Download as doc, pdf, or txt
Download as doc, pdf, or txt
You are on page 1of 15

Applications of chitin and their derivatives in the field of medicine: a

review

Abstract: Chitin has been considered as the second most abundant naturally occurring biopolymer. It

can be extracted majorly from four major sources i.e. insects, fungi, crustaceans and from mollusks.

One of the chitin derivatives is chitosan which is more useful and important tool in cure and

treatment. This review mainly highlights the importance of chitin and its derivatives in field of

medicine. The data discussed in this review is withdrawn from various journals, books and other

sources.

Keywords: Chitin, chitosan, anticancer activity, immunoadjuvant, antimicrobial property, targeted

drug delivery.

1 Introduction

Chitin is considered as the second most plentiful biopolymer widely found in nature [1–3]

after cellulose [4]. In different organisms such as yeast, insects, marine invertebrates and

fungi etc., it is present in the form of well-organized structures of microfibrils either in cell

wall or in the exoskeleton of arthropods. It is a homopolymer that is composed of 2-

acetamido-2-deoxy-β-D glucopyranose 1-4 linked units, which is glucose derivative. The

chitin can be converted to its derivative, chitosan which consists of greater than 50% of its

free amine form [5].

Chitin is not soluble in water and it exhibits mainly two polymorphs i.e. α polymorphic form

and β polymorphic form. Beta chitin is found commonly deacetylation in nature i.e. mostly

among crustaceans [6] while alpha chitin can be found from squid pens. Dissimilar to chitin,

chitosan shows solubility in dil. aqueous solutions. The chitosan can be classified into various

categories which depend on their molecular weight, patterns of acetylation, fractions of

acetylated residues, and their degree of weight distribution [4,6]. Chitosan with molecular
mass less than 3900 Da and less than 20 degree of polymerization are referred as

chitooligosaccharides [7]. Chitooligosaccharides (CHOS) are another important chitin

derivative, which can be obtained from chitosan either by chemical process i.e. acid

hydrolysis or by enzymatic process i.e. glycosyl hydrolases [4,7].

Chitin is very important in nature as it supplies strength to most invertebrates. Its composition

includes 6.9% of nitrogen therefore it is an important and useful chelating agent and has

many applications in the field of medicine [8]. Now a days chitosan and CHOS have taken

noticeable attention due to their peculiar properties and their various medicinal uses as they

are involved in drug delivery systems, wound dressings [9,10], anti-cancerous effects, anti-

microbial activity, anti-allergic responses [11] along with some others (Fig. 2).

This review concerns to analyze the anti-cancerous effects, anti-microbial effects, anti-

allergic effects and drug delivery systems of chitin, chitosan and their derivatives in the field

of medicine and their future prospective.

2 Sources of chitin

Shrimps are used as food. Forty five (45) percent of sea food is found in shrimps, which is

composed of cephalothorax and exoskeleton [12]. Cephalothorax is an environmental hazard

but it consists of beneficial products like chitin and protein [13]. Chitin is extracted from

insects, Mollusks, crustaceans and fungal exoskeleton (Fig. 1) [14]. Silkworm is also a

potential source of chitin [15]. Squid is another source of chitin [16,17]. Crustaceans like

Penaeus semisulcatus, Metapenaeus affinis, Portunus pelagicus , Thenus orientalis , Sepia

spp. have also been reported as chitin source [18].

Figure 1. Sources of chitin.


3 Extraction of chitin

There are different methods for chitin extraction. But chemical method is commonly used. It

consists of following steps. 1. Demineralization (removal of inorganic matter like calcium

carbonate), 2. deproteinization (removal of protein matter). Demineralization is done by using

HCl and deproteinization by using KOH or NaOH aqueous solutions [18]. Microorganisms

are also used for demineralization [19] and in deproteinization [20] in biological processing

which is an alternative of chemical process.

