Skip to content
BY-NC-ND 3.0 license Open Access Published by De Gruyter Open Access September 14, 2017

Therapeutic nanomedicine surmounts the limitations of pharmacotherapy

  • Arome Odiba EMAIL logo , Victoria Ottah , Comfort Ottah , Ogechukwu Anunobi , Chimere Ukegbu , Affiong Edeke , Robert Uroko and Kingsley Omeje
From the journal Open Medicine

Abstract

Science always strives to find an improved way of doing things and nanoscience is one such approach. Nanomaterials are suitable for pharmaceutical applications mostly because of their size which facilitates absorption, distribution, metabolism and excretion of the nanoparticles. Whether labile or insoluble nanoparticles, their cytotoxic effect on malignant cells has moved the use of nanomedicine into focus. Since nanomedicine can be described as the science and technology of diagnosing, treating and preventing diseases towards ultimately improving human health, a lot of nanotechnology options have received approval by various regulatory agencies. Nanodrugs also have been discovered to be more precise in targeting the desired site, hence maximizing the therapeutic effects, while minimizing side-effects on the rest of the body. This unique property and more has made nanomedicine popular in therapeutic medicine employing nanotechnology in genetic therapy, drug encapsulation, enzyme manipulation and control, tissue engineering, target drug delivery, pharmacogenomics, stem cell and cloning, and even virus-based hybrids. This review highlights nanoproducts that are in development and have gained approval through one clinical trial stage or the other.

1 Introduction

Nanotechnology has stayed with us for more than a decade now, and governments as well as independent funding organizations/agencies have pumped billions of US dollars into research that has produced tens of thousands of published papers. But what exactly are we talking about? This question is pertinent because the very next follow-up issues are considerations on how to carve-out “Nanoscience” and the associated fields into a different discipline rather than remaining an integrated concept in other disciplines. For instance, nanomedicine is already beginning to produce other sub-fields under our watch even though it is yet to be fully pronounced as an accredited discipline of its own. Go to the archives and you will see a whole lot of journal papers, books, news, editorials and the likes on nanomedicine addressing basic issues of clinical applications. Nonetheless, all that “Nanoscience” and the associated fields are trying to convey is that, in solving problems, “smaller” (specifics or details) is better than “bigger” (generalization or broad). In nanotechnology, the fact is that science has employed technology to get a job done. It has been observed however, that in the course of research that science and technology, in trying to find a solution to a particular problem or challenge, damage in another form is done [1-3]. This is obvious in the side-effects associated with drugs [4-6]. It has also been quietly noted that the rate of cancer incidence is positively correlated (or directly proportional) to technological advancements. It is obvious that Nanoscience and its associated fields such as nanotechnology and nanomedicine is actually in a relatively adolescence stage. This is, however, not to neglect the advances made to date. Providing an effective and a clear definition to the field of nanomedicine and creating a standardized approach to acquiring, sharing, and tracking relevant information on nanomedicine applications as well as products will give a better focus as to what is achievable in this field. Applications and products that are already in trials or used in humans are also briefly highlighted in this study.

1.1 Defining nanotechnology and nanomedicine

Diverse definitions as to what nanotechnology is perceived to be, have emerged. To put in perspective, “Nanoscience” is broadly viewed as the study of materials and their associated physical, biophysical, chemical and biochemical phenomena on the scale of ~1–100 nm (10-9 m). Though much of the biological processes occur on the nanoscale, we cannot say that Nanoscience has always existed. It is therefore expedient to clarify what this concept is in its entirety. Nanotechnology is a field that employs: technological tools and concepts to biology; engineering biological and chemical molecules toward acquiring functions that are different from natural processes; or manipulating biological systems via methods more precise than the conventional molecular biology, biochemical or synthetic chemical approaches. Nanotechnology has proved useful in a lot of scientific and technological breakthroughs and a few of such obvious strides include:

  1. Mechanical, heat and electrical energy derived from the heart beat and the tapping of fingers can be converted into electrical energy through nanotech materials, which can be used to recharge portable devices when built into fabrics of cloth.

  2. Nanomaterials have also been proven to stimulate the body into regenerating lost as well as damaged cells. This has been demonstrated in mice that were able to use paralyzed limbs again.

  3. Some of the major challenges of cancer treatment include off-target sites and the side-effects of treatments that can now be circumvented by the development of nanotech-coated therapeutics that has the ability to guide the drug to the target while ignoring healthy cells.

  4. Blades that can cut through stones and metal can now be made from steel containing nanotubes. This steel can last for a thousand years as evident in the popular Damascus steel

  5. Scientists can now build outfits for rescue operations on smooth surfaces of building structures without any external support. The inspiration for this was derived from geckos that possess nano-sized hairs that can fuse to smooth surfaces without falling off.

  6. The creation of an invisibility cloak, where nano-tube sheets can allow light to divert away from objects that “disappear” at extreme temperatures.

  7. Suits that are stab-resistant and bullet proof capable of stopping 9mm rounds are now made from carbon nanotubes. They are so resilient that they have to be cut into different shapes and sizes using a saw.

  8. Nanotechnology is also being employed in energy (including solar cells and -performance batteries), electronics with ultra-storage capacity [7], smart nutrients delivery, screening for contaminants [8], and single-atom transistors [9].

Even though the core definition of nanotechnology as well as nanomedicine continues to be an area of controversy without a universally agreed definition, we still have some acceptable descriptions as well as operational definitions that show what we are talking about. Etheridge et al., 2013 suggested that nanomedicine should be taken as the medical application of nanoscale or nanostructured materials that have been engineered to have unique medical effects based on their structure with at least one characteristic dimension up to 300 nm as against the common 100 nm, and tries to buttress this definition with the fact that many of the early definitions of nanotechnology took a cut-off around 100 nm. Giving the definition provided by the National Nanotechnology Initiative (NNI) as an instance, earlier definitions that established 300 nm as limit focused more on where quantum effects are most times restricted to structures on the order of one to tens of nanometers [10-12]. In addition to this defense, spectacular physiochemical behaviors sometimes show-up for nanomaterials with defining features exceeding 100 nm. For instance, the plasmon-resonance in 150 nm diameter gold nanoshells was under clinical investigation for cancer thermal therapy. Etheridge et al., 2013 concluded by saying that, although many applications of nanomedicine utilize feature sizes ≤100 nm, this cut-off rules out many applications with significant consequences hence, the choice of 300 nm to better encompass the unique physicochemical properties that is occurs on these scales.

2 The nature of nanomaterials

Engineering nanomaterials is the most rapidly growing branch of nanosciences, and their physicochemical properties prove to be more advantageous than the bulk materials that have the same compositions. Nanomaterials exhibit distinct novel attributes that the same material at large size may not possess. Carbon lattices, metal oxides, micelles, liposomes, nanotubes and polymers, and many more examples of nanomaterials are made of diameter of less than 100 nm (0.1μm). Particle aggregates are capable of being broken down to even smaller particles through milling and/or dispersion which generates nanoscale particles rather than solvated materials [13]. The relatively increased surface area, the quantum effects (in some cases), and the nano-scale size are the three principal factors that give nanomaterials their distinct properties that distinguish them from the bulk materials. When mass is kept unchanged, miniaturizing nanoparticles will produce an increase in the total surface area of particles, generating additional physico-chemical properties that puts the further miniaturized nanoparticle ahead of the corresponding bulk [14-16]. For example, a particle with a diameter of 1000 nm should have 1015 number of particles per pound, and a surface area of 3×109mm2 per pound. Comparatively, a particle with a diameter of 10 nm should have 1021 number of particles per pound, and a surface area of 3×1011 mm2 per pound. Additionally, the translocation/pharmacokinetics and distribution of nanoparticles through the body is largely and unequivocally dependent on size. Hence, the adverse or unintended effects of miniaturized nanoparticles cannot actually be easily detected or predicted from the known toxicity of the same chemical constituent when it is still at the bulk size or at micro-size [14, 17, 18]. The main factor that makes nanomaterials suitable for pharmaceutical applications is their unique size. However, size, hydrophobicity, 3D shape, electronic configuration, photo/electro-chemistry amongst many other attributes are part of what makes us find nanomaterials useful and applicable to medicine. These attributes affect the absorption, distribution, metabolism and excretion (ADME) of engineered nanoparticles in vitro and in vivo. Taking out the size factor will limit the use of nanomaterials.

We can subdivide nanoparticles into two major groups:

  1. The labile nanoparticles. This group is majorly but not limited to organic products, and they are characterized by disintegration into many molecular components upon application. Such nanoparticles include liposomes, polymers, micelles, and nanoemulsions.

  2. The insoluble nanoparticles. This group is majorly but not limited to inorganic products, such as silica dioxide (SiO2), titanium dioxide (TiO2), fullerenes and quantum dots (QD) (e.g. carbon lattices, nanotubes, metal oxides). In essence, these remain intact.

The metabolic fate of nanoparticles in the body system to a great extent depends on the surface characteristics of any particular nanomaterials. The pharmacokinetic behaviors and biological effects associated with the different class of nanoparticles require serious investigation at cellular levels as there is no single universal “nanoparticle” that is fit for all kinds of needs. Each nanomaterial is therefore to be investigated and evaluated for the entire effects associated with the whole therapeutic process.

