applications and synthesis of nanoparticles in medicine

15
www.wjpr.net Vol 5, Issue 7, 2016. 365 APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE Dr. Leena H. Sarkar Department of Chemistry, J.V.M’s Degree College, Airoli, Navi Mumbai 400 708, India. ABSTRACT Nanoparticles are materials with dimensions of the nanoscale i.e., under 100 nm in size. Recently researchers have developed interest in synthesis and applications of nanotechnology. It is considered as most active area of research and advancements have been made in the field of medicine, clinical applications, magnetic resonance imaging and drug delivery systems. However, in some cases nanoparticles also bring with them unique challenges to our society and environment, due to their toxic nature. This review aims to highlight the major contributions of nanoparticles to modern medicine and also discuss the effects of nanoparticles on environment and society. KEYWORDS: nanoparticles, contrast agents, drug delivery, tumors, quantum dots, cancer, toxicity. INTRODUCTION Nanoparticles are particles between 1 and 100 nanometers in size. [1] They are also referred as “zero-dimensional” nanomaterials. In nanotechnology, a particle is defined as a small object that behaves as a whole unit with respect to its transport and properties. Particles are further classified according to diameter. [2] Nanoparticles show unique properties compared to the bulk metals therefore a lot of research work has been reported for the synthesis and applications of metal nanoparticles. [3] Nanoparticles have a number of novel optical, electronic, magnetic, structural properties, chemical reactivity, energy absorption, and biological mobility compared with the same materials at bulk volume by virtue of their size, making them attractive. There are two main approaches for the synthesis of nanoparticles: top-down and bottom-up. In top-down approach the material size is reduced from large to nanoscale, whereas in the World Journal of Pharmaceutical Research SJIF Impact Factor 6.805 Volume 5, Issue 7, 365-379. Review Article ISSN 2277– 7105 *Corresponding Author Dr. Leena H. Sarkar Department of Chemistry, J.V.M’s Degree College, Airoli, Navi Mumbai 400 708, India. Article Received on 24 April 2016, Revised on 15 May 2016, Accepted on 03 June 2016 DOI: 10.20959/wjpr20167-6467

Upload: others

Post on 15-Oct-2021

5 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

365

Sarkar World Journal of Pharmaceutical Research

APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN

MEDICINE

Dr. Leena H. Sarkar

Department of Chemistry, J.V.M’s Degree College, Airoli, Navi Mumbai 400 708, India.

ABSTRACT

Nanoparticles are materials with dimensions of the nanoscale i.e.,

under 100 nm in size. Recently researchers have developed interest in

synthesis and applications of nanotechnology. It is considered as most

active area of research and advancements have been made in the field

of medicine, clinical applications, magnetic resonance imaging and

drug delivery systems. However, in some cases nanoparticles also

bring with them unique challenges to our society and environment, due

to their toxic nature. This review aims to highlight the major

contributions of nanoparticles to modern medicine and also discuss the

effects of nanoparticles on environment and society.

KEYWORDS: nanoparticles, contrast agents, drug delivery, tumors, quantum dots, cancer,

toxicity.

INTRODUCTION

Nanoparticles are particles between 1 and 100 nanometers in size.[1]

They are also referred as

“zero-dimensional” nanomaterials. In nanotechnology, a particle is defined as a small object

that behaves as a whole unit with respect to its transport and properties. Particles are further

classified according to diameter.[2]

Nanoparticles show unique properties compared to the

bulk metals therefore a lot of research work has been reported for the synthesis and

applications of metal nanoparticles.[3]

Nanoparticles have a number of novel optical, electronic, magnetic, structural properties,

chemical reactivity, energy absorption, and biological mobility compared with the same

materials at bulk volume by virtue of their size, making them attractive.

There are two main approaches for the synthesis of nanoparticles: top-down and bottom-up.

In top-down approach the material size is reduced from large to nanoscale, whereas in the

World Journal of Pharmaceutical Research SJIF Impact Factor 6.805

Volume 5, Issue 7, 365-379. Review Article ISSN 2277– 7105

*Corresponding Author

Dr. Leena H. Sarkar

Department of Chemistry,

J.V.M’s Degree College,

Airoli, Navi Mumbai 400

708, India.

Article Received on

24 April 2016,

Revised on 15 May 2016,

Accepted on 03 June 2016

DOI: 10.20959/wjpr20167-6467

Page 2: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

366

Sarkar World Journal of Pharmaceutical Research

bottom-up approach nanomaterials are synthesised by starting from the atomic level.[4,5]

The

top-down approach often uses the traditional methods where externally controlled tools are

used to cut, mill, and shape materials into the desired shape and order. Bottom-up approach

uses the chemical properties of single molecules to (a) self-organize or self-assemble into

some useful conformation, or (b) rely on positional assembly. The biggest problem with top

down approach is that it introduces internal stress, contaminations, surface defects leading to

imperfection of structure and crystallographic damage. These imperfections lead to extra

challenges in the fabrication. But this approach leads to the bulk production of nano

materials. Regardless of the defects produced by top down approach, they continue to play an

important role in the synthesis of nano structures. Though the bottom up approach is an old

concept observed in nature and has been used in industries for a long time. For example,

production of salt in chemical industry. Bottom up approach gives nano structures with less

defects and more homogeneous chemical composition.