4 Applications in Medicine

Various biomedical uses of Chitin and chitosan have been discussed as under and also show

in Fig. 2.

Figure 2. Medicinal applications of chitin and its derivatives.

4.1 Anticancer activity

Chitooligosaccharides (CHOS) are used in anti-tumor activity. The mechanism is that CHOS

increases the differentiation and proliferation of T-cell, that increases the immunity. And in

the result tumor formation activity is reduced [21]. Maeda and Kimura performed an

experiment and showed that chitosan of low molecular weight, have decrease tumor size in

mice by enhancing the activity of T-cell [22,23]. Experiments showed that low molecular

weight and water soluble chitosan and oligochitosan act as immunomodulator and enhance

cytotoxic activity against tumor cells [24]. Furthermore, from lymphocytes chitosan produces

cytokinins such as interferon that activate microphages. Heparanase is a beta endoglucanase

that assist metastasis and tumor invasion. Chitosan act as inhibitors of heparinase [25].

Aminoderivatized COS act as inhibitors of gastric adenocarcinoma [26]. In vivo and in vitro

both type of cancers are inhibited by COS [27,28].


4.2 Immunoadjuvant

From studies it is determined that chitin and its derivatives have an effect on microphages

that in result produce cytokines. Cytokines produce resistance in host from bacterial and viral

activity as well as tumor formation [29–33].

Shibata et al. [34] performed experiments on mice and studied how chitin work on

phagocytes and NK cells (natural killer cells; a type of lymphocytes) they use chitin particles

having sized 1-10 micrometer via intravenous administration into lungs. These chitin

particles activated phagocytes, in a results cytokines like IL-12, IL-18, tumor necrosis

factors-alpha, that lead to the production of INF-Ɣ by NK cells

They also studied that the production of cytokines is bring about by mannose receptor

mechanism in phagocytes [35]. Mannose receptors present on the plasma membrane are

means of acceptance of chitin particles. In macrophages of experimental animals these

particles are then break down by lysozymes and N acetyl beta glucosaminidase [36] from

these studies the interaction of chitin and receptors of cell surface was studied and it was

shown that chitin uses specific signals for immune regulation.

4.3 Antimicrobial property

The condition in which chitosan is used i.e. in form of hydrogels, films, coatings, or in other

combinations contribute to its antimicrobial properties. It is supposed that the negatively

charged cell membrane of microbe and positively charged molecules of chitosan interact with

each other and this ultimately leads to the distortion of cell membrane of microbe. Thereby

leakage of intracellular components takes place [37,38].

The in vitro experiments that were performed were based on the death rates of bacterial

strains like Staphylococcus saprophyticus, Pseudomonas aeruginosa, Enterococcus faecalis

and Escherichia coli [39].


Chitosans depending upon their molecular weights have showed more antibacterial results as

compared to CHOS and have shown a prominent cease in growth of most tested bacteria,

although the antibacterial effects were more for gram positive bacteria as compared to gram

negative bacteria [40].

4.4 Wound healing and Burn treatment

It is obvious that chitin is so much beneficial for the human body that it can be applied to

body from head to the tip of toe (Fig. 3). Chitosan is so important that it can bind with the

RBC’s and it rapidly clots the blood and therefore it has gained importance in USA and is

being used in hemostatic agents and bandages [41,42]. In the form of a partially permeable

biological dressing it maintains sterile wound gum and averts dehydration of wound and its

contamination, and creates best conditions for healing purpose.

A wound infection is demonstration of disrupted host-bacteria equilibrium in an injured tissue

environment favorable for bacteria. Chitosan not only has a possibility to draw a systemic

response, but is also likely to impede the process involved in development of a normal wound

healing. Every process in wound healing is affected by bacterial growth in a wound [43].

