Hyaluronan: from biomimetic to industrial business strategy

22

Transcript of Hyaluronan: from biomimetic to industrial business strategy

INFORMATION FOR AUTHORS

Full details of how to submit a manuscript for publication in Natural Product Communications are given in Information for Authors on our Web site

http://www.naturalproduct.us.

Authors may reproduce/republish portions of their published contribution without seeking permission from NPC, provided that any such republication is

accompanied by an acknowledgment (original citation)-Reproduced by permission of Natural Product Communications. Any unauthorized reproduction, transmission or storage may result in either civil or criminal liability.

The publication of each of the articles contained herein is protected by copyright. Except as allowed under national ―fair use‖ laws, copying is not permitted by any means or for any purpose, such as for distribution to any third party (whether by sale, loan, gift, or otherwise); as agent (express or implied) of any third

party; for purposes of advertising or promotion; or to create collective or derivative works. Such permission requests, or other inquiries, should be addressed

to the Natural Product Inc. (NPI). A photocopy license is available from the NPI for institutional subscribers that need to make multiple copies of single articles for internal study or research purposes.

To Subscribe: Natural Product Communications is a journal published monthly. 2011 subscription price: US$1,995 (Print, ISSN# 1934-578X); US$1,995 (Web edition, ISSN# 1555-9475); US$2,495 (Print + single site online); US$595 (Personal online). Orders should be addressed to Subscription Department,

Natural Product Communications, Natural Product Inc., 7963 Anderson Park Lane, Westerville, Ohio 43081, USA. Subscriptions are renewed on an annual

basis. Claims for nonreceipt of issues will be honored if made within three months of publication of the issue. All issues are dispatched by airmail throughout the world, excluding the USA and Canada.

NPC Natural Product Communications

EDITOR-IN-CHIEF

DR. PAWAN K AGRAWAL Natural Product Inc.

7963, Anderson Park Lane,

Westerville, Ohio 43081, USA

[email protected]

EDITORS

PROFESSOR ALESSANDRA BRACA

Dipartimento di Chimica Bioorganicae Biofarmacia,

Universita di Pisa,

via Bonanno 33, 56126 Pisa, Italy

[email protected]

PROFESSOR DEAN GUO

State Key Laboratory of Natural and Biomimetic Drugs,

School of Pharmaceutical Sciences,

Peking University,

Beijing 100083, China [email protected]

PROFESSOR YOSHIHIRO MIMAKI

School of Pharmacy,

Tokyo University of Pharmacy and Life Sciences,

Horinouchi 1432-1, Hachioji, Tokyo 192-0392, Japan

[email protected]

PROFESSOR STEPHEN G. PYNE

Department of Chemistry

University of Wollongong

Wollongong, New South Wales, 2522, Australia

[email protected]

PROFESSOR MANFRED G. REINECKE

Department of Chemistry,

Texas Christian University,

Forts Worth, TX 76129, USA

[email protected]

PROFESSOR WILLIAM N. SETZER

Department of Chemistry

The University of Alabama in Huntsville

Huntsville, AL 35809, USA

[email protected]

PROFESSOR YASUHIRO TEZUKA

Institute of Natural Medicine

Institute of Natural Medicine, University of Toyama,

2630-Sugitani, Toyama 930-0194, Japan

[email protected]

PROFESSOR DAVID E. THURSTON Department of Pharmaceutical and Biological Chemistry,

The School of Pharmacy,

University of London, 29-39 Brunswick Square,

London WC1N 1AX, UK

[email protected]

ADVISORY BOARD

Prof. Berhanu M. Abegaz

Gaborone, Botswana

Prof. Viqar Uddin Ahmad

Karachi, Pakistan

Prof. Øyvind M. Andersen

Bergen, Norway

Prof. Giovanni Appendino

Novara, Italy

Prof. Yoshinori Asakawa

Tokushima, Japan

Prof. Lee Banting

Portsmouth, U.K.

Prof. Julie Banerji

Kolkata, India

Prof. Alejandro F. Barrero

Granada, Spain

Prof. Anna R. Bilia

Florence, Italy

Prof. Maurizio Bruno

Palermo, Italy

Prof. César A. N. Catalán

Tucumán,Argentina

Prof. Josep Coll

Barcelona, Spain

Prof. Geoffrey Cordell

Chicago, IL, USA

Prof. Cristina Gracia-Viguera

Murcia, Spain

Prof. Duvvuru Gunasekar

Tirupati, India

Prof. A.A. Leslie Gunatilaka

Tucson, AZ, USA

Prof. Kurt Hostettmann

Lausanne, Switzerland

Prof. Martin A. Iglesias Arteaga

Mexico, D. F, Mexico

Prof. Jerzy Jaroszewski

Copenhagen, Denmark

Prof. Leopold Jirovetz

Vienna, Austria

Prof. Karsten Krohn

Paderborn, Germany

Prof. Hartmut Laatsch

Gottingen, Germany

Prof. Marie Lacaille-Dubois

Dijon, France

Prof. Shoei-Sheng Lee

Taipei, Taiwan

Prof. Francisco Macias

Cadiz, Spain

Prof. Imre Mathe

Szeged, Hungary

Prof. Joseph Michael

Johannesburg, South Africa

Prof. Ermino Murano

Trieste, Italy

Prof. M. Soledade C. Pedras

Saskatoon, Cnada

Prof. Luc Pieters

Antwerp, Belgium

Prof. Peter Proksch

Düsseldorf, Germany

Prof. Phila Raharivelomanana

Tahiti, French Plynesia

Prof. Monique Simmonds

Richmond, UK

Prof. Valentin Stonik

Vladivostok, Russia

Prof. Winston F. Tinto

Barbados, West Indies

Prof. Karen Valant-Vetschera

Vienna, Austria

Prof. Peter G. Waterman

Lismore, Australia

HONORARY EDITOR

PROFESSOR GERALD BLUNDEN

The School of Pharmacy & Biomedical Sciences,

University of Portsmouth,

Portsmouth, PO1 2DT U.K.

[email protected]

Hyaluronan: From Biomimetic to Industrial Business

Strategy

Erminio Muranoa,b

, Danilo Perina, Riaz Khan

a and Massimo Bergamin

a,b

aPROTOS Research Institute, via Flavia 23/1c/o BIC Incubatori FVG, 34148, Trieste, Italy

bNEALYS srl, via Flavia 23/1c/o BIC Incubatori FVG, 34148, Trieste, Italy

[email protected]

Received: December 20th

, 2010; Accepted: February 2nd

, 2011

Hyaluronan (hyaluronic acid) is a naturally occurring polysaccharide of a linear repeating disaccharide unit consisting of β-(1→4)-linked

D-glucopyranuronic acid and β-(1→3)-linked 2-acetamido-2-deoxy-D-glucopyranose, which is present in extracellular matrices, the synovial

fluid of joints, and scaffolding that comprises cartilage.

In its mechanism of synthesis, its size, and its physico-chemical properties, hyaluronan is unique amongst other glycosaminoglycans. The

network-forming, viscoelastic and its charge characteristics are important to many biochemical properties of living tissues. It is an important

pericellular and cell surface constituent; its interaction with other macromolecules such as proteins, participates in regulating cell behavior

during numerous morphogenic, restorative, and pathological processes in the body. The knowledge of HA in diseases such as various forms

of cancers, arthritis and osteoporosis has led to new impetus in research and development in the preparation of biomaterials for surgical

implants and drug conjugates for targeted delivery.

A concise and focused review on hyaluronan is timely. This review will cover the following important aspects of hyaluronan: (i) biological

functions and synthesis in nature; (ii) current industrial production and potential biosynthetic processes of hyaluronan; (iii) chemical

modifications of hyaluronan leading to products of commercial significance; and (iv) and the global market position and manufacturers of

hyaluronan.

Keywords: Hyaluronan, hyaluronic acid, biosynthesis, tissue engineering, antitumor drugs, crosslinked HA, global markets.

Hyaluronan (also known as hyaluronic acid) (HA) is a

naturally occurring polysaccharide of a linear repeating

disaccharide unit consisting of β-(1→4)-linked D-

glucopyranuronic acid and β-(1→3)-linked 2-acetamido-2-

deoxy-D-glucopyranose, which is present in extracellular

matrices, the synovial fluid of joints, and scaffolding that

comprises cartilage. However, other organisms, such as

bacterial pathogens and virus infected micro algae, are able

to synthesis this polysaccharide. HA was first isolated

from the vitreous body of the eye by Mayer and Palmer [1]

and then found in all vertebrates tissues, from skin to

synovial fluid between joins, from umbilical cord to skin

and cartilage [2-5].

Despite its simplistic structure hyaluronan behaves quite

differently from other glycosaminoglycans in its

mechanism of synthesis, its size, and its physico-chemical

properties. The network-forming, viscoelastic and its

charge characteristics are important to many biochemical

properties of living tissues. It is an important pericellular

and cell surface constituent which, through interaction

with other macromolecules such as proteins, participates in

regulating cell behavior during numerous morphogenic,

restorative and pathological processes in the body. The

knowledge of such biological functions of hyaluronan has

led to the preparation of biomaterials for surgical implants

and drug conjugates for targeted delivery.

The interest in the role of hyaluronan diseases such as

various forms of cancers, arthritis and osteoporosis has led

to new impetus in research and development. Our

laboratory has been involved in the study of chemistry and

physico-chemical properties of hyaluronan with a view to

develop biomaterials and drugs conjugates.

As a general arrangement of the review the subject is

divided into: biological function and synthesis in nature;

industrial production of hyaluronan; chemical modification

of hyaluronan; and market position of hyaluronan.

Biological function and synthesis in nature

Hyaluronan was first isolated from the vitreous body of the

eye by Mayer and Palmer [1] and then found in all

vertebrates tissues, from skin to synovial fluid between

joins, from umbilical cord to skin and cartilage [2-9].

Hyaluronan is a glycosaminoglycan that has a number of

biological functions in vertebrates, being essential for

tissue and organ integrity and function. Hyaluronan

molecules are involved in the activation of signaling

NPC Natural Product Communications 2011

Vol. 6

No. 4

555 - 572

556 Natural Product Communications Vol. 6 (4) 2011 Murano et al.

pathways that control cell proliferation, differentiation,

adhesion and migration [10-20].