These materials can be synthesized by several methods which include solid-phase, liquid-

phase and gas-phase processes. The solid-phase techniques include mechanical ball

milling[6,7]

and mechano chemical[7]

, the liquid-phase techniques which include laser

ablation[8,9]

, exploding wire[10]

, solution reduction[11]

, and decomposition process[12,13]

,

whereas the gas-phase processes include gas evaporation[14,15]

, exploding wire[16,17]

, and laser

ablation process.[18]

There are large number of applications of nanoparticles in medicine. This article reviews

synthesis, the role of nanoparticles in medicine, medical imaging and drug/gene delivery.

Also the effect of nanoparticles on environment has been discussed.

ROLE OF NANOPARTICLES (NPs) IN MAGNETIC RESONANCE IMAGING

(MRI)

Cancer is a disease for which mortality rate is very high. Treatment methods include surgery,

chemotherapy and radiation therapy. However, the results of the treatment are not positive in

many cases. The reasons behind this being ineffective early diagnosis, insufficient drug

concentrations reaching the tumour cells, adverse effects of drugs. Thus, the important issues

in improving treatment regimens are (i) development of advanced imaging technologies for

early diagnosis and (ii) utilization of targeting moieties to specifically and efficiently deliver

drugs to tumour tissues.

Page 3: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

367

Sarkar World Journal of Pharmaceutical Research

Harisinghani et al[19]

used SPIO nanoparticles to detect small metastases in the lymph node in

human patients with prostate cancer which would not be detected by conventional MRI.

SPIO nanoparticles allow the visualization of metastases that cannot be clearly observed by

conventional MRI. The significance of this work is that patients with localized disease have

the option of early treatment by surgery without being restricted to radiation therapy, the

primary treatment for advanced-stage patients.

Huh et al[20]

described the use of SPIO nanoparticles to detect cancer in vivo using a mouse

xenograft model. SPIO nanoparticles were prepared by the thermal decomposition of iron

acetylacetonate and made water-soluble by binding with 2, 3-dimercaptosuccinic acid and

then conjugated to herceptin, a cancer-targeting antibody. When administered intravenously

to mice, a rapid change was observed in the T2-weighted MRI signal from the tumor located

in the thigh of the animals.

Recently researchers at Imperial College London[21]

, have designed a new self-assembling

nanoparticle that boosts the effectiveness and sensitivity of Magnetic Resonance Imaging

(MRI) scanning, targets tumours and helps doctors for earlier diagnosis of cancer.

The nanoparticle is coated with a special protein, which looks for specific signals given off

by tumours, and when it finds a tumour it begins to interact with the cancerous cells. This

interaction strips off the protein coating, causing the nanoparticle to self-assemble into a

much larger particle so that a more powerful signal is obtained and a clearer MRI image of

the tumour is visible. This would enable doctors for faster detection of cancer and patients to

receive effective treatment sooner.

ROLE OF NANOPARTICLES IN OPTICAL IMAGING

Conventional imaging of cells and tissue sections is performed by loading organic dyes into

the sample. Dyes such as fluorescein isocyanate (FITC) and rhodamine are often attached to

biomolecules that selectively bind to cells or cell components through ligand/receptor

interactions. Inadequate fluorescence intensity and photobleaching i.e. gradual decrease in

fluorescence intensity are observed in this mode.

Fluorescent nanoparticles (NPs), such as organic dye-doped NPs, quantum dots enable highly

sensitive optical imaging of cancer at cellular and animal level.

Page 4: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

368

Sarkar World Journal of Pharmaceutical Research

NPs, when conjugated with several functional molecules like targeting moieties and imaging

probes, provides new potentials for clinical therapies and diagnostics. NP-based drug-

delivery systems based on polyethyleneimine (PEI), liposomes, micelles and silica NPs help

in easy drug delivery and reduce drug side effects.[22]

The core of the nanoparticle employs a phenomenon called plasmonics. In plasmonics,

nanostructured metals such as gold and silver[23]

resonate in light and concentrate the

electromagnetic field near the surface. In this way, fluorescent dyes are enhanced, appearing

about tenfold brighter than their natural state when no metal is present. When the core is

etched, the enhancement goes away and the particle becomes dim.