3 Application of nanotechnology in medicine

Its application in health and medicine is however, receiving more interest now, and focuses extensively on pharmaceuticals (figure 1) [19], cancer treatment [20], tissue regeneration [21], implantable materials and medical diagnosis and imaging [22, 23]. So much has also been emphasized about nanomedical applications, and it is ideal to point out that one of the major attributes of these materials is in the cytotoxic effects of certain engineered nanomaterials in destroying malignant cells/tumours. Wagner et al. summarized the findings of a 2005 study commissioned by the European Science and Technology Observatory (ETSO) [24], through which products that are in development and have gained approval through one stage or another have been clearly spotted (table 1). The study also took companies involved, economic potential and data on developing applications into consideration for some of the studies (table 2). There are five (5) basic stages that are involved from development to the use of nanomaterials in medicine. The stages are: basic nanoscience research, application development, preclinical study (animal models), clinical trials phases, and finally consumer commercial products.

Figure 1 Nanomaterials and their application in pharmaceuticals
Figure 1

Nanomaterials and their application in pharmaceuticals

Table 1

Products that are in development and have gained approval through one stage or another.

Product (s)/application (s)Source/CompanyNanocomponentMechanism of targetDisease conditionsBroad Use
MBP-Y003, MBP-Y004, MBP-Y005[25]Mebiopharm Co., LtdloposometransferrinlymphomaLymphoma
SGT-53 [26, 27]SynerGene therapeutics, Inc.LiposomestransferrinSolid tumors
Ontak [25, 28]Seragen, Inc.Protein NanoparticleIL-2 ProteinT- Lymphoma
MCC-456 [29, 30]Mitsubishi Tanabe Pharma CorpLiposomeGAH AntibodyStomach cancer
MBP-426 [29, 31]Mebiopharm Co., LtdLiposometransferrinSolid TumorsSolid tumors
CALAA-01 [26, 32]Calando PharmaceuticalsNanoparticletransferrinSolid tumors
DM-CHOC-PEN [33]DEKK-TEC, Inc.EmulsionPenetrate Blood-Brain-BarrierBrain Neoplasms
SapC-DOPS [34]Bexion Pharmaceuticals, Inc.LiposomeSaposin CSolid tumors
AS15 [27]GlaxoSmithKline BiologicalsLiposomedHER2 AntibodiMetastatic Breast Cancer
PK2 [29, 35]Pharmacia & Upjohn Inc.Polymeric NanoparticleGalactoseLiver cancer
Actinium-225-HuM195 [27]National Cancer InstituteNanoparticleHuM195 Anti-bodyLeukemia
Rexin-G, Reximmune-C [36]Epeius BiotechnologiesNanoparticlesVon Willebrand factor (Collagen-Binding)Solid tumorsSolid Tumors
Aurimune (CYT-6091) [26, 37],CytImmune Sciences,Colloid GoldTNF-αSolid tumors
Auritol (CYT-21001) [38]Inc.
Targeted EmulsionsKereos, Inc.Emulsion“Ligands”In Vivo Imagine
Opaxio [39, 40]Cell therapeutics, Inc.Polymeric NanoparticlesEnzyme-ActivatedSolid tumors
thermoDox [41]Celsion CorporationLiposomethermosensitivSolid tumors
Nanodots (Website, “Nanoco Technologies.”)Nanoco Group PLCQuantum DotFluorescent EmissionIn vivo imagingIn vivo imaging
FeraSpin (Website, “Miltenyi Biotec – FeraSpin™.”)Miltenyi BiotecIron-Oxide NanoparticleEnhanced MRI ContrastIn vivo imaging
Qdot Nanocrystals (Website, 2010, “Qdot® Nanocrystal Technology Overview.”)Invitrogen CorpoarationQuantum DotFluorescent EmissionIn vivo imaging
eFluor Nanocrystals (Website, “eBiosciences: eFluor® Nanocrystals.”)eBiosciencesQuantum DotFluorescent EmissionIn vivo imaging
TriLiteTM Nanocrystals (from: “Crystalplex Quantum Dots, Semiconductor Nanocrystals & Encoded Polystyrene Beads.”)Crystalplex CorporationQuantum DotFluorescent EmissionIn vivo imaging
NanoHC (Website, “DiagNano – Technology | DiagNano: Revolutionizing Diagnostics through Nanotechnology.”)DiagNanoQuantum DotFluorescent EmissionIn vivo imaging
Clariscan (Website, “Veridex, LLC, a Johnson & Johnson company in vitro diagnostics oncology.”)NycomedIron-Oxide Nano-particle1- Enhanced MRI ContrastIn vivo imaging
NanoHC (Website, “DiagNano – Technology | DiagNano: Revolutionizing Diagnostics through Nanotechnology.”)DiagNanoQuantum DotFluorescent EmissionIn vivo imaging
Resovist [42, 43], Supravist [44]ScheringIron-Oxide Nano particle1- Enhanced MRI ContrastIn vivo imaging
CellSearch®Epithelial Cell Kit (Website, “Veridex, LLC, a Johnson & Johnson company in vitro diagnostics oncology.”)Veridex LLC (Johnson & Johnson)Iron-Oxide Nano particleMagnetic SeparationIn vivo imaging
NanoDX (Website, “T2 Biosystems - NanoDx Nanoparticles.”)T2 BiosystemsIron-Oxide Nano particleMagnetic SeparationCell SeparationIn Vitro Cell Separation
NanoXray (45)NanobiotixProprietary Nano particlesX-Ray Induced ElectronSolid tumorSolid tumor treatment
Endorem, Lumirem, Sinerem [42, 43]GuebertIron-Oxide NanoparticleEnhanced MRI ContrastSolid tumor
Feridex IV, Gastromark Combidex (ferumoxtran-10) [42; 43]Advanced MagneticsIron-Oxide NanoparticleEnhanced MRI ContrastSolid tumor
AuroShell ([46]Nanospectra BiosciencesGold nanoshellIR Laser HeatingSolid tumorSolid Tumor hyperthermia
NanoThern [45]MagForce Nanotechnologies AGIron-Oxide NanoparticleAC Magnetic HeatingSolid tumor
Targeted Nano-Therapeutics (“Technology - Aspen Medisys, LLC.”)Aspen medisys, LLCIron-Oxide NanoparticleAC Magnetic HeatingSolid tumor

Table 2

Nano-products approved for use, with dates.

Product(s)/application(s) plus year of approvalSource/CompanyNanocomponet DescriptionBroad use
Fresenius Polysulfone®, Helixone® (1998) (“NephroCare: Fresenius Polysulfone® Helixone® Dialysers.”)NephroCareNanoporous MembraneDialysis Filter
Filtek (2008) [47]3M CompanySilica and Zrconium NanoparticlesBone substitute
OsSatura (2003) [48]Isotis Orthobiologics USHydroxapatite Nanocrystals
Vitoss (2003) [24]orthovita100 nm calcium-phosphate nanocrystals
NanOss (2005) [45]Angstrom medica, Inc.Hydroxapatite Nanocrystals
Ceram × Duo (2005) (“Ceram-X: Nano Ceramic Restorative.”)DentspleyCeramic Nanoparticles
Ostim (2004) [49]osartis20 nm Hydroxapatite
Alpha-bsm, Beta-bsm, Gamma-bsn, EquivaBone, CarriGen (2009) (“ETEX products are composed of a proprietary nano-crystalline calcium phosphate formulation that mimics the crystalline mineral structure of human bone.”)ETEX CorporationHydroxapatite Nanocrystals
Acticoat® (2005) [50, 51]Smith & Nephew, IncAnit-Microbial NanosilverMedical Dressing
ON-Q SilverSoaker/SilvaGardTM (2005) [50]I-Flow Corporation/AcryMed, Inc.Anti-Microbial NanosilverDental composite
Nano-Bond (2007) (Nano-Bond: Nano Particulate Reinforced Adhesive System, Pentron Clinical Technologies, LLC.)Pentron® Clinical Technologies, LLC“Nanoparticles”
Premise (2003) [24]Sybron Dental Specialties“Nanoparticles”
CellTracks® (2003) [24]Immunicon CorporationMagnetic NanoparticlesDevice Coating
NanoTite Implant (2008) (Website, “BIOMET 3i Implant Systems - NanoTite™ Implant - Introduction.”)BiometCalcium phosphate Nanocrystal Coating
Stratus CS (2003) [48]Dade BehringDendrimers
EnSeal Laparoscopic vessel Fusion (2005) (“Nanotechnology and Medicine / Nanotechnology Medical Applications,” The Project on Emerging Nanotechnologies)Ethicon endo-Surgery, Inc.Nanoparticle Coated Electrode
NicAlert (2002) [24]NymoxColloidal Gold
TiMESH (2004) (“Nanotechnology and Medicine / Nanotechnology Medical Applications,” The Project on Emerging Nanotechnologies)GfE Medizintechnik GmbH30 nm Titanium CoatingTissue Scarfold
MyCareTM Assays (2008) (Website, “Saladax Biomedical | Product Pipeline Assays.”)Sladax Biomedical“Nanoparticles”In Vitro Assay
Verigene (2007) [52, 53]Nanosphere, IncColloidal Gold
cellSearch® Epithelial Cell Kit (2004) (Website, “Veridex, LLC, a Johnson & Johnson company in vitro diagnostics oncology.”)Vendex, LLC (Johnson & Johnson)Iron-Oxide Nanoparticles