Chitosan dressing was refined by Ong et al. [44]. combining silver and polyphosphate with

chitosan and thereby increasing its antimicrobial and haemostatic properties. In both in vitro

and animal studies it was found that formulation of chitosan polyphosphate speeds up blood

clotting, generates faster thrombin, increased the adhesion of pellets, and absorbed more

blood than chitosan alone. Silver chitosan polyphosphate formulations showed more

antimicrobial activity than chitosan polyphosphate in vitro, and thereby achieved a complete

death of P. aeruginosa and a >99.99% death of S. aureus.

Efficiency of burn infections treatment was noted by Dai et al. [45]. Third degree burnt

wounds that had bioluminescent bacterial infections were applied the dressings of both
chitosan acetate bandage and silver dressings, and that showed a significant decrease in

luminesecence signal of bacteria as compared to untreated burnt wounds.

In hernia repair the use of mesh creates surgical site infections. Commercial absorbable

materials which are triclosan embedded are used to decrease the rate of infection in these

wounds. Cakmak et al. [46] used mesh which was coated with chitosan gel infections along

with triclosan embedding for treatment and prevention of mesh infections taking rat as a

model. It is reported that graft coatings of chitosan gel along with triclosan loading show

treatment against graft infection.

The specific biological activity that is associated with the tissue regeneration and general

growth processes is called wound healing. It advances through various overlapping and

interdependent stages of reestablishment of the integrity and replacement of tissue by the

action of cellular components [47].

Figure 3. Wound healing applications of chitosan.

4.5 Targeted Drug Delivery

Chitosan is a biodegradable, low toxic, good biocompatible compound and therefore is

preferred for many medical applications mainly in the targeted drug delivery [48,49]. During

recent years there has been a great deal of development in targeted drug delivery systems

together with anti-cancerous agents to attain superior clinical tolerance and response [50].

The chitosan particles were used in the form of a complex and that complex protected the

genetic material by encapsulating it and prevented it from the nuclease attack [51,52]. It can

also enhance the time of expression of genes. The unification of chitin, chitosan and other

derivatives together with some anti cancerous agents, enhance the anti-cancerous effects

better by decreasing the drawbacks free drug release in cancerous cells [51,53–55].
5 Conclusion

Chitin is a naturally occurring, easily available biopolymer. Recently, chitin and its

derivatives have taken noticeable attention as they have potential to cure many diseases. in

this review the medical applications of chitin and its derivatives have been discussed such as

anti-cancer activity, anti-microbial property, immunoadjuvant, wound healing and targeted

drug delivery system. The derivatives of chitin being more water soluble than chitin are more

applicable than chitin itself. This study will attract people to use chitin products for treatment

of different diseases and will facilitate continuously in future to improve the quality of human

life.

6 References

[1] Lavall RL, Assis OB, Campana-Filho SP. β-Chitin from the pens of Loligo sp.:

Extraction and characterization. Bioresour. Technol. 2007;98(13): 2465-72.

[2] Abdou ES, Nagy KS, Elsabee MZ. Extraction and characterization of chitin and

chitosan from local sources. Bioresour. Technol. 2008;99(5): 1359-67.

[3] Ngo DN, Kim MM, Kim SK. Chitin oligosaccharides inhibit oxidative stress in live

cells. Carbohydr.Polym. 2008;74(2): 228-34.

[4] Aam BB, Heggset EB, Norberg AL, Sørlie M, Vårum KM, Eijsink VG. Production of

chitooligosaccharides and their potential applications in medicine. Mar. Drugs.

2010;8(5): 1482-517.

[5] Jenkins DW, Hudson SM. Review of vinyl graft copolymerization featuring recent

advances toward controlled radical-based reactions and illustrated with chitin/chitosan

trunk polymers. Chem. Rev. 2001;101(11): 3245-74.


[6] Muzzarelli RA. Chitins and chitosans as immunoadjuvants and non-allergenic drug

carriers. Mar. Drugs. 2010;8(2): 292-312.

