Zhu, R.B.S. Roden, Y. Chen, R.
[155] C.L. Lay, H.Q. Liu, H.R. Tan, Y. Liu, Delivery of paclitaxel by physically loading onto Yang, Delivery of telomerase reverse transcriptase small interfering RNA in
poly(ethylene glycol) (PEG)-graft-carbon nanotubes for potent cancer thera- complex with positively charged single-walled carbon nanotubes suppresses
peutics, Nanotechnology 21 (2010) 065101. tumor growth, Clin. Cancer Res. 12 (2006) 4933–4936.
[156] M. Prato, K. Kostarelos, A. Bianco, Functionalized carbon nanotubes in drug [171] M.J. O'Connell, S.H. Bachilo, C.B. Huffman, V.C. Moore, M.S. Strano, E.H. Haroz, K.L.
design and discovery, Acc. Chem. Res. 41 (2008) 60–68. Rialon, P.J. Boul, W.H. Noon, C. Kittrell, J. Ma, R.H. Hauge, R.B. Weisman, R.E.
[157] N. Tagmatarchis, M. Prato, Functionalization of carbon nanotubes via 1,3-dipolar Smalley, Band gap fluorescence from individual single-walled carbon nanotubes,
cycloadditions, J. Mater. Chem. 14 (2004) 437–439. Science 297 (2002) 593–596.
[158] J. Chen, M.J. Meziani, Y.P. Sun, S.H. Cheng, Poly(ethylene glycol)-conjugated [172] N.W. Kam, M. O'Connell, J.A. Wisdom, H. Dai, Carbon nanotubes as multi-
multi-walled carbon nanotubes as an efficient drug carrier for overcoming multi functional biological transporters and near-infrared agents for selective cancer
drug resistance, Toxicol. Appl. Pharmacol. 250 (2011) 184–193. cell destruction, Proc. Natl Acad. Sci. USA 102 (2005) 11600–11605.
[159] K. Kostarelos, L. Lacerda, G. Pastorin, W. Wu, S. Wieckowski, J. Luangsivilay, S. [173] N. Shao, S. Lu, E. Wickstrom, B. Panchapakesan, Integrated molecular targeting of
Godefroy, D. Pantarotto, J.P. Briand, S. Muller, M. Prato, A. Bianco, Cellular uptake IGF1R and HER2 surface receptors and destruction of breast cancer cells using
of functionalized carbon nanotubes is independent of functional group and cell single wall carbon nanotubes, Nanotechnology 18 (2007) 315101.
type, Nat. Nanotechnol. 2 (2007) 108–113. [174] C.M. Sayes, F. Liang, J.L. Hudson, J. Mendez, W. Guo, J.M. Beach, V.C. Moore, C.D.
[160] Z. Liu, S.M. Tabakman, Z. Chen, H. Dai, Preparation of carbon nanotube Doyle, J.L. West, W.E. Billups, K.D. Ausman, V.L. Colvin, Functionalization density
bioconjugates for biomedical applications, Nat. Protoc. 4 (2009) 1372–1382. dependence of single-walled carbon nanotubes cytotoxicity in-vitro, Toxicol.
[161] Z. Liu, A.C. Fan, K. Rakhra, S. Sherlock, A. Goodwin, X. Chen, Q. Yang, D.W. Felsher, Lett. 16 (2006) 135–142.
H. Dai, Supramolecular stacking of doxorubicin on carbon nanotubes for in vivo [175] L. Lacerda, A. Bianco, M. Prato, K. Kostarelos, Carbon nanotubes as nanomedicines:
cancer therapy, Angew. Chem. Int. Ed Engl. 48 (2009) 7668–7672. from toxicology to pharmacology, Adv. Drug Deliv. Rev. 58 (2006) 1460–1470.
[162] P. Wu, X. Chen, N. Hu, U.C. Tam, O. Blixt, A. Zettl, C.R. Bertozzi, Biocompatible [176] M.L. Schipper, N. Nakayama-Ratchford, C.R. Davis, N.W.S. Kam, P. Chu, Z. Liu, X.
carbon nanotubes generated by functionalization with glycodendrimers, Angew. Sun, H. Dai, S.S. Gambhir, A pilot toxicology study of single-walled carbon
Chem. Int. Ed. 47 (2008) 5022–5025. nanotubes in a small sample of mice, Nat. Nanotechnol. 3 (2008) 216–221.
[163] Z. Liu, K. Chen, C. Davis, S. Sherlock, Q. Cao, X. Chen, H. Dai, Drug delivery with [177] Zhuang Liu, C. Davis, W. Cai, L. Hi, X. Chen, H. Dai, Circulation and long-term fate
carbon nanotubes for in vivo cancer treatment, Cancer Res. 68 (2008) 6652–6660. of functionalized, biocompatible single-walled carbon nanotubes in mice probed
[164] R.P. Feazell, N. Nakayama-Ratchford, H. Dai, S.J. Lippard, Soluble single-walled by Raman spectroscopy, Proc. Natl Acad. Sci. 105 (2008) 1410–1415.
carbon nanotubes as longboat delivery systems for platinum(IV) anticancer drug [178] S.T. Yang, X. Wang, G. Jia, Y. Gu, T. Wang, H. Nie, C. Ge, H. Wang, Y. Liu, Long-term
design, J. Am. Chem. Soc. 129 (2007) 8438–8439. accumulation and low toxicity of single-walled carbon nanotubes in intrave-
[165] W. Wu, R. Li, X. Bian, Z. Zhu, D. Ding, X. Li, Z. Jia, X. Jiang, Y. Hu, Covalently nously exposed mice, Toxicol. Lett. 181 (2008) 182–189.
combining carbon nanotubes with anticancer agent: preparation and antitumor [179] Z. Liu, W. Cai, L. He, N. Nakayama, K. Chen, X. Sun, X. Chen, H. Dai, In vivo
activity, ACS Nano 3 (2009) 2740–2750. biodistribution and highly efficient tumour targeting of carbon nanotubes in
[166] A. Kulamarva, P. Raja, J. Bhathena, H. Chen, S. Talapatra, P. Ajayan, O. Nalamasu, S. mice, Nat. Nanotechnol. 2 (2007) 47–52.
Prakash, Microcapsulecarbon nanotube devices for therapeutic applications,
Nanotechnology 20 (2009) 025612.

You might also like