3.1 Nano-science and the justification for nanomedicine

Two fields of medicine, regenerative medicine and personalized medicine, are currently seeking refuge in nanomedicine as an alternative. Medicine itself is at the mercy of diverse fields of science that have individual origins. We can actually say that medicine is a discipline that rides on the strength and knowledge of other scientific and technological disciplines by harnessing the potentials in each of these fields of study. These scientific and technological fields include: the Natural sciences, Life sciences, Biomedicine, Molecular biology, Proteomics, Genomics, Biotechnology, Pharmacology, Nanotechnology, Nanomedicine, and many more fields. The evolution and convergence of the various related fields into nanomedicine is schematically depicted in figure 2. In the midst of varied descriptions of nanomedicine, the European Science Foundation in 2004 proposed nanomedicine to be the field of science and technology of diagnosing, treating and preventing disease and traumatic injury, of relieving pain, and of preserving and improving human health, using molecular tools and molecular knowledge of the human body [54]. Since nanomaterials and the associated nanoscale devices are generally and typically comparable in size to DNA, viruses, subcellular organelles, proteins and gap junctions [55, 56], nanomedicine and other fields of medical research such as molecular medicine need to be strictly differentiated on the basis of phenomena at the nanoscale [57]. Defining Nanomedicine will not be a difficult one, as it is simply the application of nanotechnology to medicine. Summarily, nanomedicine is described as the science and technology of diagnosing, treating and preventing diseases towards ultimately improving human health. The applications of nanoparticles in nanomedicine actually stems from the enormous variety of attributes and unique characteristics of the nanoparticles under consideration. These characteristics are used in biomedical applications in the amplification of effects at the molecular level, by creating the required local concentrations of therapeutics. Currently, a lot of options are being considered in the use of nanomaterials in medicine, and some of them have received approval by various appropriate regulatory agencies such as the FDA. A few examples are briefly stated. Dendrimers capable of controlling drug delivery during gene transfection and also neutron-capture therapy are being investigated as potential therapeutics. The ability to track and monitor the therapeutic process such as tracing Quantum Dots (QDs) microscopically as well as at ultrastructural levels via the principles of probes is largely instrumental in cellular dynamics. QD (most commonly Cd–Te, Zn–S, Cd–S, Pb–S and Cd–Se nanocrystals) probes are actually nanometer-sized (with 5–10 nm suggested to be more photo stable than the usual fluorophores) semi-conductors fortified with fluorescent property that make it suitable for biological imaging of molecules to which they are tagged [58-60]. Photoexcited TiO2, capable of generating strong reactive oxygen species such as OH- and H2O2 is being investigated for the capability of suppressing the growth of tumor cells implanted in nude mice, potentiating for cancer therapy, including treatment of superficial cellular tumors in organs (such as trachea, urinary bladder, skin and oral cavity) not prone to damage by exposure to light. It is also vital to state at this point that the National Institute for Enviromental Health and Safety (NIEHS), the National Cancer Institute (NCI) led a National Nanotechnology Initiative, and the National Institute of Health, USA, have been trying to initiate, fund and foster preclinical toxicology studies to consider all the possible safety issues that could propel nanomedicine into clinical practice. The use of nanodrugs creates a dose differentiation between site of the target and the rest of the body, thereby maximising the therapeutic effects, while minimizing side-effects [62]. This is in contrast with the traditional methods used.

Figure 2 Evolution and convergence of some diverse related scientific and technological disciplines in nanomedicine.
Figure 2

Evolution and convergence of some diverse related scientific and technological disciplines in nanomedicine.

4 Nanomedicine in therapeutic medicine

The nanocarriers employed in nanomedicine are made of relatively safe materials, including but not limited to synthetic biodegradable polymers, polysaccharides and lipids. The essence of this is that, medicine tries as much as possible to stick closer to natural organic products as well as biodegradable natural products rather than inorganic and synthetic products. Various fields that play a part in therapeutic nanomedicine are depicted in Table 3.

Table 3

Diverse fields in therapeutic medicine

Nanomedicine in Therapeutic (treatment) Medicine
Molecular Biology & ProteomicsPharmaceuticsBiomedicine, Biorobotic & RoboticsBiotechnology
– Genetic therapy– Drug delivery– Enzyme manipulation & control– Tissue engineering
– Pharmacogenomics– Biopharmaceutics– Biological viral therapy– Artificial organ
– Drug encapsulation– Artificial cells & liposomes– Stem cell and cloning
– Smart drugs– Polymeric micelles & polymersomes– Virus-based hybrids
– Radiopharmaceutics – Dynamic nanoplatform nano-some
– Antibacterial & antiviral nanoparticles– MEMS surgical devices
– Photodynamic therapy– Sensory aids (e.g artificial retina)
– Fullerene-based pharmaceuticals– MEMS/Nanomaterials-based prosthetics
– Target drug delivery– Diamond-based nanorobots

Multifunctional nanoparticles play key roles in cancer therapeutics. Abraxane, one of the therapeutics approved by the FDA for the treatment of breast cancer, is a nano formulation made of paclitaxel (a very potent anticancer drug) conjugated to a nano-bead protein structure. The nano-bead conjugated protein moiety facilitates water solubility to enhance the elimination of the toxicity of cremaphor (the solvent vehicle) towards ultimately improving the therapeutic index. Likewise, nanoparticles (e.g TiO2) have been employed as optical filter carriers to protect the drug against light exposure damage and ultimately improve the photostability of pharmacologically active payload. There is also an improved hydrophilicity of TiO2, producing increased aqueous wettability and dissolution that disallows aggregation in the dispersion medium [62].

Carbon nanotubes (CNTs) are also a very important class of nanomaterials. Of the carbon nanotubes, the single-walled carbon nanotube (SWNT), usually linked covalently to the therapeutic drug of diagnostic sensor via ester disulfide bond, is of a very special interest because of its hydrophilic functionalized soluble nature. That grants it a wide application as a viable means of moving molecular cargo of different sizes and types across the highly selective cell membrane to the intended therapeutic target via various energy -dependent and/or –independent processes [63-65]. SWNTs are capable of binding to single/ double stranded DNA, as well as peptide amino acids (PAA) [66-69]. When SWNTs enters the target compartment, they come in contact with the acidic environment of endosomes or lysosomes which has a reducing property, leading to the cleavage of the disulfide/ester bonds to selectively release the active therapeutic agent [70]. It has been demonstrated that nanoformulations of SWNTs conjugated to platinum (IV) could effectively deliver a lethal dose of the anti-cancer drug inside the malignant cancer cells [71]. Cationic functionalized CNTs (usually positively charged polyelectrolyte), are often employed in binding to DNA to facilitate specific target-delivery of functional DNA and siRNA in target gene expression studies and experiments. These Cationic functionalized CNTs nanotubes are intended to balance-out negative charge on the DNA, and this has helped generate lots of DNA sensors [72]. One of the vital procedures that must be well observed in putting a methodology into practice is the ability to track and monitor biological molecular processes in vivo and in vitro, and water-soluble semiconductor Quantum dots (QDs), such as cadmium selenide (CdSe) have proven to be effective in functioning in this capacity. This is achieved by capping CdSe with a shell of PEG, with a functional biotin surface.

Likewise, the administration of doxorubicin encapsulated in PEG-PE micelles improves doxorubicin accumulation and penetration in tumors, in comparison with what is obtainable without nanotechnological modification. Liposomes have been long used as viable transporters for therapeutics such as cisplatin, doxorubicin, paclitaxel, daunomycin and amphotericin, providing a practical approach for on-target and in time delivery aimed at reducing the risk of adverse effects as well as increasing bioavailability [73].

Doxil® and Abraxane are two examples of “first generation” nano-based drugs that have been fashioned for cancer chemotherapy, functioning in their capacity due to their physicochemical behavior on the nanoscale [74, 75]. Cancer cells can be effectively targeted due to the enhanced permeation and retention effect made feasible by this delivery system [76-78]. Approximately 5%–10% of all cancer drugs that are in clinical use today are nanodrugs. The solution to various infectious diseases has also been sought using nanotechnology, and they include endocrine/exocrine disorders, skin regeneration, cardiac/vascular disorders, corrective cochlear and retinal implants, degenerative disorders [10], increased efforts in sustained drug release in treating glaucoma [79, 80] improve heart function, and improved antimicrobials [81]. Most of the currently registered clinical trials (Table 4) in nanomedicine primarily focus on therapeutic approaches in managing malignant solid tumors.