The primary structural function of HA is related to its

viscoelastic and hygroscopic properties. It acts as shock-

adsorber and offers lubrication to the joints, creates a

protective and flexible layer between tissues, influence cell

responses during a number of processes, inflammation and

cancer [21-28]. A wide range of different cell responses

are evoked by HA molecules which different molecular

weights [29]. In general, high molecular weight

hyaluronan inhibits cell differentiation and promotes cell

proliferation, being involved in tissue regeneration, wound

healing, epithelial integrity and embryogenesis. Lower

molecular weights HA are anti-angiogenic, immuno-

suppressive and hamper the differentiation by limiting the

cell-cell interactions or ligand access to cell receptors.

Finally, HA oligomers showed angiogenic and pro-

inflammatory activity, increase immune response and are

capable to slow down growth of a number of tumors in

vivo [30-37].

HA mediates his biological function through specific

protein receptors present on the different cell surface,

which include CD44, HARE, RHAMM, LYVE [29,38-43].

In nature, hyaluronan is synthesized by specific

glycosyltransferases (HA synthases or HAse) that

polymerizes the activated donor uridine diphospho- sugars

UDP-glucuronic acid and UDP-N-acetyl-glucosamine into

hyaluronan [44-47]. In nature, hyaluronan was shown to be

synthesized also by a small number of microbial pathogens

such us Pasteurella multocida and the Lancefield group A

and C streptococci among which Streptococcus equi,

the human pathogen Streptococcus pyogenes and the

bovine pathogen Streptococcus uberis. [48-58]. These

microorganisms adopted the strategy to encapsulate their

cells with hyaluronan in order to perfectly disguise from

the host animal defense system and allow the adhesion and

colonization. In Streptococcus and Pasteurella, the HA

capsules represent virulence factors affording to the

bacteria a stealth function against the recognition from the

host immune system [45,59-61]. On the contrary to other

bacterial capsule polysaccharides triggering the antibody

immune response, HA is not immunogenic because it is a

normal component of the host body conferring to these

pathogen bacteria an efficient molecular camouflage.

Moreover, the HA capsule provide a protection against the

reactive oxides released by leukocytes [62] and facilitate the

bacteria migration through epithelial layer into tissues [63].

In Streptococcus equi subsp. zooepidemicus hyaluronic

acid capsular material contributes to adherence properties

of the pathogen possibly conferring to the bacteria the

resistance to phagocytosis by macrophages [64-66].

The importance of the capsule in pharyngeal colonization

and other inflammatory syndromes caused by

Streptococcus pyogenes may reflect the role of hyaluronan

as a specific ligand for attachment of Streptococcus

pyogenes to the hyaluronan-binding protein CD44 on

pharyngeal epithelial cells [67,68].

Pasteurella multocida, a prevalent animal pathogen,

employs a hyaluronan polysaccharide capsule to avoid host

defenses. This pathogen is associated with various diseases

in many animal species; a number of studies suggested that

there is a correlation between the capsule and the virulence

of P. multocida, for example in the pathogenesis of fowl

cholera [52,69-74].

This biomimetic strategy of prokaryotic organisms

efficiently exploits the chemical identity of the bacterial

capsular HA molecules to that of the eukaryotic host

extracellular matrix. Such a biomimetic strategy adopted

by specialized prokaryotic cells to proliferate into an

animal host organism has the same goal as many types of

endogenous cells like tumor cells to invade and proliferate

into normal tissues. In fact, a large number of tumor cells

over express membrane receptors for hyaluronan in order

to more efficiently interact with this major component of

the extracellular matrix to easily spread and proliferate into

the tissues [75,76].

The existence of hyaluronan outside the animals and their

pathogens has been demonstrated in green microalgae

infected by specialized viruses. Chlorovirus is a group of

virus of Phycodnaviridae family that infects the eukaryotic

microalgae Chlorella [77,78]. Chlorovirus dsDNA genome

unusually encodes enzymes involved in sugar metabolism.

The 330-kilobase genome of the chlorovirus PBCV-1

encode other than fructose-6-phosfate aminotransferase

and UDP-glucose dehydrogenase also a HA synthase,

resulting in the production of an HA capsule surrounding

the Chlorella infected cells. However, some chlorovirus

contain both HA synthase and chitin synthase genes and

form both hyaluronan and chitin, an N-acetyl-glucosamine

based polysaccharide found in the crustaceans shell, in the

insect exoskeletons and fungi cell wall [79], on the surface

of their infected cells [80-82].

The function of a HA capsule in chlorovirus infected

chlorella cells is still unknown; however, since the genes

encoding for HA synthesis are transcribed early in

PBCV-1 infection and HA accumulates on the external

surface of the infected chlorella cells, hyaluronan synthesis

might be related to the virus life cycle and the recognition

of infected cells. The distribution of hyaluronan synthases

from vertebrates to bacterial pathogens and algal viruses

makes these fascinating glycosyltransferases an interesting

evolutionary case which can advance the knowledge on the

biological functions of a typical natural molecule like

hyaluronan.

Industrial production of hyaluronan

HA has been traditionally produced by extracting from

rooster combs and bovine vitreous humor The process is

difficult, particularly to ensure the biological safety and to

Review on hyaluronan Natural Product Communications Vol. 6 (4) 2011 557

control the molecular weight of the product [3]. Hence,

since 1980‘s, the commercial production of HA has moved

to the fermentation process using streptococcal bacteria.

The most used strains are S. equi subsp. equi and S. equi

subsp. zooepidemicus that naturally achieve good HA

production rate.

Improved HA producer streptococcal strain has been

obtained by random mutagenesis followed by a serial

selection of the mutant clones. The aim of the strain

selection was mainly oriented to the reduction of the HA-

depolymerising enzyme (hyaluronidase-negative strains)

and other extracellular proteins such as the virulence factor

streptolysins, the exotoxins responsible for the β-hemolysis

(non-hemolytic strains) [83] as well as the production of

high molecular weight HA [84,85]. Several studies were

performed in order to improve the HA yield, molecular

weight and reduce the polydispersity by an accurate

control of the growing conditions. Unfortunately,

Streptococci are lactic acid bacteria that demand expensive

complex growth medium containing yeast or animal

extract, peptone and serum supplemented with an high

glucose content [83,86,87].

The HA biosynthesis require a large amount of energy and

compete with the cell growth because of the large

pathways overlapping with the cell wall biosynthesis

(Figure 1). Indeed, when unlimited resource are available,

the optimal growth conditions is observed at 37°C and

around pH 7 while, the highest HA productivity and

molecular weight is observed at suboptimal growing

conditions. Limiting the resources available, in particular

glucose, first occurs a decline in HA productivity

followed by the HA molecular weight [55]. Streptococci

are facultative anaerobes incapable to oxidative

phosphorylation however, aerobic culturing condition it is

known to greatly improve the HA production. These

effects are probably the results of improved energy

efficiency when the NADH oxidase (NOX) removes the

excess of NADH in presence of oxygen [88].

Metabolically engineered Streptococcus strain with an

increased NOX activity resulted in 33% improvement of

ATP yield leading to a 15% improvement in biomass.

Unfortunately, any HA yield or molecular weight

improvement was not observed [89].

Even under the best conditions, and the use of continuous

cultivation, the HA yield does not exceed the practical

limit of 5-10 g/L. Further improvement requires genetic

engineering of the producer microorganism. Partial or

complete genome sequence of the most relevant HA

producing microorganisms is now available including S.

equi [90,91], S. pyogenes [92,93] and Pasteurella

multocida [94]. These information, combined with the

advanced vectors developed for a wide number of hosting

cell, allows the metabolic engineering of the HA pathway.

Moreover, it is possible to transfer the HA synthesis

capability from natural producer to non-producer hosting

cells. Even though the metabolic engineering and genomic

insertion for the improvement of HA production is

possible in streptococci, the same result was easier

achieved heterologus host microorganisms. Other

advantages of heterologus HA synthesis reside in the

exploitation of microorganism generally considered as safe

for human health and the easier culture preparation and

processing.

The enzymes involved in the HA pathway in streptococci

are encoded in the bacterial genome as a monocistronic

has operon. The has operon includes three genes: hasA

that encodes for the HA synthetase [95], hasB that

encode for UDP-glucose dehydrogenase [96] and hasC that

Figure 1: Schematic representation of the streptococci HA synthesis pathway. The enzymes and well as the correspondent gene locus is in italic; the has operon genes

(hasA, hasB and hasC) is underlined.

558 Natural Product Communications Vol. 6 (4) 2011 Murano et al.

encodes for UDP-glucose pyrophosphorylase [97].

Heterologus expression of HA synthetatse (hasA gene in

streptococci) in host bacteria such us Escherichia coli,

Enterococcus faecalis or Bacillus subtilis, has shown to be

sufficient to direct the production of HA [70,98]. However,

since HA and cell wall biosynthesis has partially common

pathway (Figure 1), the depletion of sugar precursor

caused by the high activity of HA synthetase would stop

the cell from growing. To overcome this problem, at least

a second gene hasB or its homologue must to be

introduced at the same time [40,98].

Several microorganisms have been tried for the

transformation into hosting cells for HA synthesis:

Escherichia coli, Bacillus subtilis, Lactococcus lactis and

Agrobacterium sp. The initial genetic modifications were

performed on E. coli by the cloning and expression of the

hasA gene from S. equi encoding for HA synthetase.