Quantum dots (QDs) are made of inorganic semiconductor molecules. They emit strong

fluorescent light under ultraviolet (UV) illumination. The wavelength of the emitted

fluorescent light depends on particle size. When QDs are excited by giving energy of a

specific wavelength, in the form of a photon an electron jumps from the valence band to the

conduction band and leaves behind a “hole”. When the excitation ceases, electrons come

back to the valence band, by emitting energy in form of light. Larger QDs have more

electron-hole pairs and are therefore capable of absorbing and releasing more energy. The

wavelength of emitted light decreases as QD size increases as energy is inversely

proportional to wavelength. Hence QDs emit light that is far more intense as compared to

conventional organic dyes. This is a major advantage in 3-D tissue imaging where

photobleaching is a major concern.[24]

QDs made of CdTe and capped with CdSe are capable of light emission under near infrared

excitation. This helps to locate lymph nodes up to 1 cm below the skin without the need for

surgical incisions.[25]

Akerman et al showed that QDs can be targeted to specific organs within the body by coating

them with appropriate molecules.[26]

TARGETING OF TUMORS USING NANOPARTICLES

Most anti-cancer drugs cannot differentiate between cancerous and normal cells, leading to

systemic toxicity and adverse effects. The maximal allowable dose is limited due to severe

side effects of the drugs to be delivered in a living system, resulting in inadequate drug

Page 5: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

369

Sarkar World Journal of Pharmaceutical Research

concentrations reaching the tumour cells. NPs can deliver anti-cancer drugs to tumour sites

efficiently benefiting cancer patients.[27]

Gold nanoparticles are emerging as promising agents for cancer therapy and are being

investigated as drug carriers, photothermal agents. Tumour targeting can be achieved by

actively binding tumour-specific recognition molecules such as epidermal growth factor

(EGF), transferrin, folic acid or monoclonal antibodies to nanoparticles.[28,29,30]

Scientists at UC Santa Barbara[23]

have designed silver nanoparticles that are spherical in

shape and are encased in a shell coated with a peptide that enables it to target tumor cells.

The shell is etchable so those nanoparticles that don’t hit their target can be broken down and

eliminated.

UCSB’s Ruoslahti Research Laboratory has also developed a simple etching technique which

uses biocompatible chemicals to rapidly disassemble and remove the silver nanoparticles

outside living cells. This method leaves only the intact nanoparticles for imaging or

quantification, thus revealing which cells have been targeted and how much each cell

internalized. “The disassembly is an interesting concept for creating drugs that respond to a

certain stimulus. It also minimizes the off-target toxicity by breaking down the excess

nanoparticles so they can then be cleared through the kidneys.” Since the nanoparticle has a

core shell structure, by varying its exterior coating the efficiency of tumor targeting and

internalization can be compared. Switching out the surface agent enables the targeting of

different diseases.

NANOPARTICLES IN DRUG AND GENE DELIVERY

Nowadays in drug therapy, a drug is transported to the place of action by using controlled

drug delivery systems (DDS)[31]

as a result, its effectiveness can be increased and its

influence on vital tissues and undesirable side effects can be reduced. Also, more drug

accumulates at the target site and so lower doses of drugs are required.

A drug may be adsorbed or covalently attached to the nanocarriers surface or else it can be

encapsulated into it. Covalent linking has the advantage over other ways of attaching as it

enables to control the number of drug molecules connected to the nanocarrier, i.e., a precise

control of the amount of the drug will be delivered. Cell-specific targeting with nanocarriers

Page 6: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

370

Sarkar World Journal of Pharmaceutical Research

may be accomplished by using active or passive mechanisms. Nanocarriers used for medical

applications have to be biocompatible and nontoxic.

SLN (solid lipid nanoparticles), NLC (nanostructured lipid carriers) and LDC (lipid drug

conjugates) are types of carrier systems based on solid lipid matrix i.e., lipids solid at the

body temperature.[32]

They have been exploited for the dermal[33]

, peroral[34]

, parenteral[35]

,

ocular[36]

, pulmonary[37]

, and rectal application.

Polymeric nanoparticles (PNPs) are structures obtained from synthetic polymers, such as

poly-e-caprolactone[38]

, polyacrylamide[39]

and poly-acrylate[40]

, or natural polymers, e.g.,

albumin[41]

, gelatin.[42]

Based on in vivo behavior, PNPs may be classified as biodegradable,

i.e., poly(L-lactide) (PLA)[43]

, poly-glycolide (PGA).[44]

Nanocarriers composed of

biodegradable polymers undergo hydrolysis in the body, producing biodegradable metabolite

monomers, such as lactic acid and glycolic acid. Kumari et al.[45]

reported a minimal systemic

toxicity associated with using of PLGA poly(lactic-co-glycolic acid) for drug delivery or

biomaterial applications. These biodegradable polymeric nanocarriers used for drug delivery

are stable in blood, non-toxic, and non-thrombogenic. They are also non-immunogenic as

well as non-proinflammatory, and they neither activate neutrophils nor affect

reticuloendothelial system.[46]

Some nanomers have dendritic structure eg. glycogen, amylopectin, and proteoglycans.[47]

They consist of a core with at least two identical functional groups, dendrons and surface

active groups. Dendrimers cytotoxicity and use of dendrimers in medical applications,

biocompatibility and physicochemical properties of dendrimers are determined by surface

functional groups.[48]