Table 4

Some clinical trials in nanomedicine

Clinical trial IdentifierArea of applicationNanotherapeutic/nanodiagnosticsMain focus of study
NCT02104752Schizophrenia; cognition; psychosisTheracurmin formulation of curcumin nanoparticlesCurcumin as a Novel Treatment to Improve Cognitive Dysfunction in Schizophrenia
NCT02033447Prostate cancerMagnetic Nanoparticle injectionMagnetic Nanoparticle Thermoablation-Retention and Maintenance in the Prostate: A Phase 0 Study in Men (MAGNABLATEI)
NCT01167985Endodontic treatment; irreversible pulpitis; infected pulp; etcDevice: IABNA Clinical Study: the Antibacterial Effect of Insoluble Antibacterial Nanoparticles (IABN) Incorporated in Dental Materials for Root Canal Treatment
NCT02507596Chronic periodontitis

Biological: nanocrystalline hydroxyapatit Silica Gel

Biological: bone allograft

Procedure: open flap debridement

e Evaluation of Nano-crystalline Hydroxyapasilica gel tite Silica Gel in Management of Periodontal Intrabony Defects
NCT01763710Breast CancerPaclitaxel 80 mg/m2Neurotoxicity Characterization Study of
Drug: Nab-paclitaxel 100 mg/m2 days 1,Nab-paclitaxel Versus Conventional Paclitaxel
8 and 15in Metastatic Breast Cancer
Drug: Nab-paclitaxel 150 mg/m2 days 1,
8 and 15
Drug: Nab-paclitaxel 150 mg/m2 days 1
and 15
NCT02549898Migraine headache;Drug: FerahemeInvestigation of Vascular Inflammation in
migraine without auraDrug: cilostazolMigraine Using Molecular Nano-imaging and
Other: USPIO MRIBlack Blood Imaging MRI
Other: BBI MRI
NCT01995799Myocardial infarction;Device: ferumoxytol-enhanced MRIIRon Nanoparticle Enhanced MRI in the
inflammationAssessment of Myocardial infarctioN
(IRNMAN)
NCT00711230HIV InfectionDermaVir: PlaceboRepeated DermaVir Immunizations in HIV-1
Infected Treatment-naϊve Patients
NCT02479178Urothelial carcinoma; cholangiocarcinoma; cervical cancer; squamous cell carcinoma of head and neckDrug: BIND-014 (docetaxel nanoparticles for injectable suspension)A Study of BIND-014 in Patients With Urothelial Carcinoma, Cholangiocarcinoma, Cervical Cancer and Squamous Cell Carcinoma of the Head and Neck (iNSITE2)
NCT01929785Cardiac Transplant FailureApplication of In-House Developed Nanomedicine Technology for Diagnosis and Management of Post-Transplant Heart Allograft PatientsBIIR Gene to Manage Heart Allograft Patients
NCT00712530HIV InfectionDermaVir|Drug: HAARTSingle DermaVir Immunization in HIV-1
Infected Patients on HAART
NCT01463072Male breast cancer; recurrent breast cancer; stage IV breast cancer; etcPaclitaxel albumin-stabilized nanoparticle formulationPaclitaxel Albumin-Stabilized Nanoparticle Formulation in Treating Older Patients With Locally Advanced or Metastatic Breast Cancer
NCT00918840HIV InfectionDermaVir: Placebo|Drug: HAARTAntiretroviral-Sparing Concept With HIV-specific T Cell Precursors With High Proliferative
Capacity (PHPC)
NCT02006108Renal transplant rejectionDrug: FerahemeImaging Kidney Transplant Rejection Using
Other: MRI-GEHealthcare 3 Tesla magnetFerumoxytol-Enhanced Magnetic Resonance
NCT02755870Healthy Volunteers - MaleCNM-Au8|Other: PlaceboA Phase I SAD and MAD Clinical Trial of
and FemaleCNM-Au8 in Healthy Male and Female Volunteers
NCT02467673Menopausal syndromeDrug: micronized estradiol + progesterone Drug: nanoparticulate estradiol + progesteroneNanoparticulate Versus Micronized Steroids Delivery for Transdermal Hormone Replacement Therapy (Nanoparticle)

4.1 Nanomedicine, stem cells, cancer, immune activation management, neurological diseases and regenerative medicine

Cancer is the major target of nanomedical research in the development of individualised diagnoses and treatments. Common anti-cancer agents have a poor biodistribution, and poor pharmacokinetics, creating adverse effects on healthy tissues [82]. By identifying disease biomarkers such as a protein, mutant gene, lipid, carbohydrate RNA, or small metabolites, targeted treatments can be designed using multifunctional nanoparticles that are able to detect and image tumours [83]. Some of the nanobased anti-cancer drugs currently used to treat various forms of cancer are reported in table 5. The broad mechanisms of the delivery of nano-therapeutic materials to cancerous tissue progresses generally via passive or active nanomedicine drug targeting. The passive targeting occurs due to the spontaneous drug accumulation in cancerous sites which possesses typically leaky vasculature. This ultimately provides enhanced permeation and retention effect [84]. Active targeting methods, on the other hand, exploits or takes advantage of the fact that, at the cellular level, a large number of pathological states may be identified by particularly displayed surface proteins that are absent in healthy cells. In some cases, these proteins are relatively over expressed when compared to healthy cells. This surface functionalization with ligands, which bind selectively to these proteins that are expressed on the surface of the unhealthy tissue, makes provisions for specific targeting of the nano-based therapeutics to the target diseased site [85]. The first sets of nanoparticles used for cancer treatment were biodegradable and biocompatible liposomes capable of carrying both hydrophobic and hydrophilic molecules [86]. They consist of a lipid bilayer surrounding an aqueous core, encapsulating the drug [83], highly endowed with the ability to permeate cell membranes easily to bind to DNA or RNA for transfection [86]. Dendrimers are also used for a range of therapeutics and diagnostics by carrying drugs either by encapsulation or conjugation in cancer, and in photodynamic therapy and hyperthermia therapies using AuNPs. Another landmark on the progress to curing cancer has been cancer stem cells (CSCs). They have similar properties to normal stem cells, but found within the tumour, therefore are tumorigenic, generating tumours through the stem cell processes. Targeting of CSCs within a tumour permits two types of targeting; active targeting that uses the binding of a specific target ligand to a specific receptor molecule, and passive targeting that relies on haemodynamic changes conventionally linked to the blood supply to tumours. Metastasis, tissue invasion, growth self-sufficiency, sustained angiogenesis, evasion of apoptosis, endless potential for replication and insensitivity to anti-growth signals, are all major hallmarks of cancer cells, and are hinged on a complex set of errors in the governing regulatory circuitry that defines homeostasis. In addition to their perfectly suited preferential transport of therapeutic agents to target cancer sites, nano- and micro-particulates are really ideal probes for study. Transport within and across body compartments and biological barriers differentiates healthy tissues and cancer cells [87]. This has been tested using variants of nanoparticles that can be manufactured in combinatory large sets, so that varying only one designed parameter at a time can explain the effects. This is mostly guided by a careful form of logic usually referred to as the Logic-Embedded Vectors “(LEV)” which forms ideal probes in application to nanoparticles for the determination of the laws of mass transport in and across tumors. This is to function as an imaging contrast enhancer. The full comprehension of the transport differentials expressed in cancer, often termed ‘transport oncophysics’ redefines a new hopeful frontier in oncology, producing greater efficacy in addition to reduced adverse effects. The Logic-Embedded Vectors “(LEV)” actually helps in boycotting the challenges of the barriers to transport of nanoparticles from the site of administration to the subcellular localization in target cells, fostering specificity when the biosensors are incorporated. The specific and preferential targeting and locating of cancer sites by the administration of nanoparticles has greatly evolved as an attractive treatment strategy with a greater therapeutic index, topping the conventional formulations and designs of the same agents [87-91]. The first designed nanodrugs which were approved in the mid-Nineties were doxorubicin [92, 93] and liposomal encapsulated antifungals [94]. Another design employed the conjugation of albumin nanoparticles to Cremaphor®-free taxanes, and secured FDA approval in 2005 for the treatment of Breast cancer [95, 96]. The chaperoning effect of albumin is largely credited to the improved therapeutic index of taxane producing an enhanced extravasation in target tumors. It is recorded that Nanoparticles commands the market in excess of US$ 5.4 Billion per year [97]. Before nanotechnology emerged in medicine, radiopharmaceuticals [98], liposomal drug carriers [98, 99] antisense oligonucleotides, receptor-specific delivery of immunotherapy [100], and imaging contrast agents [101], have been used in the treatments of what nanoparticles have now proven better when properly employed. The ‘first generation’ nanotherapeutics have no particular biomolecular target recognition specificity, and they include the highly recognized clinically-approved liposomes as well as the albumin nanoparticles. Second-generation nanoparticles are the main focus of current research and are expected to perform other functions beyond the first generation. The main focus here is ‘active targeting’, that is the biomolecular targeting of cancer cell surface epitopes via nanoparticle surface-appended recognition moieties, such as peptides, antibodies or aptamers. This has been the focus for some years.

Table 5

Some of the nanobased anti-cancer drugs currently used to treat various forms of cancer

DrugNano-drugTarget/action
Tretinoin [102]Loaded nanocapsules; LiposomesAcute promyelitic leukaemia
Cisplatin [103]DMPG-complexed, entrapped in liposomesMelanomas
Mitoxantrone [103]Cytostatic complex with lipophilic acidMetastatic breast cancer, acute myeloid leukaemia, non-Hodgkin’s lymphoma, acute lymphoblastic leukaemia
Daunomycin [104]DaunoXome (liposome)Leukaemia
Doxorubicin [103]Doxil/ Caelyx-encapsulation in PEGylated liposome Myocet-non-PEGylatedAcute leukaemia, lymphoma, breast carcinoma, osteosarcoma, haematological malignancies, Kaposi’s sarcoma Myocet-metastatic breast cancer in
Liposomecombination with cyclophosphamide
Paclitaxel/Taxol [105]Lipophilic prodrug paclitaxel oleate in sterically stabilizedLung, ovarian, and breast cancer
Annamycin [103]LiposomeLeukaemia, reticulosarcoma
Vincristine [106]LiposomeChemotherapy-nephroblastoma, lymphoma, lymphoblastic leukaemia
Hydroxyrubicin (lipophilic prodrug) [103]LiposomeLeukaemia, reticulosarcoma

One major challenge in therapeutic medicine is tissue or organ rejection during transplant. Nanosized porous materials have been employed in immunoisolation of transplants to overcome this challenge. Via an encapsulation mechanism, semi-permeable membranes allow for the transport of oxygen and nutrients between the host environment and the transplant (cells), but blocks bigger immune cells such as monocytes and leukocytes from attacking the transplanted cells [107].