[7] Lodhi G, Kim YS, Hwang JW, Kim SK, Jeon YJ, Je JY, Ahn CB, Moon SH, Jeon

BT, Park PJ. Chitooligosaccharide and its derivatives: preparation and biological

applications. Biomed Res. Int. 2014.

[8] Reddy N, Yang Y. Innovative bio bers from renewable resources. Springer; 2015.

[9] Agnihotri SA, Mallikarjuna NN, Aminabhavi TM. Recent advances on chitosan-based

micro-and nanoparticles in drug delivery. J Controll Release. 2004;100(1): 5-28.

[10] Kumar MR, Muzzarelli RA, Muzzarelli C, Sashiwa H, Domb AJ. Chitosan chemistry

and pharmaceutical perspectives. Chem. Rev. 2004;104(12): 6017-84.

[11] Park BK, Kim MM. Applications of chitin and its derivatives in biological medicine.

Int JMol Sci. 2010;11(12): 5152-5164.

[12] Ibrahim HM, Salama MF, El‐Banna HA. Shrimp's waste: Chemical composition,

nutritional value and utilization. Nahrung. 1999;43(6): 418-23.

[13] Roberts GA. Chitin chemistry. Macmillan International Higher Education; 1992.

[14] Minke RA, Blackwell J. The structure of α-chitin. J. Mol. Biol. 1978;120(2): 167-81.

[15] Zhang M, Haga A, Sekiguchi H, Hirano S. Structure of insect chitin isolated from

beetle larva cuticle and silkworm (Bombyx mori) pupa exuvia. Int. J. Biol. Macromol.

2000;27(1): 99-105.

[16] Hunt S, El Sherief A. A periodic structure in the ‘pen’chitin of the squid Loligo

vulgaris. Tissue Cell. 1990;22(2): 191-7.


[17] Methacanon P, Prasitsilp M, Pothsree T, Pattaraarchachai J. Heterogeneous N-

deacetylation of squid chitin in alkaline solution. Carbohydr. Polym. 2003;52(2): 119-

23.

[18] Al Sagheer FA, Al-Sughayer MA, Muslim S, Elsabee MZ. Extraction and

characterization of chitin and chitosan from marine sources in Arabian Gulf.

Carbohydr. Polym. 2009;77(2): 410-419.

[19] G. Hall, S.S.-A. Chitin and chitosan. London: Elsevier; 1992.

[20] Jung WJ, Jo GH, Kuk JH, Kim KY, Park RD. Extraction of chitin from red crab shell

waste by cofermentation with Lactobacillus paracasei subsp. tolerans KCTC-3074 and

Serratia marcescens FS-3. Appl. Microbiol. Biotechnol. 2006;71(2): 234–237

[21] Suzuki K, Mikami T, Okawa Y, Tokoro A, Suzuki S, Suzuki M. Antitumor effect of

hexa-N-acetylchitohexaose and chitohexaose. Carbohydr. Res. 1986;151:403-8.

[22] Kobayashi M, Watanabe T, Suzuki S, Suzuki M. Effect of N-acetylchitohexaose

against Candida albicans infection of tumor-bearing mice. Microbiol.Immunol.

1990;34(5): 413-26.

[23] Maeda Y, Kimura Y. Antitumor effects of various low-molecular-weight chitosans

are due to increased natural killer activity of intestinal intraepithelial lymphocytes in

sarcoma 180–bearing mice. J. Nutr. 2004;134(4): 945-50.

[24] Kimura Y, Okuda H. Prevention by chitosan of myelotoxicity, gastrointestinal

toxicity and immunocompetent organic toxicity induced by 5‐fluorouracil without

loss of antitumor activity in mice. Jpn J Cancer Res. 1999;90(7): S765-74.


[25] Quan H, Zhu F, Han X, Xu Z, Zhao Y, Miao Z. Mechanism of anti-angiogenic

activities of chitooligosaccharides may be through inhibiting heparanase activity.