For example, changing the surface amine groups into hydroxyl ones may result in lower

levels of cytotoxicity. The presence of several surface functional groups enables a

simultaneous interaction with a number of receptors increasing its biological activity. The

drug may either be encapsulated in the internal structure of dendrimers[49]

when drugs are

labile, toxic, or poorly Soluble or else it can be chemically attached or physically adsorbed on

dendrimers surface[50]

which helps to control quantity of drugs on dendrimers surface by

controlling the number of covalent bonds.[51]

Page 7: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

371

Sarkar World Journal of Pharmaceutical Research

The surface of dendrimers provides an excellent platform for an attachment of specific

ligands, which may include folic acid[51]

, antibodies[52]

, cyclic targeting peptides – arginine-

glycine-aspartic acid (RGD).[53]

Poly(amido amide) (PAMAM) is a dendrimer which is

frequently used in biomedical applications.

Carbon nanotubes (CNTs) and nanohorns (CNH) are used as nanocarriers. Biocompatibility

of these nanotubes can be increased by chemical modification of their surface.[54]

Such

modification can be implemented by covalent anchoring of PAMAM dendrimers[55]

,

amphiphilic diblock copolymers[56]

or PEG layers[57]

on CNTs surface. Due to their

mechanical strength, SWCNTs (Single Walled Carbon Nanotubes have been used to improve

properties of other carriers, e.g., polymeric or non-polymeric composites.[58]

Drug

immobilization in carbon nanocarriers can be done by - encapsulation of a drug in the carbon

nanotube[59]

, chemical adsorption on the surface or in the spaces between the nanotubes (by

electrostatic, hydrophobic, p-p interactions and hydrogen bonds)[60,61]

, and attachment of

active agents to functionalized carbon nanotubes (f-CNTs). Encapsulation has the advantage

over the two remaining methods as the drug is protected from degradation during its transport

to the cells and is released only in specific conditions.[62]

To prevent the unwanted release of

the drug, the open ends of CNTs are sealed with polypyrrole (PPy) films.[63]

CANCER

Nanoparticles are used for the treatment of various types of cancers. Paclitaxel is used for

treatment of various types of cancer (such as skin, ovarian, lung and oesophageal.[64,65,66]

This drug interferes with the functions of cancer cells by microtubule stabilization, resulting

in apoptosis.[67]

A major shortcoming of this approach is the side effects associated with

Cremophor®, including hypersensitivity reactions, necessitating the administration of steroids

and antihistamines as premedications [68]. Abraxane® is a different form of paclitaxel where

paclitaxel is embedded within nanoparticles of a natural polymer, albumin, using a high-

pressure emulsification process. This form eliminates the side effects of Cremophor®[69]

and

the albumin carrier improves transport of the drug from the bloodstream to the tumor site and

allows higher drug dosing compared with Taxol.®

If a large number of cancerous cells are removable, then they should be removed surgically,

but even after removing a bigger tumor, residual cancer cells can be left behind in the area.

These residual cells can grow into tumors, leaving the possibility of the cancer recurring.

Detecting single residual cancer cells is not easy, however a cluster as small as three cancer

Page 8: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

372

Sarkar World Journal of Pharmaceutical Research

cells can be detected using the gold nanoparticle method. This method then becomes a very

useful option for two reasons: 1) detecting residual cancer cells, indicating whether more

tissue needs to be removed and 2) using this method to obliterate residual cancer cells. The

researchers had success with both methods, resulting in no cancer reappearance in their trials.

Removing residual cancer cells with nanoparticles was deemed a new type of surgery by the

researchers. They called it “PNB nanosurgery” which stands for Plasmonic Nano Bubble

nanosurgery.[70]

NEURODEGENERATIVE DISEASES

Treatment for neurodegenerative diseases is a challenge due to difficulty in Drug delivery to

the central nervous system.[71]

Nanoparticles made from poly (hexadecyl cyanoacrylate) and

related compounds have been shown to facilitate drug transport across the blood–brain barrier

(BBB). Kreuter et al[72, 72a]

adsorbed dalargin (an analgesic) onto poly(butyl cyanoacrylate)

(PBCA) nanoparticles and demonstrated penetration across the BBB in rats. More recently,

Siegemund et al[73]

showed how PBCA nanoparticles loaded with thioflavins can target

fibrillar amyloid β in a murine model of Alzheimer’s disease.

HIV/AIDS

De Jaeghere et al[74]

investigated the delivery of an HIV-1 protease inhibitor, CGP 70726,

using pH-sensitive nanoparticles made from a copolymer of methacrylic acid and ethyl

acrylate. This copolymer is commercially available under the name Eudragit®

L100–55. The

HIV-1 Tat protein has recently emerged as a potential candidate for a prophylactic or

therapeutic vaccine against HIV-1/AIDS.[75]

Study is still going on for solutions for the

disease.