The Blood Brain Barrier (BBB), composed of brain endothelial cells, functions as the separation of the brain from the other parts of the systemic circulation, and being the main entry route of drugs into the CNS is a major limitation in the treatment of CNS diseases. This restriction of transport is via tight junctions, and through enzymes functioning as metabolic barriers [108]. All these factors are to be considered when designing nanocarriers. However, once molecules overcome the BBB to the cerebrum, they are extensively and quickly distributed throughout the brain, due to the high vascular density in the brain. For instance, modified liposomes have the record of being used in the management of neurodegenerative disorders. It is important to note that while liposomes without modification are usually incapable of crossing the BBB, PEGylated liposomes that are conjugated to monoclonal antibodies to glial fibrillary acidic protein (GFAP) would cross the BBB at the disease site [109]. There are also other methods of making liposomes transportable across BBB, including coupling it to transferring insulin receptors or mannose.

A very important field of medical research known as regenerative medicine describes the regeneration of lost or damaged tissue cells that are no longer functional. It focuses more on the regeneration of cells in the central nervous system (CNS). Diseases such as heart failure, pancreatic cells diseases, stroke, Parkinson’s disease, Alzheimer’s disease, paralysis due to spinal cord injuries and Multiple Sclerosis [110], can be restored in processes encompassed by regenerative medicine (Table 6). This includes regeneration of teeth, and musculoskeletal elements of tendon, bones, cartilages and ligaments [110]. PLGA and Nanosized titanium implants have been used extensively to bringing this to fruition [111].

Table 6

Some of the nanobased neurological drugs currently used to treat neuronal diseases

DrugNano-drugTarget/action
5-chloro-7-iodo-8-hydroxyquinoline CQ [112]Polymeric encapsulationCu/Zn chelator - Alzheimer’s Disease
D-penicillamineNanoparticle encapsulationReverse metal induced
(Cu(I) chelator) [112]Preceptiation
H0-1 gene [113]Reducible poly (oligo-d-arginine) peptideProtect brain cells from IR related injury, including stroke
Thioflavin-T [112]Butylcyanoacrylate polymer encapsulationAlzheimer’s – detection of A_plaques
BCNU [114]Magnetic nanoparticle with Fe3O4 coreGlioma
Mito-Q10 [115]Attached to triphenyl phosphoniumParkinson’s disease
Doxorubicin [116]PEGylated dendrimer with transferrinG6 Glioma cells

5 Conclusion

Nanotechnology which involves engineering and utilizing materials at the nano-scale has shown a lot of promise in medicine and therapeutics. Nanomedicine, being a product of nanotechnology, is the process of using molecular tools and molecular knowledge of the human body for the purpose of diagnosing and treating illness. It may certainly change the ability to screen, diagnose and treat medical conditions that has hitherto posed as threats to human health. Medical conditions such as cancer, cardiovascular disease, endocrine diseases like diabetes, and other infections, may find cures through nanomedicine. Nanotechnology has already found useful applications in drug delivery e.g. delivery of chemotherapy drugs directly to cancer cells, drug release when sheer force is applied through sections of artery. Nanoparticles that deliver vaccine, sponge-like matrix delivering insulin, and nanoparticle enzymes that prevent reproduction of viruses are part of applications of nanotechnology in medicine. Other areas to which nanomedicine has been found useful include: nanotube-based biosensing devices that provide in vivo diagnostic testing capabilities, such as tracking electrolyte and blood glucose levels, synthetic biomaterials that mimic body tissues thereby enabling organ regeneration, faster and accurate infection and genetic testing tools that are less invasive than conventional methods. Some nanomedicine drug-delivery systems are already in use in anti-cancer drugs as already stated. Although nanomedicine has shown many possible breakthroughs in the management of diseases, it is necessary to employ some precautionary measures to protect the health and safety of patients, workers, the public and the environment. Areas that may require more research so as to avert possible future problems include the investigation of the toxicity profile of free nanoparticles which may be able to avoid the body’s defenses and interfere with fundamental natural processes. Others include genetic alteration due to operation of some therapies at the chromosomal level which may be harmful to the unborn child, and issues of environmental and workplace safety as nanoparticles are often too small to be curtailed and may accumulate in water, air or plants, with unpredictable effects.

  1. Conflict of interest

    Conflict of interest statement: Authors state no conflict of interest.

References

[1] Smith K. Chronic-pain treatment without side effects. Nature 2008. 10.1038/news.2008.443Search in Google Scholar

[2] Eugen L, Michael JK, Steven W, Dmitri M, Jacques H, Jeremy LJ, Paul L, Eckhard W, Allison KD, Serge C, Brian K, Shoichet LU. Large-scale prediction and testing of drug activity on sideeffect targets. Nature 2012, 486, 361-36710.1038/nature11159Search in Google Scholar PubMed PubMed Central

[3] Odiba AS, Ottah VE, Anunobi OO, Edeke AA, Ukegbu CY, Chukwunonyelum I, Onosakponome I, Joshua PE. Research Progress in Oncology: Highlighting and Exploiting the Roles of Several Strategic Proteins in Understanding Cancer Biology. Open Life Sci 2016; 11: 331–347. 10.1515/biol-2016-0045Search in Google Scholar

[4] Lee S, Lee KH, Song M, Lee D. Building the process-drug-side effect network to discover the relationship between biological processes and side effects. BMC Bioinformatics 2011, 12 (Suppl 2), S210.1186/1471-2105-12-S2-S2Search in Google Scholar PubMed PubMed Central

[5] Odiba AS, Wannang N, Joshua EP, Iroha I, Ukegbu C, Onosakponome Y, Chukwunonyelum I, Anunobi OO. Curing HIV-1 Infection via in vitro Ultra- Sensitive Modification of HIV-1 Uninfected CD4 + Cells using Antiviral Agents Curing HIV-1 Infection via in vitro Ultra-Sensitive Modification of HIV-1 Uninfected CD4 + Cells using Antiviral Agents. Adv. Life Sci. Technol 2014, 24, 1Search in Google Scholar

[6] Odiba A, Ukegbu C, Anunobi O, Chukwunonyelum I, Esemonu J. Making drugs safer: improving drug delivery and reducing the side effect of drugs on the human biochemical system. Nanotechnol. Rev 2016, 1-1310.1515/ntrev-2015-0055Search in Google Scholar

[7] Vettiger P, Cross G, Despont M, Drechsler U, Durig U, Gotsmann B, Haberle W, Lantz M, Rothuizen HE, Stutz R, et al. The ‘millipede’ - nanotechnology entering data storage. IEEE Transactions on nanotechnology. 2002, 1(1):39-5510.1109/TNANO.2002.1005425Search in Google Scholar

[8] Sozer N, Kokini JL. Nanotechnology and its applications in the food sector. Trends in Biotechnology 2009, 27(2):82-89. [PubMed: 19135747]10.1016/j.tibtech.2008.10.010Search in Google Scholar PubMed

[9] De Franceschi S, Kouwenhoven L. Electronics and the single atom. Nature 2002, 417(6890):701–702. [PubMed: 12066169]10.1038/417701aSearch in Google Scholar PubMed

[10] Etheridge ML, Campbell SA, Erdman AG, Haynes CL, Wolf SM, McCullough J. The big picture on nanomedicine: the state of investigational and approved nanomedicine products. Nanomedicine 2013,9:1-1410.1016/j.nano.2012.05.013Search in Google Scholar PubMed PubMed Central

[11] Smith AM, Nie S. Semiconductor Nanocrystals: Structure, Properties, and Band Gap Engineering. Acc. Chem. Res 2010, 43(2):190-200. [PubMed: 19827808]10.1021/ar9001069Search in Google Scholar PubMed PubMed Central

[12] Daniel M-C, Astruc D. Gold Nanoparticles: Assembly, Supramolecular Chemistry, Quantum-Size-Related Properties, and Applications toward Biology, Catalysis, and Nanotechnology. Chemical Reviews 2004, 104(1):293–346. [PubMed: 14719978]10.1021/cr030698+Search in Google Scholar

[13] Jia L. Nanoparticle formulation Increases oral bioavailability of poorly soluble drugs: experimental evidences, theory, and approaches. Curr. Nanosci 2005, 3(1):237-24310.2174/157341305774642939Search in Google Scholar

[14] Oberdorster GOE, Oberdorster J. Nanotoxicology: An Emerging Discipline Evolving from Studies of Ultrafine Particles. Environ. Health Perspect 2005b, 113:823-839. [PubMed: 16002369]10.1289/ehp.7339Search in Google Scholar