However, the experiments demonstrated that, over

expression of HA synthetase itself severely compromise

the cells ability to growth [95]. More recently, a better

result in the E. coli HA production was achieved by the co-

expression of HA synthetase hasA gene (derived from P.

multocida or S. pyogenes) respectively with hasB

homologue (UDP-glucose dehydrogenase) or hasB + hasC

homologues (UDP-glucose dehydrogenase + UDP-glucose

pyrophosphorylase) from E. coli. The aim was to improve

the intracellular availability of sugar precursor and prevent

the inhibition of cell growing. The genes were organized

into an artificial operon designed and constructed onto the

backbone of proper plasmidic vectors. The HA yield was

about 2 g/L but raised up to 3.8 g/L when the culture

media was supplemented with glucosamine, suggesting

close connection between energy metabolism and

precursor supply. Even if the HA production was lower

than the streptococci strains or others recombinant

microorganisms, this work suggests suggest engineering

routs to be exploited and the potential use of E. coli for

industrial HA production [99,100].

Interesting was the works performed onto food grade

microorganisms Agrobacterium sp. and Lactococcus lactis

[101,102]. Agrobacterium is already used in the production

of curdlan for food applications and is generally

considered as safe. In 2007, Mao and Chen, successfully

transfered HA synthetase gene (hasA) from P. multocida

and an UDP-glucose dehydrogenase homologue gene

(hasB) from E. coli into Agrobacterium host. The co-

expression of the two enzymes enables the host

microorganism to produce HA up to 0.3 g/L. Even if the

HA production yield was considerably lower than standard

streptococci, the recombinant synthesis of HA in

Agrobacterium specie was the first example of HA

production into Gram-negative bacteria that naturally

produce a food product [102].

A similar result was obtained by Chien and Lee [101] on

Lactococcus lactis, a food grade Gram-positive bacterium

extensively used in the production of buttermilk and

cheese. HA synthetase gene hasA from S. equi subsp.

zooepidemicus was expressed alone or on combination

with hasB gene (UDP-glucose dehydrogenase). The

authors use a nisin-controlled expression vector (NICE) in

order to separate the cell expansion from the HA

production. When the desired rate cell density was

reached, nisin was added to the culture to induce the

expression of the foreign genes and finally, the HA

production. The HA yield obtained was 0.08 g/L and 0.65

g/L respectively when HA synthetase (hasA) was

expressed alone or in combination with UDP-glucose

dehydrogenase (hasB). Even the production yield needs

improvement, because the host microorganism is a food-

grade lactic acid bacteria, the HA production has high

potential to applied in the field of food and biomedical

industry [101].

Novozymes obtained a successful result in the recombinant

production of HA in Bacillus subtilis [103-105]. The

microorganism is a well known non-pathogenic, Gram-

positive bacterium that was extensively studied and with a

wide range of vector available that make it an obvious

potential host for HA production. The authors produce a

series of expression cassette containing the gene hasA (HA

synthetase from S. equisiilis) alone or in combination with

B. subtilis homologues of hasB (tuaD in Bacillus,

encoding for UDP-glucose dehydrogenase), hasC (gtaB in

Bacillus, encoding for UDP-glucose pyrophosphorylase),

hasD (gcaD in Bacillus, encoding for Uridyltransferase/N-

acetyltransferase bifunctional enzyme). The expression

cassettes were cloned into appropriate vectors to construct

artificial operons that were stable introduced into a specific

locus of Bacillus subtilis host genome. The production

yield obtained by the transformed Bacillus strain resulted

to be comparable with the streptococci strains when hasA

and tuaD genes were co-expressed by the artificial operon

[104]. Interestingly, the overexpression of hasA alone does

not seem to hinder the cell growth. Indeed, the authors

report that, in contrast to E. coli, the Bacillus strain

expressing HA synthetase alone resulted in HA production

even if lower than others transformed strains. The HA

production was dramatically improved by the co-

expression of hasA and tuaD into the same operon. The

meaning of the tuaD needing reside in the evidence that,

endogenous tuaD gene is expressed only under phosphate

starvation [55]. B. subtilis is inherently more metabolic

robust than E. coli, at least regarding the HA synthesis,

allowing a simultaneous support of both cell growing and

HA synthesis [105]. In spite of no benefit in terms of HA

yield and molecular weight compared to streptococci

fermentation, recovery from B. subtilis is much easier

because HA is not cell-associated and there is no

contamination by streptococcal pathogenic factors [40].

As an alternative to the prokaryotic HA production, it is

possible to mention the HA synthesis occurring Chlorella

cells, a unicellular green algae, when infected by

Chlorovirus [106]. Experimentally, approximately 0.5-1

g/L of HA was recovered from infected Chlorella cells

Review on hyaluronan Natural Product Communications Vol. 6 (4) 2011 559

after 4 hours of infection [107]. The yield is lower than typically ones achieved in streptococcal fermentation but further implementation is possible to improve it [40].

Finally, it is interesting the work of Takeo and colleagues [108] on the cloning and expression of mammalian HA synthetase. The authors genetically modify the genome of the fruit fly Drosophila melanogaster, a non-HA-synthesizing animal, by introduction of the single gene encoding mouse HA synthetase. The expression of HA synthetase in vivo resulted in massive HA accumulation in the extracellular space of the Drosophila tissues and caused various morphological defect. The original aim of the work was the development of an in vivo model to study the biosynthetic machinery of the polysaccharides besides, this could represent an alternative route for the HA production in eukaryotic cells [108].

Chemical modification of hyaluronan

The interest in the role of hyaluronic acid in diseases such as various forms of cancers, arthritis and osteoporosis has led to new impetus in research and development. Our laboratory has been involved in the study of chemistry and physico-chemical properties of hyaluronic acid with a view to develop biomaterials and drugs conjugates [109].

The repeating disaccharide unit, β-(1→4)-linked D-gluco-pyranuronic acid and β-(1→3)-linked 2-acetamido-2-deoxy-D-glucopyranose, of hyaluronic acid contains two free hydroxyl groups at C-4,6 positions in the β-D-GlcpNAc moiety and in the β-D-GlcpA a carboxylic group at C-5’ and two free hydroxyl groups at C-2’,3’ positions, as shown in Figure 2. The chemical modifications on the hyaluronic acid backbone occur in these positions. In our laboratory, we have focused our research on regioselective modifications of the hydroxyl groups of HA. In particular, reactions at the 4- and 6-OH groups of the GlcNAc residues of the HA have led to a large number of well characterized derivatives.

Figure 2: Chemical structure of the repeating unit β-(1→4)-linked D-glucopyranuronic acid and β-(1→3)-linked 2-acetamido-2-deoxy-D-glucopyranose

Unlike simple carbohydrates, chemical reactions of hyaluronic acid usually do not result in discrete compounds; a certain proportion of the molecule may be partially substituted or unsubstituted.

High-resolution NMR experiments have been used to determine the position and the degree of substitution and in some cases to study on a qualitative basis the effect of specific substitution on the conformation of the molecule. Molecular weight and molecular weight distribution determination, using High Performance Size Exclusion Chromatography (HP-SEC), has been performed to ascertain the molecular integrity of the reaction products. The low solubility of hyaluronic acid in organic solvents has been overcome by converting it into different organic counterions such as tetrabutylammonium, pyridinium and collidinium salts [110]. Such a derivatization imparts complete solubility in organic solvents and allows the chemical reaction to occur.

6-halodeoxy-hyaluronic acid: Recently, HA was used as sodium salt without any derivatization in organic solvent (DMF) and in presence of methanesulfonyl chloride to generate regioselective 6-deoxy-6-chloro derivatives [111]. The reaction occurred in heterogeneous phase yielding halogenated HA derivatives suitable for further derivatization with drugs bearing nucleophilic groups. The complex methanesulfonyl chloride-DMF complex formed during the reaction permits selective replace of primary hydroxyl groups by chlorine [112].

Alkyl and Aryl Esters derivatives of HA: The carboxyl group of hyaluronic acid has been esterified by treatment with tetrabutylammonium salt in an aprotic solvent such as DMF or DMSO. Hyaluronic esters of ethyl, propyl, benzyl and dodecyl alcohols have been described as medical and pharmaceutical materials as medical sutures, films, microspheres, pellets, membranes, corneal shields and implants [113,114].

6-amino-6-deoxyhyaluronic acid: The amino-deoxy derivatives of carbohydrates are of interest because they are components of biological materials such as glycoproteins and bacterial polysaccharides. They are usually synthesized by catalytic reduction of the corresponding azido derivatives, which in turn can be prepared from the corresponding halodeoxy compounds [115]. A direct, high yielding, synthesis of 6-amino-6-deoxyhyaluronic acid has been achieved by selective amination of the C6-chlorinated HA in aqueous media under NH3 pressure [116].

Hyaluronic acid and methotrexate conjugate: Small molecule drugs such as antitumor compounds have been conjugated to synthetic and natural polymers. The advantages envisaged in this strategy are reduced toxicity, increased solubility and stability, localisation and controlled release of the drug. Methotrexate is used in the treatment of diseases such as inflammatory pathologies, autoimmune or neoplastic diseases. However, its therapeutic use is limited because of its high systemic toxicity. With a view to reduce the toxicity and enhance the efficacy of the drug, regioselective 6-O-methotrexyl-hyaluronic acid has been prepared by treatment of 6-chloro-6-deoxy HA with methotrexate in DMF [111].

OO O

OO HO

HO

OHN

H3C

HO

OH

O

Na

n

1'

2'

3'

4'5'

6'

1

23

4

5

6

560 Natural Product Communications Vol. 6 (4) 2011 Murano et al.

Hyaluronic acid and taxol conjugate: Taxol (paclitaxel),

an antileukemic and antitumor agent, was first isolated

from the bark of the Pacific yew tree, Taxus bravifolia;

has a limited solubility in water. Hyaluronic acid is over

expressed at sites of tumor and provides a matrix to

facilitate invasion. Hence, to overcome the solubility

problem and to target the tumor cells, a first attempt to

synthesize an hyaluronic acid conjugate of taxol was

performed by Luo et al. by linking the taxol 2‘-OH by

way of succinate ester to adipic dihydrazide-modified HA

(HA-ADH) [117].

Recently, Lee et al conjugated paclitaxel and HA utilizing

a novel solubilization method in a single organic phase.