Besides this nanoparticles have also been effectively used in the treatment of ocular

diseases[76,77]

and respiratory disease.[78,79,80]

EFFECTS OF NANOPARTICLES ON ENVIRONMENT AND SOCIETY

With the development of nanotechnology, the effects of nanomaterials, on the environment

and public health has received considerable attention in recent years. On one hand,

nanotechnological innovations have helped to improve the health of people and at the same

time different kinds of risks are associated with different nanoparticles.

Page 9: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

373

Sarkar World Journal of Pharmaceutical Research

Air pollution affects human health. Nanoparticles can be present in inhaled particulate matter

and may get deposited in the lungs. They can move from the lungs to other organs such as the

brain, the liver and possibly the foetus in pregnant women. The effects of inhaled

nanoparticles in the body may include lung inflammation, heart problems and other health

problems.

Studies have been done and toxic effects of Quantum dots[81,82]

; metallic, polymeric and

liposomal nanoparticles[83,84,85,86]

have been explored.

CONCLUSION

Nanoparticles have been recently used in the field of medicine for medical imaging,

drug/gene delivery. Innovations are also associated with certain harmful effects on

environment and society. We have to overcome these challenges.

REFERENCES

1. ASTM International E 2456-06 Terminology for nanotechnology. West Conshohocken,

PA: ASTM International, 2006.

2. Module 3: Characteristics of Particles – Particle Size Categories. epa.gov.

3. 5- Sulekh Chandra, Avdhesh Kumar, Praveen Kumar Tomar., Thermal and spectral

studies. Spectrochimica Acta Part A, 2012; 92: 392–397.

4. Charinpanitkul, T., Faungnawakij, K., Tanthapanichakoon, W., Adv Powder Technol,

2008; 19: 443.

5. Parvez Iqbal, Jon A. Preece, Paula M. Mendes, Nanotechnology: The “Top-Down” and

“Bottom-Up” Approaches, Nanotechnology Published Online: 15 Mar. 2012,

DOI: 10.1002/9780470661345.smc195, Copyright © 2012 John Wiley & Sons.

6. Thakur Prasad Yadav, Ram Manohar Yadav, Dinesh Pratap Singh, Nanoscience and

Nanotechnology, 2012; 2: 22-48.

7. Boldyrev, V.V. & Tkacova, K., Journal of Materials Synthesis and Processing, 2000; 8:

121-132.

8. Pronko P. P., Zhang Z., VanRompay P. A., App. Surf. Sci., 2003; 208: 492-501.

9. Ullmann M., Friedlander S. K., Schmidt-Ott A. J., Nanopart. Res., 2002; 4: 499-509.

10. P Sen, Joyee Ghosh, Alqudami Abdullah, Prashant Kumar, Vanada, Proc. Indian Acad.

Sci. (Chem. Sci.), 2003; 115: 499–508.

11. Vemal Raja Manikam, Kuan Yew Cheong, Khairunisak Abdul Razak, Materials Science

and Engineering:B, 2011; 176: 187–203.

Page 10: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

374

Sarkar World Journal of Pharmaceutical Research

12. Heting Li, Mohammed J. Meziani, Fushen Lu, Christopher E. Bunker, Elena A. Guliants,

and Ya-Ping Sun, J. Phys. Chem. C, 2009; 113: 20539–20542.

13. N. A. Niaz, Ishaq Ahmad, S. Nasir, Z. Wazir, R. Hussain, N. R. Khalid, S. Tajammul

Hussain, Digest Journal of Nanomaterials and Biostructures, 2013; 8: 423–431.

14. Sun X. K., Xu J., Chen W. X., Wei W. D., Nanostruct. Mater, 1994; 4: 337-44.

15. Puszynski J. A., Proc. Int. Pyrotech. 29th, 2002; 191-202.

16. L Yap, Y S Lee, W H Tay, C S Wong, Proceedings of the International Workshop On

Plasma Computations & Applications (IWPCA2008) 60-64.

17. T. K. Sindhu, R. Sarathi, S. R. Chakravarthy, Bulletin of Materials Science, 2007; 30:

187-195.

18. Han M., Gong Y., Zhou J., Yin C., Song F., Muto M., Takiya T., Iwata Y., Phys. Lett.,

2002; 302: 182-189.

19. Harisinghani MG, Barentsz J, Hahn PF, et al. Noninvasive detection of clinically occult

lymphnode metastases in prostate cancer. N Engl J Med., 2003; 348: 2491–99.

20. Huh YM, Jun YW, Song HT, et al. In vivo magnetic resonance detection of cancer by

using multifunctional magnetic nanocrystals. J. Am. Chem. Soc., 2005; 127: 12387–91.

21. Source: Imperial College London Date: July 16, 2014

22. Sinha R., Kim G. J., Nie S. M., Shin D. M., Nanotechnology in cancer therapeutics:

bioconjugated nanoparticles for drug delivery. Mol. Cancer Ther., 2006; 5: 1909–

1917. (doi:10.1158/1535-7163.MCT-06-0141)

23. Julie Cohen, Sunday, June 8, 2014; 10: 00, Santa Barbara, CA.

24. Michalet X, Pinaud FF, Bentolila LA, et al. Quantum dots for live cells, in vivo imaging,

and diagnostics. Science, 2005; 307: 538–44.