[15] Nel A, Xia T, Madler L, Li N. Toxic potential of materials at the nanolevel. Science 2006, 311:622- 627. [PubMed: 16456071]10.1126/science.1114397Search in Google Scholar

[16] Hoet PH, Bruske-Hohlfeld I, Salata OV. Nanoparticles – known and unknown health risks. J. Nanobiotechnol 2004, 2:1210.1186/1477-3155-2-12Search in Google Scholar

[17] De Jong WH, Roszek B, Geertsma RE. Nanotechnology in medical applications: possible risks for human health. 2005RIVM report 265001002/2005Search in Google Scholar

[18] Zhao, Y, Nalwa, HS. Nanotoxicology. California: American Scientific Publishers. 2007. 2007Search in Google Scholar

[19] Sahoo SK, Labhasetwar V. Nanotech approaches to drug delivery and imaging. Drug Discovery Today 2003, 8(24):1112-1120. [PubMed: 14678737]10.1016/S1359-6446(03)02903-9Search in Google Scholar

[20] Nie S, Xing Y, Kim GJ, Simons JW. Nanotechnology applications in cancer. Biomedical Engineering 2007, 910.1146/annurev.bioeng.9.060906.152025Search in Google Scholar PubMed

[21] Engel E, Michiardi A, Navarro M, Lacroix D, Planell JA. Nanotechnology in regenerative medicine: the materials side. Trends in biotechnology 2008, 26(1):39-47. [PubMed: 18036685]10.1016/j.tibtech.2007.10.005Search in Google Scholar PubMed

[22] Jain KK. Nanotechnology in clinical laboratory diagnostics. Clinica Chimica Acta 2005, 358(1-2):37-5410.1016/j.cccn.2005.03.014Search in Google Scholar PubMed

[23] Wickline SA, Lanza GM. Nanotechnology for Molecular Imaging and Targeted Therapy. Circulation. 2003, 107(8):1092-1095. [PubMed: 12615782]10.1161/01.CIR.0000059651.17045.77Search in Google Scholar PubMed

[24] Wagner V, Dullaart A, Bock AK, Zweck A. The emerging nanomedicine landscape. Nature Biotechnology 2006, 24(10):1211-121810.1038/nbt1006-1211Search in Google Scholar PubMed

[25] Faraji AH, Wipf P. Nanoparticles in cellular drug delivery. Bioorganic & medicinal chemistry 2009, 17(8):2950-2962. [PubMed: 19299149]10.1016/j.bmc.2009.02.043Search in Google Scholar PubMed

[26] Bawa R. Nanoparticle-based therapeutics in humans: a survey. Nanotechnology Law & Business 2008, 5(2):135-155Search in Google Scholar

[27] Safety Study of Infusion of SGT-53 to Treat Solid Tumors - Full Text View - ClinicalTrials.gov, ClinicalTrials.gov, SynerGene Therapeutics, Inc.Search in Google Scholar

[28] Kanaoka E. A novel and simple type of liposome carrier for recombinant interleukin-2. J.Pharm. Pharmacol 2001, 53(3):295. [PubMed: 11291744]10.1211/0022357011775523Search in Google Scholar PubMed

[29] Matsumura Y, Kataoka K. Preclinical and clinical studies of anticancer agent-incorporating polymer micelles. Cancer science 2009, 100(4):572-579. [PubMed: 19462526]10.1111/j.1349-7006.2009.01103.xSearch in Google Scholar PubMed

[30] Hamaguchi T, Matsumura Y, Nakanishi Y, Muro K, Yamada Y, Shimada Y, Shirao K, Niki H, Hosokawa S, Tagawa T, et al. Antitumor effect of MCC-465, pegylated liposomal doxorubicin tagged with newly developed monoclonal antibody GAH, in colorectal cancer xenografts. Cancer science 2004, 95(7):608-613. [PubMed: 15245599]10.1111/j.1349-7006.2004.tb02495.xSearch in Google Scholar PubMed

[31] Sankhala KK, Mita AC, Adinin R, Wood L, Beeram M, Bullock S, Yamagata N, Matsuno K, Fujisawa T, Phan A. A phase I pharmacokinetic (PK) study of MBP-426, a novel liposome encapsulated oxaliplatin. J. Clin. Oncol 2009, 27:15s10.1200/jco.2009.27.15_suppl.2535Search in Google Scholar

[32] Heidel JD, Yu Z, Liu JY, Rele SM, Liang Y, Zeidan RK, Kornbrust DJ, Davis ME. Administration in non-human primates of escalating intravenous doses of targeted nanoparticles containing ribonucleotide reductase subunit M2 siRNA. Proceedings of the National Academy of Sciences 2007, 104(14):571510.1073/pnas.0701458104Search in Google Scholar PubMed PubMed Central

[33] Morgan LR, Struck RF, Waud WR, LeBlanc B, Rodgers AH, Jursic BS. Carbonate and carbamate derivatives of 4-demethylpenclomedine as novel anticancer agents. Cancer Chemother Pharmacol 2009, 64(4):829-835. [PubMed: 19255760]10.1007/s00280-009-0933-9Search in Google Scholar PubMed PubMed Central

[34] Qi X, Chu Z, Mahller YY, Stringer KF, Witte DP, Cripe TP. Cancer-Selective Targeting and Cytotoxicity by Liposomal-Coupled Lysosomal Saposin C Protein. Clinical Cancer Research 2009, 15(18):5840-5851. [PubMed: 19737950]10.1158/1078-0432.CCR-08-3285Search in Google Scholar PubMed

[35] Seymour LW, Ferry DR, Anderson D, Hesslewood S, Julyan PJ, Poyner R, Doran J, Young AM, Burtles S, Kerr DJ. Hepatic drug targeting: phase I evaluation of polymer-bound doxorubicin. Journal of clinical oncology 2002, 20(6):1668. [PubMed: 11896118]10.1200/JCO.2002.20.6.1668Search in Google Scholar PubMed

[36] Morse M. Technology evaluation: Rexin-G, Epeius Biotechnologies. Current Opinion in Molecular Therapeutics 2005, 7(2):164. [PubMed: 15844625]Search in Google Scholar

[37] Paciotti GF, Kingston DG, Tamarkin L. Colloidal gold nanoparticles: a novel nanoparticle platform for developing multifunctional tumor-targeted drug delivery vectors. Drug development research 2006, 67(1):47-5410.1002/ddr.20066Search in Google Scholar

[38] Dobrovolskaia MA. Immunological properties of engineered nanomaterials. Nature nanotechnology 2007, 2(8):46910.1142/9789814287005_0029Search in Google Scholar

[39] Shaffer SA, Baker-Lee C, Kennedy J, Lai MS, Vries P, Buhler K, Singer JW. In vitro and in vivo metabolism of paclitaxel poliglumex: identification of metabolites and active proteases. Cancer Chemother Pharmacol 2006, 59(4):537-548. [PubMed: 16924498]10.1007/s00280-006-0296-4Search in Google Scholar PubMed

[40] Debbage P, Jaschke W. Molecular imaging with nanoparticles: giant roles for dwarf actors. Histochem Cell Biol 2008, 130(5):845-875. [PubMed: 18825403]10.1007/s00418-008-0511-ySearch in Google Scholar PubMed

[41] Lindner LH. Dual role of hexadecylphosphocholine (miltefosine) in thermosensitive liposomes: Active ingredient and mediator of drug release. J. Controlled Release 2008, 125(2):11210.1016/j.jconrel.2007.10.009Search in Google Scholar PubMed

[42] Wang Y-X, Hussain S, Krestin G. Superparamagnetic iron oxide contrast agents: physicochemical characteristics and applications in MR imaging. European Radiology 2001, 11(11):2319-2331. [PubMed: 11702180]10.1007/s003300100908Search in Google Scholar PubMed

[43] Hayat, MA. Cancer imaging: Instrumentation and applications. Academic Press; 2007Search in Google Scholar

[44] Zhang X, Bowen C, Gareua P, Rutt B. Quantitative Analysis of SPIO and USPIO Uptake Rate by Macrophages: Effects of Particle Size, Concentration, and Labeling Time. Proc. Intl. Soc. Mag. Reson. Med 2001; 9Search in Google Scholar

[45] Gil PR. Nanopharmacy: Inorganic nanoscale devices as vectors and active compounds. Pharmacol Res. 2010, 62(2):115-25. 10.1016/j.phrs.2010.01.009Search in Google Scholar PubMed

[46] Schwartz JA. Feasibility study of particle-assisted laser ablation of brain tumors in orthotopic canine model. Cancer Res 2009, 69(4):1659. [PubMed: 19208847]10.1158/0008-5472.CAN-08-2535Search in Google Scholar PubMed

[47] Schmid G, Bäumle M, Geerkens M, Heim I, Osemann C, Sawitowski T. Current and future applications of nanoclusters. Chemical Society Reviews 1999, 28(3):179-18510.1039/a801153bSearch in Google Scholar

[48] Roszek B, Jong WH, Geertsma RE. Nanotechnology in medical applications: state-of-the-art in materials and devices. 2005 RIVM Report 265001001Search in Google Scholar