Hydrophilic HA was completely dissolved in anhydrous

DMSO with addition of poly(ethylene glycol) (PEG) by

forming nano-complexes. Paclitaxel was then chemically

conjugated to HA in the DMSO phase via an ester linkage

without modifying extremely hydrophilic HA [118]. The

conjugate exhibited selective toxicity toward the human

cancer cell lines (breast, colon and ovarian) that are

known to express hyaluronic acid receptors; no toxicity

was noted against a mouse fibroblast cell line at the same

concentrations.

Hyaluronic acid and mitomycin C: Mitomycin C was

linked to the HA molecule by way of an amide

bond between the drug and the carboxyl group of the

D-glucuronic acid moiety. An amide linkage formation is a

dehydration reaction and requires anhydrous systems.

However, as hyaluronic acid is difficult to dissolve in an

anhydrous organic solvent, reaction conditions were

developed to use a water based system [119]. Other

strategies envisage the use of a succinylated HA as active

intermediate for the conjugation with the drug [120].

Hyaluronic acid and camptothecin conjugate: A novel

methodology for making drug conjugates using hyaluronan

as a carrier was recently developed. This strategy involves

a completely regioselective two-step synthesis of 6-amino-

6-deoxyhyaluronan, which is then easily functionalized

making use of a simple succinate linker [116].

Hyaluronic acid and other drugs: A number of

antitumor drugs, such as doxorubicin [121] and

daunorubicin, have been linked at the amino function of

the drug to the carboxylic group of an amino acid or a

peptide spacer arm forming an amide linkage, and then

through the terminal amino group of the amino acid or the

peptide to the carboxyl group of the HA.

Hyaluronic acid and methylprednisolone: Steroid-type

of anti-inflammatory drugs such as methylprednisolone

has proved to be efficacious in reducing symptoms

associated with osteoarthritis of the knee [122]. Alkyl and

aryl esters of HA have been used in the preparations

containing methylprednisolone, both as polymer

matrices in the form of microspheres wherein the drug is

physically incorporated, and as substrates on which

methylprednisiolone is chemically linked [123,124].

Hyaluronic acid and antioxidants: Reactive oxygen

species (ROS) such as superoxide radicals generated by

metabolic processes can depolymerise hyaluronic acid

[125]. It has been demonstrated that many anti-

inflammatory drugs and free radical scavengers are

efficacious in protecting hyaluronic acid depolymerisation

by free radicals [126-128].

Hyaluronic acid and propofol conjugate: 2,6-

Diisopropylphenol (propofol) is an anaesthetic for

intravenous administration whose efficacy as a free radical

scavenger is due to its ability to form stable radicals. Its

effectiveness as a scavenger for hydroxyl radicals

generated by xanthine oxidase has been demonstrated by

assessing in vitro the depolymerisation of hyaluronic acid

in artificial synovial fluid [129]. Low-molecular-mass

hyaluronic acid oligosaccharides have major biological

effects: notably, they appear to play a role in the

recruitment and activation of inflammatory macrophages,

which may result in an exacerbation of the inflammatory

process and tissue damage [130,131]. For this reason,

propofol was proposed as a shield for hyaluronic acid

[124].

Cross-linked Hyaluronic Acid-based materials: Hyaluronic acid is rapidly metabolised in vivo by enzymes

such as hyaluronidase [132] and by free radical oxidations

[131], which limit its use in native form as a biomaterial.

In, addition hyaluronic acid is highly soluble in water. In

order to overcome these limitations, the physico-chemical

properties of hyaluronic acid have been modified using a

variety of cross-linking reagents. These chemical

modifications have led to a number of hyaluronic acid

derivatives with a variety of physical formats such as

liquids of various viscosities, fibers, weaves, sponges and

fleeces. Cross-linked hyaluronic acids with special

properties and structure, such as different degradation rate,

different surface characteristics, different porosities, have

applications as tissue engineering scaffolds for the delivery

of cells, gene transfer, wound healing, post-surgical

adhesion prevention, and implantation of bioactive

compounds in vivo repair sites.

Ossipov et al. developed a new methodology exploiting a

protective group strategy based on initial mild cleavage of

a disulfide bond followed by elimination of the generated

2-thioethoxycarbonyl moiety ultimately affording free

amine-type functionality. The strategy therefore

encompasses a new approach for mild and highly

controlled functionalization of HA with both nucleophilic

and electrophilic chemoselective functionalities with the

emphasis for the subsequent conjugation and in situ

crosslinking [133].

Li et al. synthesized a polymeric nanogel, with tumor

targeting properties and a controllable phototoxicity,

utilizing a low molecular weight-hyaluronic acid

photosensitizer conjugate, resulting in the formulation of

self-organizing nanogels in aqueous solutions [134] while

Review on hyaluronan Natural Product Communications Vol. 6 (4) 2011 561

Kim et al synthesized hyaluronic acid/quantum dots

(QDots) conjugates. The reaction was performed through

amide bond between carboxyl groups of QDots and amine

groups of adipic acid dihydrazide modified HA (HA-

ADH) assessing the possibility of HA derivatives as

target-specific drug delivery carriers for the treatment of

liver diseases [135]. In the field of delivery systems

alkylamino hydrazide hyaluronic acid (HA) derivatives

were prepared to design new biocompatible films able to

release hydrophobic drugs. These films were found to be

more resistant to degradation by hyaluronidase compared

to solutions of unmodified HA at the same concentration

[136]. Bencherif et al developed a nanostructured

hydrogel made of thiolated HA and reacted with

biodegradable POEO300MA-co-PHEMA nanogels

derivatized with acryloyl groups. The resulting scaffold

was obtained through selective atom transfer radical

polymerization (ATRP) and Michael-type addition

reactions, affording a biocompatible matrix for cell and

protein encapsulation in both tissue engineering and drug

delivery applications [137]. Skoza et al exploited the

specificity of hyaluronate lyase to biodegrade HA and

introduce a double bond in the glucuronic acid moiety

followed by the generation of free aldehyde group via

ozonolysis and the subsequent reduction of the generated

ozonide. This gives rise to a new reactive HA substrate to

be derivatized with several molecules (such as biotin,

polymers, or proteins) with application in tissue

engineering and biomaterials production [138].

A strong commercial interest drives the extensive use of

HA hydrogels in cosmetics and tissue engineering. Due to

the frequent use of HA derivatives in aesthetic surgery,

novel, biocompatible, and non-toxic dermal fillers

hyaluronic acid (HA) hydrogels were studied. Yeom et al

successfully developed new hydrogel for tissue

augmentation applications. Instead of using highly

reactive cross-linkers such as divinylsulfone (DVS) [139]

for Hylaform, 1,4-butanediol diglycidyl ether (BDDE) for

Restylane, and 1,2,7,8-diepoxyoctane (DEO) for Puragen,

HA hydrogels were prepared by direct amide bond

formation between the carboxyl groups of HA and

hexamethylenediamine (HMDA) with an optimized

carboxyl group modification for effective tissue

augmentation [140]. Crosslinked hyaluronic acid

hydrogels have been specially designed and synthesized

to promote tissue repair. A range of glycidyl

methacrylate-hyaluronic acid (GMHA) conjugates have

been synthesized and photopolymerized to lead to a

number of crosslinked GMHA hydrogels with different

physical properties such as swelling, mesh size. As

expected, the amount of crosslinking and the degree of

degradation was dependent on the amount of the

conjugated methacrylate; the more crosslinking the less

degradation rate [141,142]. Su et al. developed an

hydrogel formed by oxidated hyaluronic acid (oxi-HA)

cross-linked with adipic acid dihydrazide (ADH). HA was

oxidized by sodium periodate to create aldehyde

functional groups, which could be cross-linked by ADH.

The result was a colorless, transparent and injectable

hydrogel as potential vitreous substitute [143]. Other type

of hydrogel were those made by Oldinsky et al,

characterized by both a hydrogel and a hydrophobic

polymer for biomedical applications. A series of

amphiphilic graft copolymers consisting of HA, a

glycosaminoglycan, and high-density polyethylene

(HDPE), that is, HA-co-HDPE, were fabricated.

Exploiting an esterification reaction between HA and

functionalized HDPE the resulting material was a

semicrystalline, insoluble powder applicable in saline

suspension or molded form, including orthopedic tissue

repair [144].

The extreme versatility of HA is emphazised also by its

employment in material science with carbon nanotubes.

Hybrid hyaluronic acid (HA) hydrogels with single wall

nanotubes (SWNTs) were formed by cross-linking with

divinyl sulfone, producing considerable change in the

morphology of the lyophilized hybrid hydrogels compared

to HA hydrogels itself. As a final result, HA plays a dual

role of matrix and linker for the rigid reinforcing

nanofibers [145]. A similar approach was used by Mendes

et al to produce hydrogels able to be used as potential

biomaterials for the restoration of bone defects [146].

Finally, a current cutting-edge topic is the development of

biocompatible delivery systems to be used as a carrier for

genetic material. Many attempts have been made to

produce the ―perfect‖ carrier, able to overcome all the

issues related to the crossing of the biological barriers

(enzyme degradation, endocytosis, intracellular

trafficking), once administered in the body. Mok et al

produced a Green covalently linked fluorescent protein

(GFP) antisense oligodeoxynucleotide (ODN) / (HA)

conjugate via a reducible disulfide linkage, and the HA-

ODN was complexed with protamine to increase the extent

of cellular uptake and enhance the gene inhibition

efficiency of GFP expression [147,148]. Furthermore, HA

was derivatized exploiting the conjugation with

polyethyleneimine (PEI) chains through the carboxylic

groups to obtain a favorable positively charged

environment for siRNA complexation [149-151]. This

strategy could have potential applications as safe and

effective nonviral carriers for ODN and siRNA nucleic

acid therapeutics

Global markets and manufacturers

The use of hyaluronic acid (HA) has been extensively

investigated in blends with other biopolymer and growth

factors to form matrices to mimic the extracellular matrix

surrounding the living cells. In this section commercially

valuable application of hyaluronic acid as biomaterial will

be described.

The HA market is led by several manufacturers who

compete in different market segments related to their

specific application. Seikagaku Corporation,

manufacturing Artz HA viscosupplementant, is the largest

562 Natural Product Communications Vol. 6 (4) 2011 Murano et al.

company in the global HA market with its dominance in

the Japanese market and its presence in Europe and US.