25. Kim S, Lim YT, Soltesz EG, et al. Near-infrared fluorescent type II quantum dots for

sentinel lymph node mapping. Nat Biotechnol, 2004; 22: 93–7.

26. Akerman ME, Chan WCW, Laakkonen P, et al. Nanocrystal targeting in vivo. Proc Natl

Acad Sci U S A., 2002; 99: 12617–21.

27. Byrne J. D., Betancourt T., Brannon-Peppas L., Active targeting schemes for nanoparticle

systems in cancer therapeutics. Adv. Drug Del. Rev., 2008; 60: 1615-1626.

(doi:10.1016/j.addr.2008.08.005)

28. Chithrani BD, Ghazani AA, Chan WC. Determining the size and shape dependence of

gold nanoparticle uptake into mammalian cells. Nano Lett., 2006; 6: 662–8.

Page 11: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

375

Sarkar World Journal of Pharmaceutical Research

29. El-Sayed IH, Huang X, El-Sayed MA. Surface plasmon resonance scattering and

absorption of anti-EGFR antibody conjugated gold nanoparticles in cancer diagnostics:

applications in oral cancer. Nano Lett., 2005; 5: 829–34.

30. Eghtedari M, Liopo AV, Copland JA, Oraevsky AA, Motamedi M. Engineering of

hetero-functional gold nanorods for the in vivo molecular targeting of breast cancer

cells.Nano Lett, 2009; 9: 287–91.

31. Nevozhay D, Kañska U, Budzyñska R, Boratyñski J: Current status of research on

conjugates and related drug delivery systems in the treatment of cancer and other diseases

(Polish). Postepy HigMed Dosw, 2007; 61: 350–360.

32. Wissing SA, Kayser O, Müller RH: Solid lipid nanoparticles for parenteral drug delivery.

Adv Drug Deliv Rev, 2004; 56: 1257–1272.

33. Abdel-Mottaleb MMA, Neumann D, Lamprecht A: Lipid nanocapsules for dermal

application: A comparative study of lipid-based versus polymer-based nanocarriers. Eur J

Pharm Biopharm, 2011; 79: 36–42.

34. Muchow M, Maincent P, Müller RH: Lipid nanoparticles with a solid matrix (SLN, NLC

LDC) for oral drug delivery. Drug Dev Ind Pharm, 2008; 34: 1394–1405.

35. Nayak AP, Tiyaboonchai W, Patankar S, Madhusudhan B, Souto EB: Curcuminoids-

loaded lipid nanoparticles: novel approach towards malaria treatment. Colloids Surf B

Biointerfaces, 2010; 81: 263–273.

36. Attama AA, Schicke BC, Paepenmüller T, Müller-Goymann CC: Solid lipid

nanodispersions containing mixed lipid core and a polar heterolipid: characterization. Eur

J Pharm Biopharm, 2007; 67: 48–57.

37. Liu J, Gong T, Fu H,Wang C,Wang X, Chen Q, Zhang Q, He Q, Zhang Z: Solid lipid

nanoparticles for pulmonary delivery of insulin, Int J Pharm, 2008; 356: 333–344.

38. Bilensoy E, Sarisozen C, Esendagl G, Dogan LA, Aktas Y, Sen M, Mangan AN:

Intravesical cationic nanoparticles of chitosan and polycaprolactone for the delivery of

Mitomycin C to bladder tumors. Int J Pharm, 2009; 371: 170–176.

39. Bai J, Li Y, Du J, Wang S, Zheng J, Yang O, Chen X: One-pot synthesis of

polyacrylamide-gold nanocomposite. Mater Chem Phys, 2007; 106: 412–415.

40. Turos E, Shim JY, Wang Y, Greenhalgh K, Reddy GS, Dickey S, Lim DV: Antibiotic-

conjugated polyacrylate nanoparticles: New opportunities for development of anti-MRSA

agents. Bioorg Med Chem Lett, 2007; 17: 53–56.

Page 12: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

376

Sarkar World Journal of Pharmaceutical Research

41. Martinem A, Iglesias I, Lozano R, Teijon JM, Blanco MD: Synthesis and characterization

of thiolated alginate-albumin nanoparticles stabilized by disulfide bonds. Evaluation as

drug delivery systems. Carbohydr Polym, 2011; 83: 1311–1321.

42. Saraog GK, Gupta P, Gupta UD, Jain NK, Agrawal GP: Gelatin nanocarriers as potential

vectors for effective management of tuberculosis. Int J Pharm, 2010; 385: 143–149.

43. Mainardes RM, Khalil NM, Gremião MPD: Intranasal delivery of zidovudine by PLA

and PLA–PEG blend nanoparticles. Int J Pharm, 2010; 395: 266–271.

44. Park J, Fong PM, Lu J, Russell KS, Booth KJ, Saltzman WM, Fahmy TM: PEGylated

PLGA nanoparticles for the improved delivery of doxorubicin. Nanomedicine, 2009; 5:

410-418.