Med. Hypotheses. 2009;73(2): 205-206.

[26] Karagozlu MZ, Kim JA, Karadeniz F, Kong CS, Kim SK. Anti-proliferative effect of

aminoderivatized chitooligosaccharides on AGS human gastric cancer cells. Process

Biochem. 2010;45(9): 1523-1528.

[27] Prashanth KH, Tharanathan RN. Depolymerized products of chitosan as potent

inhibitors of tumor-induced angiogenesis. , Biochim.Biophys.Acta Gen. Subjects.

2005;1722(1): 22-29.

[28] Wu H, Yao Z, Bai X, Du Y, Ma X. Chitooligosaccharides inhibit nitric oxide

mediated migration of endothelial cells in vitro and tumor angiogenesis in vivo.

Carbohydr.Polym. 2010;82(3): 927-32.

[29] Ellouz F, Adam A, Ciorbaru R, Lederer E. Minimal structural requirements for

adjuvant activity of bacterial peptidoglycan derivatives. Biochem. Biophys. Res.

Commun. 1974;59(4): 1317-1325.

[30] Azuma I, Sugimura K, Taniyama T, Yamawaki M, Yamamura Y, Kusumoto S,

Okada S, Shiba T. Adjuvant activity of mycobacterial fractions. Immunological

properties of synthetic N-acetylmuramyldipeptide and related compounds. Infect.

Immun. 1976;14: 18-27.

[31] Nishimura K, Nishimura S, Nishi N, Saiki I, Tokura S, Azuma I. Immunological

activity of chitin and its derivatives. Vaccine. 1984;2(1): 93-99.


[32] Nishimura SI, Nishi N, Tokura S, Nishimura K, Azuma I. Bioactive chitin derivatives.

Activation of mouse-peritoneal macrophages by O-(carboxymethyl) chitins. ,

Carbohydr. Res. 1986;146(2): 251-8.

[33] Ober C, Tan Z, Sun Y, Possick JD, Pan L, Nicolae R, Radford S, Parry RR,

Heinzmann A, Deichmann KA, Lester LA. Effect of variation in CHI3L1 on serum

YKL-40 level, risk of asthma, and lung function. N. Eng1. J. Med. 2008;358(16):

1682-1691.

[34] Shibata Y, Metzger WJ, Myrvik QN. Chitin particle-induced cell-mediated immunity

is inhibited by soluble mannan: mannose receptor-mediated phagocytosis initiates IL-

12 production. J. Immunol. 1997;159(5): 2462-7.

[35] Shibata Y, Foster LA, Metzger WJ, Myrvik QN. Alveolar macrophage priming by

intravenous administration of chitin particles, polymers of N-acetyl-D-glucosamine,

in mice. Infect immune. 1997;65(5): 1734-1741.

[36] Bourbouze R, Raffi F, Dameron G, Hali-Miraftab H, Loko F, Vilde JL. N-acetyl-β-D-

Glucosaminidase (NAG) isoenzymes release from human monocyte-derived

macrophages in response to zymosan and human recombinant interferon-γ. Clin.

Chim. Acta. 1991;199(2): 185-194.

[37] Rabea EI, Badawy ME, Stevens CV, Smagghe G, Steurbaut W. Chitosan as

antimicrobial agent: applications and mode of action. Biomacromolecules. 2003;4(6):

1457-65.

[38] Li P, Poon YF, Li W, Zhu HY, Yeap SH, Cao Y, Qi X, Zhou C, Lamrani M,

Beuerman RW, Kang ET. A polycationic antimicrobial and biocompatible hydrogel

with microbe membrane suctioning ability. Nat. Mater. 2011;10(2): 149-56.


[39] Dai T, Tanaka M, Huang YY, Hamblin MR. Chitosan preparations for wounds and

burns: antimicrobial and wound-healing effects. Expert Re v Anti Infect Ther. 2011

Jul 1;9(7):857-79.