Among the competitors, Seikagaku is followed by

Genzyme Biosurgery. Q-Med dominates in the field of

dermal filler while the largest competitor in the ophtalmic

viscoelastics market is Advanced Medical Optics (AMO).

Table 1 summarizes the composition of the main

competitors in the field of HA-based biomaterials:

Table 1: Leading competitors in the HA-based treatment market (by

segment Global, 2005) [152].

Company Market share

Genzyme

Biosurgery 16.3%

Q-Med 12.3%

Fidia Farmaceutici 9.0%

AMO 7.9%

Alcon 7.3%

Chugai

Pharmaceuticals 5.3%

Santen 1.9%

Bausch&Lomb 1.9%

Inamed 1.8%

Corneal 1.5%

Bioniche Life

Science 1.3%

Others 11.1%

An emerging company in the HA manufacturer‘s scenario

is Novozymes, which has developed a patented process for

production of HA form an bioengineered strain of Bacillus

Subtilis. Also Anika Therapeutics recently strengthened its

position market acquiring part of Fidia Advanced

Biopolymers, producing hyaluronic acid-based products in

several therapeutic areas including the regeneration of

connective and structural tissues damaged by injuries,

aging or degenerative diseases.

It is worth mentioning that in 2010 the global HA

treatment market is foreseen to generate over $2 billion in

revenues, demonstrating an overall compound annual

growth rate (CAGR) in excess of 8%. The main sectors of

extreme commercial interest are those related to:

- Dermal fillers

- Ophtalmic viscoelastics

- Viscosupplementation

In addition to these sectors other areas are being deeply

investigated for their usefulness in commercial

applications such as:

- Tissue engineering

- Adhesion barriers

- Wound healing/wound dressing

Dermal fillers: HA is one of the most used materials for

dermal filler products. In fact, according to recent

statistics, more than 85% of all dermal filler procedures

occurred with a hyaluronic acid derivative [153,154].

Dermal fillers for face rejuvenation were proposed on the

market many years ago and received a large interest with

the introduction of injectable bovine collagen. As the

procedures for wrinkle correction with dermal fillers

became more popular, the search began for safer, longer

lasting fillers [155-157]. The next generation of filler after

those made of bovine collagen was crosslinked hyaluronic

acid derivatives, which was an improvement over bovine

collagen in that it did not require a skin test for

hypersensitivity and appeared to be longer lasting. The

search for safer and more effective dermal fillers has

continued because of the patients requirements to have

fillers which predictably last longer than 6 months. In spite

of a number of new temporary fillers composed of

crosslinked hyaluronic acid that are available in Europe

and are undergoing clinical testing in the United States, it

is not fully understood what parameters control the

performance of these fillers [158]. Nevertheless, the

customer demand for such a products is very high and

demonstrated by the list below, summarizing the most used

crosslinked HA-based materials as dermal fillers (Table 2):

Several studies have been carried out to explain the

properties [159] and the mechanism of action of dermal

fillers. Wang et al. proposed that the injection of

crosslinked hyaluronic acid stimulates collagen synthesis,

partially restoring dermal matrix components damaged by

environmental conditions (air, UV radiation). They

hypothesize that this stimulatory effect may be induced by

mechanical stretching of the dermis, which could activate

the dermal fibroblasts. These findings suggest that cross-

linked hyaluronic acid may be useful for stimulating

collagen production therapeutically, particularly in

atrophic skin conditions [160]. Table 2: Producers, products and functions of the most used dermal

fillers [152].

To improve the efficacy of the injections and the whole

final esthetic result, Kenner proposed the simultaneous

administration of HA and botulinum neurotoxin to

strengthen and improve the rejuvenation effect of the

treatment, showing for the first time that hyaluronic acid

and botulinum neurotoxin can be delivered in combination

in the same syringe and at the same time for the treatment

of the upper face [161,162]. Hasson et al evaluated

Esthélis, a new formulation sold as injectable non-animal

crosslinked hyaluronic acid. It is characterized by a

polydense cohesive matrix (CPM®), which produces a gel

of uniform consistency with better biointegration to the

Company Market share Product

Q-Med 71,2% Restylene

Inamed 10,6%

Hylaform

Captique Juvederm

Corneal 8,5% Juvederm

Surgiderm

Merz Pharmaceuticals 1,9% Hyal-System

Boletero

Fidia 1,3% Hyal-System

L.E.A. Derm 1,1% Juwederm

Mentor 0,9% Puragen

Others 4,4% -

Review on hyaluronan Natural Product Communications Vol. 6 (4) 2011 563

tissues and a longer duration in the treatment of atrophic

scars [163]. Monheit et al. studied a new type of dermal

filler based on a Dermal gel extra (DGE) crosslinked HA-

based material containing lidocaine, engineered to resist

deformation and degradation. The results obtained on

patients indicate that DGE could provide a comfortable

and cost-effective dermal filler option for clinicians and

patients [164].

Table 3: Producers, products and relative market share of the mainly used

dermal fillers [152].

Brand Name Manufacturer Function

Hylaform Inamed Moderate to severe wrinkles

Hylaform Plus Inamed Moderate to severe wrinkles

Restylene Touch Q-Med AB Superficial lines

Restylene Q-Med AB Moderate to severe wrinkles

Restylene

Perlane Q-Med AB Deep dermis and lips

Restylene SubQ Q-Med AB Add volume

Restylene Lipp Q-Med AB Lips

Juvederm 24 LEA Derm Medium to deep wrinkles

Juvederm 24HV LEA Derm Mid dermis and lips

Juvederm 30 LEA Derm Mid dermis and lips

Juvederm 30HV LEA Derm Medium to deep wrinkles

and lips

Esthelis Anteis Mid or deep dermis and lips

Puragen Mentor Mid dermis and lips

Yeom et al. proposed a new generation of dermal fillers

based on HA for tissue augmentation. Instead of using

highly reactive crosslinkers such as divinyl sulfone (DVS)

for Hylaform, 1,4-butanediol diglycidyl ether (BDDE) for

Restylane, and 1,2,7,8-diepoxyoctane (DEO) for Puragen,

HA hydrogels were prepared by direct amide bond

formation between the carboxyl groups of HA and

hexamethylenediamine (HMDA) with an optimized

carboxyl group modification for effective tissue

augmentation [140]. The research mentioned above

indicates products which are under the spotlight of the

cosmetic and pharmaceutic industries; the market segment

of this type of products is mainly subdivided among the

companies listed in Table 3:

Ophthalmic viscoelastics: Hyaluronic acid is widely use

in ophthalmology due to its viscosupplementant properties

and its presence in the cornea, the sclera and the vitreous

humor [165]. Ophthalmic viscoelastics are HA-based

products used to maintain the structure of the eye‘s

capsule, mainly in the phacoemulsification (removal of

the eye‘s vitreous humor) step of cataract surgery [166],

but also in other surgical procedures such as vitreoretinal

surgery, anterior segment surgery, glaucoma surgery, and

corneal transplantation [167]. HA-based ophthalmic

viscoelastics are used in every cataract procedure and

intraocular lens implantation. Maintaining the eye‘s

structural integrity without the fluid vitreous humor,

viscoelastics also provide lubrification and protection to

sensitive corneal tissue. Some studies have been carried

out to produce a biocompatible materials for the vitreous

humor replacement after vitrectomy using a new

crosslinked HA through adipic acid dihydrazide bridges

[143] and for cell sheet delivery applications [168]. By

bonding to the fragments of the cloudy lens, viscoelastics

act to facilitate natural lens removal toward the end of the

phacoemulsification process. In this sector the main

competitors are listed in Table 4:

Table 4: Producers, products and relative market share of the manly used

ophthalmic viscoelastics [152].

Company Market share Product

AMO 39,1% Healon

Alcon 36,4% DisCoVisc

Santen 9,6% Opegan

Bausch&Lomb 9,3% AmVisc Plus

Staar Surgical 2,6% StaarVisc

Others 3,0% -

Some efforts have been made to prepare crystalline lens

made of biodegradable hyaluronic acid and a non-

degradable polymeric gel, showing good biocompatibility

in the body [169].

The use of these materials is also extended to the eye‘s

cosmetic surgery. In fact, soft-tissue fillers have been

successfully used for volume augmentation in the orbit, the

tear trough, the upper eyelid/sub-brow region and in the

brow and temple region. Non-animal stabilized hyaluronic

acid fillers are the most commonly used in orbital and

periorbital volume augmentation. As a matter of the fact,

the role of injectable soft-tissue fillers in treating

periorbital pathologies is gaining much interest, due to the

results obtained in the treatment of paralytic

lagophthalmos, lower eyelid retraction and cicatricial

ectropion [170].

Viscosupplementation: Intra-articular viscosupplementa-

tion was approved by the Food and Drug Administration

(FDA) in 1997. HA viscosupplementation is a treatment

for osteoarthritis (OA) sufferers where injectable, artificial

produced HA supplements the body‘s own HA. The

product acts to reduce joint pain while increasing joint

mobility [171,172]. The positive effects of HA are due in

part to the fact that it influences physical properties, such

as viscoelasticity and lubrification, and HA molecules

interacts with the tissues in the joints. HA products are

delivered as a series of injections and provide varying

lengths of pain relief. It should be recognized that HA

viscosupplementation doesn‘t cure osteoarthritis but rather

serves as an intermediate treatment for patient who have

not been successfully treated with pharmaceuticals [173].

The pain relief activity of this treatments is of course, not

limited to OA itself but also to disease-related pain

persistent after arthroscopic surgery [174], such as

meniscectomy or cartilage damage in joints.

Strauss et al. highlighted as intra-articular hyaluronic acid

viscosupplementation was gaining popularity as a

treatment option in the nonoperative management of

patients with osteoarthritis. Recent clinical studies have

564 Natural Product Communications Vol. 6 (4) 2011 Murano et al.

demonstrated that the anti-inflammatory, anabolic, and

chondroprotective actions of hyaluronic acid reduce pain

and improve patient function. With evidence mounting in

support of the efficacy of this treatment modality for

patients with osteoarthritis, its potential use in patient

populations affected by pathologies of weight-bearing joint

such as knee [175] and ankles [176] is deeply investigated.