45. Kumari A, Yadav SK, Yadav SC: Biodegradable polymeric nanoparticles based drug

delivery systems. Colloids Surf B Biointerfaces, 2010; 75: 1–18.

46. des Rieux A, Fievez V, Garinot M, Schneider YJ, Préat V: Nanoparticles as potential oral

delivery systems of proteins and vaccines: a mechanistic approach. J Control Release,

2006; 116: 1–27.

47. Svenson S, Tomalia DA: Dendrimers in biomedical applications – reflections on the field.

Adv Drug Deliv Rev, 2005; 57: 2106–2129.

48. Caminade AM, Laurent R, Majoral JP: Characterization of dendrimers. Adv Drug Deliv

Rev, 2005; 57: 2130–2146.

49. D’Emanuele A, Attwood D: Dendrimer-drug interactions. Adv Drug Deliv Rev, 2005; 57:

2147–2162.

50. Menjoge AR, Kannan RM, Tomalia DA: Dendrimerbased drug and imaging conjugates:

design considerations for nanomedical applications. Drug Discov Today, 2010; 15: 171–

187.

51. Singh P, Gupta U, Asthana A, Jain NK: Folate and Folate-PEG-PAMAM dendrimers:

synthesis, characterization, and targeted anticancer drug delivery potential in tumor

bearing mice. Bioconjugate Chem, 2008; 19: 2239–2252.

52. Wängler C, Moldenhauer G, Eisenhut M, Haberkorn U, Mier W: Antibody-dendrimer

conjugates: the number, not the size of the dendrimers, determines the imunoreactivity.

Bioconjug Chem, 2008; 19: 813–820.

53. Waite CL, Roth ChM: PAMAM-RGD conjugates enhance siRNA delivery through a

multicellular spheroid model of malignant glioma. Bioconjug Chem, 2009; 20: 1908–

1916.

Page 13: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

377

Sarkar World Journal of Pharmaceutical Research

54. Foldvari M, Bagonluri M: Carbon nanotubes as functional excipients for nanomedicines:

I. Pharmaceutical properties. Nanomedicine, 2008; 4: 173–182.

55. Zhang B, Chen Q, Tang H, Xie Q, Ma M, Tan L, Zhang Y, Yao S: Characterization of

and biomolecule immobilization on the biocompatible multi-walled carbon nanotubes

generated by functionalization with polyamidoamine dendrimers. Colloids Surf B

Biointerfaces, 2010; 80: 18–25.

56. Di Crescenzo A, Velluto D, Hubbell JA, Fontana A: Biocompatible dispersions of carbon

nanotubes: a potential tool for intracellular transport of anticancer drugs. Nanoscale,

2011; 3: 925–928.

57. Bhirde AA, Patel S, Sousa AA, Patel V, Molinolo AA, Ji Y, Leapman RD et al.:

Distribution and clearance of PEG-single-walled carbon nanotube cancer drug delivery

vehicles in mice. Nanomedicine, 2010; 5: 1535–1546.

58. Shin US, Yoon IK, Lee GS, Jang WC, Knowles JC, Kim HW: Carbon nanotubes in

nanocomposites and hybrids with hydroxyapatite for bone replacements. J Tissue Eng,

2011. (doi:10.4061/2011/674287).

59. Arsawang U, Saengsawang O, Rungrotmongkol T, Sornmee P, Wittayanarakul K,

Remsungnen T, Hannongbua S: How do carbon nanotubes serve as carriers for

gemcitabine transport in a drug delivery system? J Mol Graph Model, 2011; 29: 591–596.

60. Chen Z, Pierre D, He H, Tan S, Pham-Huy C, Hong H,Huang J: Adsorption behavior of

epirubicin hydrochloride on carboxylated carbon nanotubes. Int J Pharm, 2011; 28: 405,

153–161.

61. Zhang D, Pan B, Wu M, Wang B, Zhang H, Peng H, Wu D, Ning P: Adsorption of

sulfamethoxazole on functionalized carbon nanotubes as affected by cations and anions.

Environ Pollut., 2011; 159: 2616–2621.

62. Perry JL, Martin CR, Stewart JD: Drug-delivery strategies by using template-synthesized

nanotubes. Chemistry, 2011; 17: 6296–6302.

63. Luo X, Matranga C, Tan S, Alba N, Cui XT: Carbon nanotube nanoreservior for

controlled release of anti-inflammatory dexamethasone. Biomaterials, 2011; 32: 6316–

6323.

64. Kikuchi A, Sakamoto H, Yamamoto T. Weekly carboplatin and paclitaxel is safe, active,

and well tolerated in recurrent ovarian cancer cases of Japanese women previously treated

with cisplatin containing multidrug chemotherapy. Int J Gynecol Cancer, 2005; 15: 45–9.