[49] Spies CKG. The efficacy of Biobon and Ostim within metaphyseal defects using the Göttinger Minipig. Arch. Orthop.Trauma Surg 2009, 129(7):979. [PubMed: 18677495]10.1007/s00402-008-0705-8Search in Google Scholar PubMed

[50] Hayman ML. Emerging Product and Patent Landscape for Nanosilver-Containing Medical Devices. The. Nanotech. L. & Bus 2009, 6:148Search in Google Scholar

[51] Lu S, Lu. Construction, application and biosafety of silver nanocrystalline chitosan wound dressing. Burns 2008, 34(5):623. [PubMed: 18226459]10.1016/j.burns.2007.08.020Search in Google Scholar

[52] Dobson MG. Emerging technologies for point-of-care genetic testing. Expert Review of Molecular Diagnostics 2007, 7(4):359. [PubMed: 17620045]10.1586/14737159.7.4.359Search in Google Scholar

[53] Buxton DB. Current status of nanotechnology approaches for cardiovascular disease: a personal perspective. Wiley interdisciplinary reviews (WIRES): Nanomedicine and nanobiotechnology 2009, 1(2):14910.1002/wnan.8Search in Google Scholar

[54] European Science Foundation (ESF). Nanomedicine: An ESF–European Medical Research Councils (EMRC) Forward Look Report. Strasbourg: ESF 2005, Available from: http://www.esf.org/fileadmin/Public_documents/Publications/Nanomedicine.pdf. Accessed November 25, 2015Search in Google Scholar

[55] Buzea C, Pacheco II, Robbie K. Nanomaterials and nanoparticles: sources and toxicity. Biointerphases 2007, 2:MR17–MR71.10.1116/1.2815690Search in Google Scholar

[56] Whitesides GM. Nanoscience, nanotechnology, and chemistry. Small. 2005, 1:172-17910.1002/smll.200400130Search in Google Scholar

[57] Webster TJ. Nanomedicine: what’s in a definition? Int. J. Nanomedicine 2006, 1:115-11610.2147/nano.2006.1.2.115Search in Google Scholar

[58] Yacoby A, Heiblum M, Mahalu D, Shtrikman H. Coherence and Phase Sensitive Measurements in a Quantum Dot. Phys. Rev. Lett 1995, 74:4047-4050. [PubMed: 10058399]10.1103/PhysRevLett.74.4047Search in Google Scholar

[59] Chan WC, Maxwell DJ, Gao X, Bailey RE, Han M, Nie S. Luminescent quantum dots for multiplexed biological detection and imaging. Curr. Opin. Biotechnol 2002, 13:40–46. [PubMed: 11849956]10.1016/S0958-1669(02)00282-3Search in Google Scholar

[60] Wu X, Liu H, Liu J, Haley KN, Treadway JA, Larson JP, Ge N, Peale F, Bruchez MP. Immunofluorescent labeling of cancer marker Her2 and other cellular targets with semiconductor quantum dots. Nat. Biotechnol 2003, 21:41-46. [PubMed: 12459735]10.1038/nbt764Search in Google Scholar PubMed

[61] Schätzlein AG. Delivering cancer stem cell therapies - a role for nanomedicines? Euro. J. cancer (Oxford, England: 1990) 2006, 42:1309-131510.1016/j.ejca.2006.01.044Search in Google Scholar PubMed

[62] Li N, Kommireddy DS, Lvov Y, Liebenberg W, Tiedt LR, De Villiers MM. Nanoparticle multilayers: surface modification of photosensitive drug microparticles for increased stability and in vitro bioavailability. J. Nanosci. Nanotechnol 2006, 6:3252-3260. [PubMed: 17048544]10.1166/jnn.2006.421Search in Google Scholar PubMed

[63] Kam NW, O’Connell M, Wisdom JA, Dai H. Carbon nanotubes as multifunctional biological transporters and near-infrared agents for selective cancer cell destruction. Proc. Natl. Acad. Sci. USA 2005, 102:11600–11605. [PubMed: 16087878]10.1073/pnas.0502680102Search in Google Scholar PubMed PubMed Central

[64] Pantarotto D, Briand JP, Prato M, Bianco A. Translocation of bioactive peptides across cell membranes by carbon nanotubes. Chem. Commun. (Camb.) 2004, 1:16-17. [PubMed: 14737310]10.1039/b311254cSearch in Google Scholar PubMed

[65] Kostarelos KLL, Pastorin G, Wu W, Wieckowski S, Luangsivilay J, Godefroy S, Pantarotto D, Briand J, Muller S, Prato M, Bianco A. Cellular uptake of functionalized carbon nanotubes is independent of functional group and cell type. Nat. Nanotechnol 2007, 2:108-113. [PubMed: 18654229]10.1038/nnano.2006.209Search in Google Scholar PubMed

[66] Zheng M, Jagota A, Semke ED, Diner BA, McLean RS, Lustig SR, Richardson RE, Tassi NG. DNAassisted dispersion and separation of carbon nanotubes. Nat. Mater 2003, 2:338-342. [PubMed: 12692536]10.1038/nmat877Search in Google Scholar PubMed

[67] Rajendra J, Baxendale M, Dit Rap LG, Rodger A. Flow linear dichroism to probe binding of aromatic molecules and DNA to single-walled carbon nanotubes. J. Am. Chem. Soc 2004, 126:11182-11188. [PubMed: 15355099]10.1021/ja048720jSearch in Google Scholar PubMed

[68] Rajendra J, Rodger A. The binding of single-stranded DNA and PNA to single-walled carbon nanotubes probed by flow linear dichroism. Chemistry 2005, 11:4841-4847. [PubMed: 15954149]10.1002/chem.200500093Search in Google Scholar PubMed

[69] Audette GF, van Schaik J, Hazes B, Irvin RT. DNA-binding protein nanotubes. Nano. Lett 2004, 4:1897-190210.1021/nl048942fSearch in Google Scholar

[70] Arunachalam B, Phan UT, Geuze HJ, Cresswell P. Enzymatic reduction of disulfide bonds in lysosomes: characterization of a gamma-interferon-inducible lysosomal thiol reductase (GILT). Proc. Natl. Acad. Sci. USA 2000, 97:745-750. [PubMed: 10639150]10.1073/pnas.97.2.745Search in Google Scholar PubMed PubMed Central

[71] Feazell RP, Nakayama-Ratchford N, Dai H, Lippard SJ. Soluble single-walled carbon nanotubes as longboat delivery systems for platinum (IV) anticancer drug design. J. Am. Chem. Soc 2007, 129:8438-8439. [PubMed: 17569542]10.1021/ja073231fSearch in Google Scholar PubMed PubMed Central

[72] He P, Bayachou M. Layer-by-layer fabrication and characterization of DNA-wrapped single-walled carbon nanotube particles. Langmuir 2005, 21: 6086-6092. [PubMed: 15952864]10.1021/la050581bSearch in Google Scholar PubMed

[73] Chougule M, Padhi B, Misra A. Nano-liposomal dry powder inhaler of tacrolimus: preparation, characterization, and pulmonary pharmacokinetics. Int. J. Nanomed 2007, 2:675-688Search in Google Scholar

[74] Green AE, Rose PG. Pegylated liposomal doxorubicin in ovarian cancer. Int J Nanomedicine 2006, 1:229-239Search in Google Scholar

[75] Miele E, Spinelli GP, Miele E, Tomao F, Tomao S. Albumin-bound formulation of paclitaxel (Abraxane ABI-007) in the treatment of breast cancer. Int J Nanomedicine. 2009, 4:99-10510.2147/IJN.S3061Search in Google Scholar

[76] Serda RE, Godin B, Blanco E, Chiappini C, Ferrari M. Multi-stage delivery nano-particle systems for therapeutic applications. Biochim Biophys Acta 2011, 1810:317-32910.1016/j.bbagen.2010.05.004Search in Google Scholar PubMed PubMed Central

[77] Ventola CL. The nanomedicine revolution: part 2: current and future clinical applications. P T 2012, 37:582-591Search in Google Scholar

[78] Bertrand N, Wu J, Xu X, Kamaly N, Farokhzad OC. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev 2014, 66:2-2510.1016/j.addr.2013.11.009Search in Google Scholar PubMed PubMed Central

[79] Venkatraman S. Has nanomedicine lived up to its promise? Nanotechnology 2014, 25:37250110.1088/0957-4484/25/37/372501Search in Google Scholar PubMed

[80] Natarajan JV, Darwitan A, Barathi VA, et al. Sustained drug release in nanomedicine: a long-acting nanocarrier-based formulation for glaucoma. ACS Nano 2014, 8:419-42910.1021/nn4046024Search in Google Scholar PubMed

[81] Murday JS, Siegel RW, Stein J, Wright JF. Translational nanomedicine: status assessment and opportunities. Nanomedicine 2009, 5:251-27310.1016/j.nano.2009.06.001Search in Google Scholar PubMed

[82] Lammers T, Subr V, Ulbrich K, et al. Polymeric nanomedicines for image-guided drug delivery and tumortargeted combination therapy. Nano Today 2010, 5:197-21210.1016/j.nantod.2010.05.001Search in Google Scholar

[83] Ahmad A, Wang Z, Ali R, et al. Apoptosis-inducing effect of garcinol is mediated by NF-kappaB signaling in breast cancer cells. J Cell Biochem 2010, 109:1134-114110.1002/jcb.22492Search in Google Scholar PubMed