Bencke et at. showed that the number of patients with

osteoarthritis (OA) is expected to be growing.

Viscosupplementation, in which hyaluronic acid is injected

into the knee joint, has evolved into an important part of

our current therapeutic regimen in addressing the patient

with knee pain due to OA. Although suffering from lack of

an "evidence-based" approach there is a growing body of

data demonstrating the efficacy of HA in decreasing pain

and improving function in patients with knee OA. The

extensive use in clinic of HA has led to the introduction on

the market of various forms of HA, although little data are

available to justify one over the other [177]. Further proofs

about the successful concept of HA as viscosupplementant

were given by Mathieu et al during in vitro studies on

rheological behavior of synovial fluids mixed with linear

and crosslinked HA. They showed as in vitro tests

highlighted the stability of HA over a period of 6 week

without relevant alteration of the rheologic behavior and

no pronounced degradation by catabolytic enzymes [178].

The pharmaceutical companies are very active in this field

since these are high-demand products. The leading

companies in the viscosupplementation field and the

relative HA-based products are summarized in Table 5.

Table 5: Manufacturers, products and share market of the most used

viscosupplementants [152].

Company Market share Product

Seikagaku corporation 35,80% ARTZ

Genzyme Biosurgery 26,10% Synvisc (Hylan G-F20)

Fidia Pharmaceuticals 14,30% Hyalgan

Chugai Pharmaceuticals 8,40% Suvenyl

Bioniche Life Science 2,00% Suplasyn

Others 13,40% -

Tissue engineering: The production of artificial 3D

matrices is nowadays a challenging topic [179],

considering the in vitro tissue growth for surgical

application a strong market need. Several approaches have

been proposed to promote the cell growth on 3D-matrices

made of biocompatible components. Due to a strong

market demand for those kind of artificial tissues research

in regenerative medicine is rapidly expanding to cope with

this new need. Based on the concept of tissue regeneration

triggered by HA-based materials, some products are

commercially available for the treatment of deep dermal

lesions such as non healing ulcers, deep II degree and III

degree burns [180] (HyalograftTM

3D), for autologous skin

replacement (Laserskin®), for autologous fibroblasts and

keratinocytes replacement (Tissuetech) and for wound

healing/dressing, whose use will be discussed later on in

this chapter.

In general, there is nowadays a trend towards supplying

cells with a material to speed up the tissue healing process.

Hydrogel encapsulation made of different biopolymers

provides cells with a three dimensional environment

similar to that experienced in vivo and therefore may allow

the maintenance of normal cellular function in order to

produce tissues similar to those found in the body [181].

Fan et al. synthesized galactosylated hyaluronic acid

(GHA) prepared through the covalent coupling of

lactobionic acid with HA. The derivative was then used to

make highly porous three-dimensional sponges composed

by a blend of chitosan and GHA. The mixture formed a 3D

matrix stabilized by the electrostatic interaction between

the two biopolymers. The matrix was applied to

hepatocytes for liver-specific functions analysis. The

addition of GHA not only improved the wettability and

changed their mechanical properties, but also significantly

influenced the cell attachment ratio. Moreover, liver

functions of the hepatocytes entrapped in the CS/GHA

scaffolds, such as albumin secretion, urea synthesis and

ammonia elimination were improved in comparison with

those in similar scaffolds made of chitosan [182].

Kasahara et al. have developed an original three-

dimensional (3D) scaffold to meet improved

biomechanical and biological requirements. The constructs

were made of chitosan-based hyaluronic acid hybrid

polymer fibers. Previous studies have shown that these

hybrid polymer fibers have superior adhesion of

chondrocytes and the ability to maintain chondrocyte

phenotype. The study aimed at regenerating hyaline-like

cartilage with sufficient mechanical properties combining

a volume-reduced 3D scaffold and a bioreactor system.

The implantation of such a biocomposites was proposed as

a model approach for various cartilaginous lesions [183].

Other studies, such that of Lee et al. focused on the

fabrication of 3D highly porous scaffolds and their

biochemical binding affinity to realize various biomimetic

functionalizations. This was achieved by using a binding

mechanism that incorporated molecules, which strongly

bind to each other. A good example of such a binding

molecule pair was avidin and biotin. In this work, a porous

hybrid scaffolds composed of natural materials such as

collagen and HA was prepared to evaluate morphology,

mechanical strength, in vitro enzymatic degradation and

cytotoxicity of the scaffolds as well as cell growth within

the scaffolds [184]. Other attempts were proposed by Chou

et al., who prepared a glue made of autologous fibrin as a

potential scaffold with very good biocompatibility for

neocartilage formation. However, fibrin glue has been

reported not to provide enough mechanical strength, but

with many growth factors to interfere the tissue growth.

Gelatin/hyaluronic acid/chondroitin-6-sulfate (GHC6S)

tricopolymer sponge was prepared as scaffold for cartilage

tissue engineering and showed very good results in cell

seeding and cell distribution [185]. The results indicated

that the chondrocytes cultured in GHC6S-fibrin glue

would effectively promote extracellular matrix secretion,

inhibiting the degradation. The evidence could support that

Review on hyaluronan Natural Product Communications Vol. 6 (4) 2011 565

GHC6S-fibrin glue would be a promising scaffold for

articular cartilage tissue engineering [186].

Hu and al reported the formation of biocompatible

hydrogels using physically cross-linked biopolymers.

Gellation of silk fibroin (from B. mori silkworm) aqueous

solution was effected by ultrasonication and used to entrap

blended, uncrosslinked, hyaluronic acid (HA) without

chemical cross-linking. This is a novel approach to HA

hydrogel systems, which otherwise require chemical cross-

linking. Further, these systems exploit the beneficial

material and biological properties of both polymers. These

novel non-chemically cross-linked blend hydrogels may be

useful for biomedical applications due to biocompatibility

and the widespread utility of hydrogel systems [187].

Adhesion barriers: Adhesions are a major cause of post-

surgical morbidity and mortality and entail a substantial

medical economic burden. It has been estimated that 90%

of patients undergoing major abdominal surgery and 55%

to 100% of women undergoing pelvic surgery develop

adhesions. These phenomena are tightly linked to two risk

factors: tissue injury and inflammation response. Clinically

significant outcomes associated with adhesions include

chronic pelvic and abdominal pain, bowel obstruction, and

infertility. Consequently, methods for preventing such

adhesions have been the focus of extensive research. Few

barrier devices based on polysaccharides such as

hyaluronic acid or oxidized regenerated cellulose are in

commercial use, since part of them are based on synthetic

polymers (polytetrafluoroethylene) [188,189]. One of the

most used adhesion barrier is Seprafilm®, a composite

made of HA and carboxymethylcellulose employed in a

wide range of applications such as in bowel obstruction in

gynecological malignancies [190], tendons repair [191]

and after myomectomy, which is a procedure to surgically

removed uterine fibroids from the uterus. This procedure

often causes adhesion formation and decreases subsequent

fertility. This was the reason to evaluate the effectiveness

of several antiadhesion barrier materials in preventing

adhesion after myomectomy. Tsuji et al. prospectively

classified 63 women undergoing myomectomy alone into

four groups according to the type of antiadhesion material

used: Hyaluronic acid-carboxymethylcellulose film

(Seprafilm®), Dextran 40 (10% Dextran 40 Low

Injection®), factor 13 with fibrinogen (Beriplast

®) and

control. Seprafilm® was highly effective and was superior

to the other antiadhesion materials tested in preventing

uterine adhesions after myomectomy [192]. Other HA-

based product available on the market is INCERT®-S

(Anika), an anti-adhesion barrier gel for use in spinal

surgeries such as discectomies with a laminectomy or

laminotomy.

Other examples of dangerous adhesions are peritoneal

adhesions. These are among the most frequent cases of

tissue connections that form within the abdominopelvic

cavity following surgery or other injuries. They can cause

major medical complications. For this reason, researchers

are combining barrier devices and pharmacological agents

to be used in the prevention of this type of adhesion

formation. It was hypothesized that an adhesion barrier,

which also delivers anti-adhesion drugs, can address both

physical and physiological causes for adhesion formation.

For this reason an in situ cross-linking hyaluronan

hydrogel (barrier device) was prepared containing the

glucocorticoid receptor agonist budesonide [193]. A

similar approach to prevent peritoneal adhesions was the

application of physical barriers, which can separate the

injured regions during peritoneal healing. The hybrid

cross-linked HA-nanoparticles system appears to be a

biocompatible and highly effective adhesion barrier. This

―hybrid system‖ is able to prevent postoperative peritoneal

adhesions and combine the biocompatibility and ease of

application of in situ cross-linkable hydrogels with the

controlled release features of polymeric nanoparticles

[194]. Furthermore, postoperative peritoneal adhesions can

have serious, potentially lethal consequences.

Pharmacotherapy and barrier devices can reduce adhesion

formation to varying degrees, but their efficacy is limited

by rapid clearance from the peritoneum and lack of

biological activity, respectively. To overcome these

limitations, Yeo et al. have delivered tissue-type

plasminogen activator (tPA), which is deficient in the first

2-3 postoperative days, using a highly cross-linked in situ

forming hyaluronan gel. They demonstrated this

formulation's anti-adhesion activity in animal model that

involved recurrent adhesions [193]. Bioresorbable

membranes composed of hyaluronic acid and

carboxymethylcellulose (HA/CMC) are the most effective

method to prevent intra-abdominal adhesions; however,

their efficacy may be limited to the site of application.

Previous studies have shown that the intraperitoneal

administration of a neurokinin-1 receptor antagonist (NK-

1RA) reduces adhesions; however, the co-administration

of HA/CMC plus an NK-1RA has not been studied. The

co-administration of HA/CMC plus NK-1RA not only

increases the efficacy of the membrane at the site of

application, but also significantly reduces adhesions

formation at distal unprotected sites. This combination

may represent an emerging concept in more effective

adhesion prevention throughout the peritoneum [195].