Page 14: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

378

Sarkar World Journal of Pharmaceutical Research

65. Chao Y, Li CP, Chao TY, et al. An open, multi-centre, phase II clinical trial to evaluate

the efficacy and safety of paclitaxel, UFT, and leucovorin in patients with advanced

gastric cancer. Br J Cancer, 2006; 95: 159–63.

66. Roof KS, Coen J, Lynch TJ, et al. Concurrent cisplatin, 5-FU, pacli-taxel, and radiation

therapy in patients with locally advanced esophageal cancer. Int J Radiat Oncol Biol

Phys., 2006; 65: 1120–8.

67. Koziara JM, Whisman TR, Tseng MT, et al. In-vivo efficacy of novel paclitaxel

nanoparticles in paclitaxel-resistant human colorectal tumors. J Control Release, 2006;

112: 312–19.

68. Zhang JA, Anyarambhatla G, Ma L, et al. Development and characterization of a novel

Cremophor (R) EL free liposome-based paclitaxel (LEP-ETU) formulation. Eur J Pharm

Biopharm, 2005a; 59: 177–87.

69. Micha JP, Goldstein BH, Birk CL, et al. Abraxane in the treatment of ovarian cancer: The

absence of hypersensitivity reactions. Gynecol Oncol, 2006; 100: 437–8.

70. Meier, J. D., Oliver, D. A. & Varvares, M. A. Surgical margin determination in head and

neck oncology: current clinical practice. The results of an International American Head

and Neck Society Member Survey. Head Neck, 2005; 27: 952–958.

71. Garcia-Garcia E, Andrieux K, Gil S, et al. Colloidal carriers and blood-brain barrier

(BBB) translocation: A way to deliver drugs to the brain? Int J Pharm., 2005; 298: 274–

92.

72. Kreuter J, Ramge P, Petrov V, et al. Direct evidence that polysorbate-80-coated

poly(butylcyanoacrylate) nanoparticles deliver drugs to the CNS via specific mechanisms

requiring prior binding of drug to the nanoparticles. Pharm Res., 2003; 20: 409–16. a

http://www.ncbi.nlm.nih.gov/pmc/articles

73. Siegemund T, Paulke BR, Schmiedel H, et al. Thioflavins released from nanoparticles

target fibrillar amyloid beta in the hippocampus of APP/PS1 transgenic mice. Int J Dev

Neurosci, 2006; 24: 195–201.

74. De Jaeghere F, Allemann E, Kubel F, et al. Oral bioavailability of a poorly water soluble

HIV-1 protease inhibitor incorporated into pH-sensitive particles: effect of the particle

size and nutritional state. J Control Release, 2000; 68: 291–8.

75. Caputo A, Gavioli R, Ensoli B. Recent advances in the development of HIV-1 Tat-based

Vaccines. Curr HIV Res, 2004; 2: 357–76.

76. Ludwig A. The use of mucoadhesive polymers in ocular drug delivery. Adv Drug Deliv

Rev., 2005; 57: 1595–639.

Page 15: APPLICATIONS AND SYNTHESIS OF NANOPARTICLES IN MEDICINE

www.wjpr.net Vol 5, Issue 7, 2016.

379

Sarkar World Journal of Pharmaceutical Research

77. Pignatello R, Bucolo C, Ferrara P, et al. Eudragit RS100 (R) nano-suspensions for the

ophthalmic controlled delivery of ibuprofen. Eur J Pharm Sci., 2002a: 16: 53–61.

78. John AE, Lukacs NW, Berlin AA, et al. Discovery of a potent nanoparticle P-selectin

antagonist with anti-inflammatory effects in allergic airway disease. FASEB J, 2003; 17:

2296–8.

79. Pison U, Welte T, Giersig M, et al. Nanomedicine for respiratory diseases. Eur J

Pharmacol, 2006; 533: 341–50.

80. Kumar M, Kong X, Behera A, et al. Chitosan IFN-γ-pDNA nanoparticle (CIN) therapy

for allergic asthma. Genet Vaccines Ther, 2003; 1: 3.

81. Derfus AM, Chan WCW, Bhatia SN. Probing the cytotoxicity of semiconductor quantum

dots. Nano Lett, 2004; 4: 11–18.

82. Ballou B, Lagerholm BC, Ernst LA, et al. Noninvasive imaging of quantum dots in

mice. Bioconjugate Chem, 2004; 15: 79–86.

83. Muldoon LL, Sandor M, Pinkston KE, et al. Imaging, distribution, and toxicity of

superparamagnetic iron oxide magnetic resonance nanoparticles in the rat brain and

intracerebral tumor. Neurosurgery, 2005; 57: 785–96.

84. Hainfeld JF, Slatkin DN, Focella TM, et al. Gold nanoparticles: a new X-ray contrast

agent. Br J Radiol, 2006; 79: 248–53.

85. Alonso MJ. Nanomedicines for overcoming biological barriers. Biomed

Pharmacother, 2004; 58: 168–72.

86. Owens DE, Peppas NA. Opsonization, biodistribution, and pharma-cokinetics of

polymeric nanoparticles. Int J Pharm, 2006; 307: 93–102.