[84] Khanna VK. Targeted delivery of nanomedicines. ISRN Pharmacol 2012, 57139410.5402/2012/571394Search in Google Scholar PubMed PubMed Central

[85] Luo D, Carter KA, Lovell JF. Nanomedical engineering: shaping future nanomedicines. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2015, 7:169-18810.1002/wnan.1315Search in Google Scholar

[86] Emerich DF, Halberstadt C, Thanos C. Role of nanobiotechnology in cell-based nanomedicine: A concise review. J Biomed Nanotechnol 2007, 3:235-24410.1166/jbn.2007.036Search in Google Scholar

[87] Ferrari M. Cancer nanotechnology: opportunities and challenges. Nat Rev Cancer 2005, 5:161-171. [PubMed: 15738981]10.1038/nrc1566Search in Google Scholar

[88] Langer R. Drug delivery and targeting. Nature 1998, 392:5-10. [PubMed: 9579855]Search in Google Scholar

[89] Langer R. Drug delivery. Drugs on target. Science 2001, 293:58-59. [PubMed: 11441170]10.1126/science.1063273Search in Google Scholar

[90] Peer D, Karp J, Hong S, Farokhzad O, Margalit R, Langer R. Nanocarriers: Emerging platforms for cancer therapy. Nature Nanotechnology 2007, 2:751-76010.1201/9780429399039-2Search in Google Scholar

[91] Tanaka T, Decuzzi P, Cristofanilli M, Sakamoto JH, Tasciotti E, Robertson FM, Ferrari M. nanotechnology for breast cancer therapy. Biomed. Microdevices 2009, 11:49–63. [PubMed: 18663578]10.1007/s10544-008-9209-0Search in Google Scholar

[92] Campos S. Liposomal anthracyclines: adjuvant and neoadjuvant therapy for breast cancer. Oncologist 2003, 8:10-16. [PubMed: 13679591]10.1634/theoncologist.8-suppl_2-10Search in Google Scholar

[93] Lyass O, Uziely B, Ben-Yosef R, Tzemach D, Heshing NI, Lotem M, Brufman G, Gabizon A. Correlation of toxicity with pharmacokinetics of pegylated liposomal doxorubicin (Doxil) in metastatic breast carcinoma. Cancer 2000, 89:1037-1047. [PubMed: 10964334]10.1002/1097-0142(20000901)89:5<1037::AID-CNCR13>3.0.CO;2-ZSearch in Google Scholar

[94] Moen MD, Lyseng-Williamson KA, Scott LJ. Liposomal amphotericin B: a review of its use as empirical therapy in febrile neutropenia and in the treatment of invasive fungal infections. Drugs 2009, 69:361-392. [PubMed: 19275278]10.2165/00003495-200969030-00010Search in Google Scholar

[95] Sparreboom A, Scripture CD, Trieu V, Williams PJ, De T, Yang A, Beals B, Figg WD, Hawkins M, Desai N. Comparative preclinical and clinical pharmacokinetics of a cremophor-free, nanoparticle albumin-bound paclitaxel (ABI-007) and paclitaxel formulated in Cremophor (Taxol). Clin Cancer Res 2005, 11:4136-4143. [PubMed: 15930349]10.1158/1078-0432.CCR-04-2291Search in Google Scholar

[96] Gradishar WJ. Albumin-bound nanoparticle paclitaxel. Clin Adv Hematol Oncol 2005, 3:348–9. [PubMed: 16167008]Search in Google Scholar

[97] Zhang L, Gu FX, Chan JM, Wang AZ, Langer RS, Farokhzad OC. Nanoparticles in medicine: therapeutic applications and developments. Clin Pharmacol Ther 2008, 83:761-769. [PubMed: 17957183]10.1038/sj.clpt.6100400Search in Google Scholar

[98] Muller C, Hohn A, Schubiger PA, Schibli R. Preclinical evaluation of novel organometallic 99mTcfolate and 99mTc-pteroate radiotracers for folate receptor-positive tumour targeting. Eur J Nucl Med Mol Imaging 2006, 33:1007-1016. [PubMed: 16763819]10.1007/s00259-006-0111-9Search in Google Scholar

[99] Pan X, Lee RJ. Tumour-selective drug delivery via folate receptor-targeted liposomes. Expert Opin Drug Deliv 2004, 1:7-17. [PubMed: 16296717]10.1517/17425247.1.1.7Search in Google Scholar

[100] Lu Y, Sega E, Low PS. Folate receptor-targeted immunotherapy: induction of humoral and cellular immunity against haptendecorated cancer cells. Int. J. Cancer 2005, 116:710-719. [PubMed: 15828051]10.1002/ijc.21126Search in Google Scholar

[101] Shi X, Wang S, Meshinchi S, Van Antwerp ME, Bi X, Lee I, Baker JR Jr. Dendrimer-entrapped gold nanoparticles as a platform for cancer-cell targeting and imaging. Small 2007, 3:1245-1252. [PubMed: 17523182]10.1002/smll.200700054Search in Google Scholar

[102] Brisaert M, Gabriels M, Matthijs V, et al. Liposomes with tretinoin: a physical and chemical evaluation. J. Pharm. Biomed. Anal 2001, 26:909-91710.1016/S0731-7085(01)00502-7Search in Google Scholar

[103] Gulati M, Grover M, Singh S, et al. Lipophilic drug derivatives in liposomes. Int J Pharmaceut 1998, 165:129-16810.1016/S0378-5173(98)00006-4Search in Google Scholar

[104] Archakov AI. Nanobiotechnologies in medicine: Nanodiagnostics and nanodrugs. Biochemistry (Moscow) Supplement Series B: Biomed Chem 2010, 4(1):2-1410.1134/S1990750810010026Search in Google Scholar

[105] Lundberg BB, Risovic V, Ramaswamy M, et al. A lipophilic paclitaxel derivative incorporated in a lipid emulsion for parenteral administration. J. Contr. Rel 2003, 86:93-10010.1016/S0168-3659(02)00323-1Search in Google Scholar

[106] Zhigaltsev IV, Maurer N, Akhong QF, et al. Liposomeencapsulated vincristine, vinblastine and vinorelbine: a comparative study of drug loading and retention. J Contr. Rel 2005, 104:103-11110.1016/j.jconrel.2005.01.010Search in Google Scholar PubMed

[107] Narang AS, Mahato RI. Biological and biomaterial approaches for improved islet transplantation. Pharmacol Rev 2006, 58:194-24310.1124/pr.58.2.6Search in Google Scholar PubMed

[108] Fernandes C, Soni U, Patravale V. Nano-interventions for neurodegenerative disorders. Pharmacol Res 2010, 62:166-17810.1016/j.phrs.2010.02.004Search in Google Scholar PubMed

[109] Juillerat-Jeanneret L. The targeted delivery of cancer drugs across the blood-brain barrier: chemical modifications of drugs or drug-nanoparticles? Drug Discov. Today 2008, 13:1099-110610.1016/j.drudis.2008.09.005Search in Google Scholar PubMed

[110] Webber MJ, Kessler JA, Stupp SI. Emerging peptide nanomedicine to regenerate tissues and organs. J Intern Med 2010, 267:71-8810.1111/j.1365-2796.2009.02184.xSearch in Google Scholar PubMed PubMed Central

[111] Khang D, Carpenter J, Chun YW, et al. Nanotechnology for regenerative medicine. Biomed Microdevices 2010, 12:575-58710.1007/s10544-008-9264-6Search in Google Scholar PubMed

[112] Roney C, Kulkarni P, Arora V, et al. Targeted nanoparticles for drug delivery through the blood-brain barrier for Alzheimer’s disease. J Contr. Release 2005, 108:193-21410.1016/j.jconrel.2005.07.024Search in Google Scholar PubMed

[113] Hyun H, Won YW, Kim KM, et al. Therapeutic effects of a reducible poly (oligo-D-arginine) carrier with the heme oxygenase-1 gene in the treatment of hypoxic-ischemic brain injury. Biomaterials 2010, 31:9128-913410.1016/j.biomaterials.2010.08.038Search in Google Scholar PubMed

[114] Hua MY, Liu HL, Yang HW, et al. The effectiveness of a magnetic nanoparticle-based delivery system for BCNU in the treatment of gliomas. Biomaterials 2011, 32:516-52710.1016/j.biomaterials.2010.09.065Search in Google Scholar PubMed

[115] Ghosh A, Chandran K, Kalivendi SV, et al. Neuroprotection by a mitochondria-targeted drug in a Parkinson’s disease model. Free Radical Bio Med 2010, 49:1674-168410.1016/j.freeradbiomed.2010.08.028Search in Google Scholar PubMed PubMed Central

[116] He H, Li Y, Jia XR, et al. PEGylated Poly(amidoamine) dendrimer-based dual-targeting carrier for treating brain tumors. Biomaterials 2011, 32:478-48710.1016/j.biomaterials.2010.09.002Search in Google Scholar PubMed

Received: 2017-6-13
Accepted: 2017-7-17
Published Online: 2017-9-14

© 2017 Arome Odiba et al.

This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 3.0 License.

Downloaded on 29.3.2024 from https://www.degruyter.com/document/doi/10.1515/med-2017-0041/html
Scroll to top button