Zhao et al. prepared a sodium carboxymethylcellulose-

Hyaluronic acid-Carboxymethylchitosan (CMCS) blend

films. The results suggested that the material was held

together by strong molecular interaction and hinted good

compatibility in the blend films. The blend films are

expected to be used as controllable degradation

biomaterials in the field of postsurgical hemostasis and

anti-adhesion [196].

Wound healing and dressing: Successful repair of

wounds and tissues remains a major healthcare and

biomedical challenge in the 21st Century. In particular,

chronic wounds often lead to loss of functional ability,

increased pain and decreased quality of life. Advanced

healing therapies employing biological dressings, skin

substitutes, growth factor-based therapies and synthetic

acellular matrices, all of which aim to correct irregular and

566 Natural Product Communications Vol. 6 (4) 2011 Murano et al.

dysfunctional cellular pathways present in chronic wounds,

are becoming more popular [197]. Commercially available

HA-based products (for healing and dressing) on the

market are Hyalofill-f, Hyalofill-R, Connettivina,

Connettivina Plus, Jaloskin®, Hyalomatrix®. Hyalofill

family products are absorbent, soft and conformable

fibrous fleece (F) or rope (R) composed of HYAFF®, an

ester of hyaluronic acid. Jaloskin® is a transparent film

dressing for the treatment of superficial moderately

exuding wounds. Hyalomatrix® is a bilayered, sterile,

flexible, and conformable wound dressing that acts as an

advanced wound care device.

HA uses in wound healing are strongly justified by its non-

antigenic properties and the number of forms it can be

manufactured, such as gels, creams to sheets of solid

material through lightly woven meshes. Epidermal

engraftment is superior to most of the available

biotechnologies and, as such, the material shows great

promise in both animal and clinical studies of tissue

engineering. Ongoing research works focus on the ability

of the biopolymer to enhance angiogenesis and the

conversion of chronic wounds into acute wounds [198].

Moreover, Gao et al investigated the biological roles of

hyaluronan (HA) fragments in angiogenesis acceleration.

Studies have confirmed that oligosaccharides of HA (o-

HA) are capable of stimulating neovascularization in vitro

and promoting blood flow or angiogenesis in animal

models. However, few laboratories have studied the

function of o-HA as an exogenous treatment in injured

tissue repair in vivo. It is thought that o-HA may lose its

activities when used topically in vivo due to its small size,

which may be absorbed quickly by the surrounding tissues.

A special slow-releasing gel containing a mixture of o-HA

at defined size was prepared and the healing effects, by

topical application to an acute wound model, was studied.

The therapy by o-HA was compared with high molecular

weight HA and the known angiogenesis stimulator, VEGF,

suggesting that o-HA therapy might be useful in acute

wound repair [200]. Ferguson et al worked on HA

fragments together to bioresponsive polymers to enhance

the circulation time and stability of biologically active

proteins and peptides, while reducing their

immunogenicity and toxicity. Recently, crosslinked

material made of HA at different molecular weights

blended with arginine and epidermal growth factor (EGF)

[201] and dextrin-EGF [202] conjugates were proposed,

the latter exploiting the so called Polymer-masked

UnMasked Protein Therapy (PUMPT) concept. The

hypothesis is that conjugation of a biodegradable polymer

to a protein would protect it and mask activity in transit,

while enabling controlled reinstatement of activity at the

target site by triggered degradation of the polymeric

component. It has been developed and shown as potential

modulators of impaired wound healing. In this contest, HA

fragments (Mw 90,000 g/mol) were conjugated to trypsin

as a model enzyme. HA-trypsin conjugates exhibited 52%

greater stability in the presence of elastase, compared to

free trypsin, demonstrating the potential use of HA

conjugates as modulators of tissue repair. Ibrahim et al.

showed that exogenous hyaluronic acid oligomers (o-HA)

stimulate functional endothelialization, though native long-

chain HA was more bioinert and possibly more

biocompatible. Additionally, the author created hydrogels

containing high molecular weight HA (1 × 106 Da) and o-

HA mixtures (HA-o: 0.75-10 kDa) crosslinked with

divinylsulfone (DVS). The greatest endothelial cell (EC)

attachment and proliferation was observed on gels with an

high amount of o-HA. The study showed that the

beneficial EC response to o-HA and biocompatibility of

HA is mostly unaltered by their chemical derivatization

and crosslinking into an hydrogel [203].

References

[1] Meyer K, Palmer JW. (1934) The polysaccharide of the vitreous humor. Journal of Biological Chemistry, 107, 629-634.

[2] Meyer K, Chaffee E. (1941) The mucopolysaccharides of skin. Journal of Biological Chemistry, 138, 491-499. [3] O'Regan M, Martini I, Crescenzi F, De Luca C, Lansing M. (1994) Molecular mechanisms and genetics of hyaluronan

biosynthesis. International Journal of Biological Macromolecules, 16, 283-286. [4] Fraser JR, Laurent TC, Laurent UB. (1997) Hyaluronan: its nature, distribution, functions and turnover. Journal of Internal

Medicine, 242, 27-33.

[5] Nusgens BV. (2010) [Hyaluronic acid and extracellular matrix: a primitive molecule?]. Annales de Dermatologie et de Venereologie, 137 Suppl 1, S3-8.

[6] Laurent TC, Fraser JR. (1992) Hyaluronan. The Journal of the Federation of American Societies for Experimental Biology, 6, 2397-2404.

[7] Poole AR, Kojima T, Yasuda T, Mwale F, Kobayashi M, Laverty S. (2001) Composition and structure of articular cartilage: a

template for tissue repair. Clinical Orthopaedics & Related Research, S26-33. [8] Kogan G, Soltes L, Stern R, Gemeiner P. (2007) Hyaluronic acid: a natural biopolymer with a broad range of biomedical and

industrial applications. Biotechnology Letters 29, 17-25. [9] Manuskiatti W, Maibach HI. (2007) Hyaluronic acid and skin: wound healing and aging. International Journal of Dermatology 35,

539-544 [10] Chen WYJ, Abatangelo G. (1990) Functions of hyaluronan in wound repair. Wound Repair and Rigeneration, 7. [11] Miyake K, Underhill CB, Lesley J, Kincade PW. (1990) Hyaluronate can function as a cell adhesion molecule and CD44

participates in hyaluronate recognition. The Journal of Experimental Medicine, 172, 69-75. [12] Inoue M, Katakami C. (1993) The effect of hyaluronic acid on corneal epithelial cell proliferation. Investigative Opthalmology &

Visual Science, 34, 2313-2315.

Review on hyaluronan Natural Product Communications Vol. 6 (4) 2011 567

[13] Foger N, Marhaba R, Zoller M. (2000) CD44 supports T cell proliferation and apoptosis by apposition of protein kinases. European Journal of Immunology, 30, 2888-2899.

[14] Itano N, Atsumi F, Sawai T, Yamada Y, Miyaishi O, Senga T, Hamaguchi M, Kimata K. (2002) Abnormal accumulation of hyaluronan matrix diminishes contact inhibition of cell growth and promotes cell migration. Proceedings of the National Academy

of Sciences of the United States of America, 99, 3609-3614. [15] Toole BP. (2002) Hyaluronan in morphogenesis. Seminars in Cell & Developmental Biology, 12, 79-87.

[16] Nochi H, Shinomiya T, Tamoto K. (2006) Characterization of hyaluronan-binding proteins on guinea pig polymorphonuclear leukocytes: possible involvement of complement receptor type 3 (CR3, CD11b/CD18) in the hyaluronan-leukocyte interaction. The Journal of Biochemistry, 139, 59-70.

[17] Zhu H, Mitsuhashi N, Klein A, Barsky LW, Weinberg K, Barr ML, Demetriou A, Wu GD. (2006) The role of the hyaluronan receptor CD44 in mesenchymal stem cell migration in the extracellular matrix. Stem Cells, 24, 928-935.

[18] Gao F, Koenitzer JR, Tobolewski JM, Jiang D, Liang J, Noble PW, Oury TD. (2008) Extracellular superoxide dismutase inhibits inflammation by preventing oxidative fragmentation of hyaluronan. The Journal of Biological Chemistry, 283, 6058-6066.

[19] Ghandi NS, Mancera RL. (2008) The structure of glycosaminoglycans and ntheir interactions with proteins. Chemical Biology &

Drug Design, 72, 455-482. [20] Schaefer L, Schaefer RM. (2010) Proteoglycans: from structural compounds to signaling molecules. Cell and Tissue Research, 339,

237-246. [21] Lipponen P, Aaltomaa S, Tammi R, Tammi M, Agren U, Kosma VM. (2001) High stromal hyaluronan level is associated with

poor differentiation and metastasis in prostate cancer. European Journal of Cancer, 37, 849-856. [22] Toole BP, Wight TN, Tammi MI. (2002) Hyaluronan-cell interactions in cancer and vascular disease. The Journal of Biological

Chemistry, 277, 4593-4596.

[23] Misra S, Ghatak S, Zoltan-Jones A, Toole BP. (2003) Regulation of multidrug resistance in cancer cells by hyaluronan. The Journal of Biological Chemistry, 278, 25285-25288.

[24] Hascall VC, Majors AK, De La Motte CA, Evanko SP, Wang A, Drazba JA, Strong SA, Wight TN. (2004) Intracellular hyaluronan: a new frontier for inflammation? The Journal of Biological Chemistry, 1673, 3-12.

[25] Toole BP. (2004) Hyaluronan: from extracellular glue to pericellular cue. Nature Reviews Cancer, 4, 528-539.

[26] Jameson JM, Cauvi G, Sharp LL, Witherden DA, Havran WL. (2005) Gammadelta T cell-induced hyaluronan production by epithelial cells regulates inflammation. The Journal of Experimental Medicine, 201, 1269-1279.

[27] Bharadwaj AG, Rector K, Simpson MA. (2007) Inducible hyaluronan production reveals differential effects on prostate tumor cell growth and tumor angiogenesis. The Journal of Biological Chemistry, 282, 20561-20572.