[40] No HK, Park NY, Lee SH, Meyers SP. Antibacterial activity of chitosans and

chitosan oligomers with different molecular weights. Int. J. Food Microbiol.

2002;74(1-2): 65-72.

[41] Kozen BG, Kircher SJ, Henao J, Godinez FS, Johnson AS. An alternative hemostatic

dressing: comparison of CELOX, HemCon, and QuikClot. Acad Emerg Med.

2008;15(1): 74-81.

[42] Millner RW, Lockhart AS, Bird H, Alexiou C. A new hemostatic agent: initial life-

saving experience with Celox (chitosan) in cardiothoracic surgery. Ann. Thorac. Surg.

2009;87(2): e13-14.

[43] Robson MC. Wound infection: a failure of wound healing caused by an imbalance of

bacteria. Surg Clin North Am. 1997;77(3): 637-650.

[44] Ong SY, Wu J, Moochhala SM, Tan MH, Lu J. Development of a chitosan-based

wound dressing with improved hemostatic and antimicrobial properties. Biomaterials.

2008;29(32): 4323-32.

[45] Dai T, Tegos GP, Burkatovskaya M, Castano AP, Hamblin MR. Chitosan acetate

bandage as a topical antimicrobial dressing for infected burns. Antimicrob. Agents

Chemother. 2009;53(2): 393-400.

[46] Çakmak A, Çirpanli Y, Bilensoy E, Yorganci K, Çaliş S, Saribaş Z, Kaynaroğlu V.

Antibacterial activity of triclosan chitosan coated graft on hernia graft infection

model. Int. J. Pharm. 2009;381(2): 214-9.


[47] Boateng JS, Matthews KH, Stevens HN, Eccleston GM. Wound healing dressings and

drug delivery systems: a review. , J. Pharm. Sci. 2008;97(8): 2892-2923.

[48] Sinha VR, Singla AK, Wadhawan S, Kaushik R, Kumria R, Bansal K, Dhawan S.

Chitosan microspheres as a potential carrier for drugs. Int. J. Pharm. 2004;274(1-2):

1-33.

[49] Denkbas EB, Ottenbrite RM. Perspectives on: chitosan drug delivery systems based

on their geometries. J. Bioact. Compat. Polym. 2006;21(4): 351-68.

[50] Iwamoto T. Clinical application of drug delivery systems in cancer chemotherapy:

review of the efficacy and side effects of approved drugs. Biol Pharm Bull. 2013 May

1;36(5): 715-8.

[51] Mao HQ, Roy K, Troung-Le VL, Janes KA, Lin KY, Wang Y, August JT, Leong

KW. Chitosan-DNA nanoparticles as gene carriers: synthesis, characterization and

transfection efficiency. J Control Release. 2001;70(3): 399-421.

[52] Yuan Y, Tan J, Wang Y, Qian C, Zhang M. Chitosan nanoparticles as non-viral gene

delivery vehicles based on atomic force microscopy study. Acta Biochim Biophys Sin.

2009;41(6): 515-26.

[53] De Campos AM, Sanchez A, Alonso MJ. Chitosan nanoparticles: a new vehicle for

the improvement of the delivery of drugs to the ocular surface. Application to

cyclosporin A. Int. J. Pharm. 2001;224(1-2): 159-68.

[54] Janes KA, Fresneau MP, Marazuela A, Fabra A, Alonso MJ. Chitosan nanoparticles

as delivery systems for doxorubicin. J Control Release. 2001;73(2-3): 255-267.

[55] Xu Y, Du Y. Effect of molecular structure of chitosan on protein delivery properties

of chitosan nanoparticles. Int. J. Pharm. 2003;250(1): 215-26.


Figure 1. Sources of chitin.

Figure 2. Medicinal applications of chitin and its derivatives.


Figure 3. Wound healing applications of chitosan.

You might also like