[28] Twarock S, Tammi MI, Savani RC, Fischer JW. (2010) Hyaluronan stabilizes focal adhesions, filopodia, and the proliferative phenotype in esophageal squamous carcinoma cells. The Journal of Biological Chemistry, 285, 23276-23284.

[29] Stern R, Asari AA, Sugahara KN. (2006) Hyaluronan fragments: an information-rich system. European Journal of Cell Biology,

85, 699-715. [30] Deed R, Rooney P, Kumar P, Norton JD, Smith J, Freemont AJ, Kumar S. (1997) Early-response gene signalling is induced by

angiogenic oligosaccharides of hyaluronan in endothelial cells. Inhibition by non-angiogenic, high-molecular-weight hyaluronan. International Journal of Cancer, 71, 251-256.

[31] Noble PW. (2002) Hyaluronan and its catabolic products in tissue injury and repair. Matrix Biology, 21, 25-29.

[32] Termeer C, Benedix F, Sleeman J, Fieber C, Voith U, Ahrens T, Miyake K, Freudenberg M, Galanos C, Simon JC. (2002) Oligosaccharides of Hyaluronan activate dendritic cells via toll-like receptor 4. The Journal of Experimental Medicine, 195, 99-111.

[33] Trabucchi E, Pallotta S, Morini M, Corsi F, Franceschini R, Casiraghi A, Pravettoni A, Foschi D, Minghetti P. (2002) Low molecular weight hyaluronic acid prevents oxygen free radical damage to granulation tissue during wound healing. International

Journal of Tissue Reactivity, 24, 65-71. [34] Mascarenhas MM, Day RM, Ochoa CD, Choi WI, Yu L, Ouyang B, Garg HG, Hales CA, Quinn DA. (2004) Low molecular

weight hyaluronan from stretched lung enhances interleukin-8 expression. American Journal of Respiratory Cell and Molecular

Biology, 30, 51-60. [35] Powell JD, Horton MR. (2005) Threat matrix: low-molecular-weight hyaluronan (HA) as a danger signal. Immunologic Research,

31, 207-218. [36] Wang CT, Lin YT, Chiang BL, Lin YH, Hou SM. (2006) High molecular weight hyaluronic acid down-regulates the gene

expression of osteoarthritis-associated cytokines and enzymes in fibroblast-like synoviocytes from patients with early osteoarthritis.

Osteoarthritis Cartilage, 14, 1237-1247. [37] Toole BP, Slomiany MG. (2008) Hyaluronan: a constitutive regulator of chemoresistance and malignancy in cancer cells. Seminars

in Cancer Biology, 18, 244-250. [38] Aruffo A, Stamenkovic I, Melnick M, Underhill CB, Seed B. (1990) CD44 is the principal cell surface receptor for hyaluronate.

Cell, 61, 1303-1313. [39] Turley EA, Noble PW, Bourguignon LY. (2002) Signaling properties of hyaluronan receptors. The Journal of Biological

Chemistry, 277, 4589-4592.

[40] Yamada T, Kawasaki T. (2005) Microbial synthesis of hyaluronan and chitin: New approaches. Journal of Bioscience and Bioengineering, 99, 521-528.

[41] Goueffic Y, Guilluy C, Guerin P, Patra P, Pacaud P, Loirand G. (2006) Hyaluronan induces vascular smooth muscle cell migration through RHAMM-mediated PI3K-dependent Rac activation. Cardiovascular Research, 72, 339-348.

[42] Kyosseva SV, Harris EN, Weigel PH. (2008) The hyaluronan receptor for endocytosis mediates hyaluronan-dependent signal

transduction via extracellular signal-regulated kinases. The Journal of Biological Chemistry, 283, 15047-15055. [43] Maxwell CA, McCarthy J, Turley E. (2008) Cell-surface and mitotic-spindle RHAMM: moonlighting or dual oncogenic functions?

Journal of Cell Science, 121, 925-932.

568 Natural Product Communications Vol. 6 (4) 2011 Murano et al.

[44] DeAngelis PL, Papaconstantinou J, Weigel PH. (1993) Isolation of a Streptococcus pyogenes gene locus that directs hyaluronan biosynthesis in acapsular mutants and in heterologous bacteria. Journal of Biological Chemistry, 268, 14568-14571.

[45] DeAngelis PL. (1999) Hyaluronan synthases: fascinating glycosyltransferases from vertebrates, bacterial pathogens, and algal viruses. Cellular and Molecular Life Sciences, 56, 670-682.

[46] Weigel PH, DeAngelis PL. (2007) Hyaluronan synthases: a decade-plus of novel glycosyltransferases. The Journal of Biological Chemistry, 282, 36777-36781.

[47] Kooy FK. (2010) Enzymatic production of hyaluronan oligo- and polysaccharides, University of Wageningen, The Netherlands. PhD Thesis.

[48] Kendall FE, Heidelberger M, Dawson MH. (1937) A serologically inactive polysaccharide elaborated by mucoid strains of Group

A hemolytic Streptococcus. The Journal of Biological Chemistry, 118, 61-69. [49] Carter GR, Annau E. (1953) Isolation of capsular polysaccharides from colonial variants of Pasteurella multocida. American

Journal of Veterinary Research, 14, 475-478. [50] Maclennan AP. (1956) The production of capsules, hyaluronic acid and hyaluronidase by 25 strains of group C streptococci.

Journal of General Microbiology, 15, 485-491.

[51] Boyce JD, Chung JY, Adler B. (2000) Pasteurella multocida capsule: composition, function and genetics. J Biotechnol 83, 153-160 [52] Chung JY, Wilkie I, Boyce JD, Townsend KM, Frost AJ, Ghoddusi M, Adler B. (2001) Role of capsule in the pathogenesis of fowl

cholera caused by Pasteurella multocida serogroup A. Infection and Immunity, 69, 2487-2492. [53] Harrington DJ, Sutcliffe IC, Chanter N. (2002) The molecular basis of Streptococcus equi infection and disease. Microbes and

Infection, 4, 501-510. [54] Ouskova G, Spellerberg B, Prehm P. (2004) Hyaluronan release from Streptococcus pyogenes: export by an ABC transporter.

Glycobiology, 14, 931-938.

[55] Fong Chong B, Blank LM, McLaughlin R, Nielsen LK. (2005) Microbial hyaluronic acid production. Applied Microbiology and Biotechnology, 66, 341-351.

[56] Gao HJ, Du GC, Chen J. (2006) Analysis of metabolic fluxes for hyaluronic acid (HA) production by Streptococcus zooepidemicus. World Journal of Microbiology and Biotechnology, 22, 399-408.

[57] Abbot EL, Smith WD, Siou GP, Chiriboga C, Smith RJ, Wilson JA, Hirst BH, Kehoe MA. (2007) Pili mediate specific adhesion of

Streptococcus pyogenes to human tonsil and skin. Cellular Microbiology, 9, 1822-1833. [58] Voyich JM, Musser JM, DeLeo FR. (2007) Streptococcus pyogenes and human neutrophils: a paradigm for evasion of innate host

defense by bacterial pathogens. Microbes and Infection, 6, 1117-1123. [59] Wessels MR, Moses AE, Goldberg JB, DiCesare TJ. (1991) Hyaluronic acid capsule is a virulence factor for mucoid group A

streptococci. Proceedings of the National Academy of Sciences of the United States of America, 88, 8317-8321. [60] Schmidt KH, Gunther E, Courtney HS. (1996) Expression of both M protein and hyaluronic acid capsule by group A streptococcal

strains results in a high virulence for chicken embryos. Medical Microbiology and Immunology 184, 169-173.

[61] Jing W, DeAngelis PL. (2003) Analysis of the two active sites of the hyaluronan synthase and the chondroitin synthase of Pasteurella multocida. Glycobiology, 13, 661-671.

[62] Cleary PP, Larkin A. (1979) Hyaluronic acid capsule: strategy for oxygen resistance in group A streptococci. Journal of Bacteriology, 140, 1090-1097.

[63] Cywes C, Wessels MR. (2001) Group A Streptococcus tissue invasion by CD44-mediated cell signalling. Nature 414, 648-652

[64] Wayan Teguh Wibawan I, Pasaribu FH, Utama IH, Abdulmawjood A, Lammler C. (2002) The role of hyaluronic acid capsular material of Streptococcus equi subsp. zooepidemicus in mediating adherence to HeLa cells and in resisting phagocytosis. Research

in Veterinary Science, 67, 131-135. [65] Kim SJ, Park SY, Kim CW. (2006) A novel approach to the production of hyaluronic acid by Streptococcus equi subsp.

zooepidemicus. Journal of Microbiology and Biotechnology, 16, 1849-1855. [66] Chen WY, Marcellin E, Hung J, Nielsen LK. (2009) Hyaluronan molecular weight is controlled by UDP-N-acetylglucosamine

concentration in Streptococcus zooepidemicus. The Journal of Biological Chemistry, 284, 18007-18014.

[67] Levin JC, Wessels MR. (1998) Identification of csrR/csrS, a genetic locus that regulates hyaluronic acid capsule synthesis in group A Streptococcus. Molecular Microbiology, 30, 209-219.

[68] Schrager HM, Alberti S, Cywes C, Dougherty GJ, Wessels MR. (1998) Hyaluronic acid capsule modulates M protein-mediated adherence and acts as a ligand for attachment of group A Streptococcus to CD44 on human keratinocytes. Journal of Clinical Investigation, 101, 1708-1716.

[69] Pruimboom IM, Rimler RB, Ackermann MR, Brogden KA. (1996) Capsular hyaluronic acid-mediated adhesion of Pasteurella multocida to turkey air sac macrophages. Avian Diseases, 40, 887-893.

[70] DeAngelis PL, Jing W, Drake RR, Achyuthan AM. (1998) Identification and molecular cloning of a unique hyaluronan synthase from Pasteurella multocida. The Journal of Biological Chemistry, 273, 8454-8458.

[71] DeAngelis PL. (1999) Molecular directionality of polysaccharide polymerization by the Pasteurella multocida hyaluronan synthase. The Journal of Biological Chemistry, 274, 26557-26562

[72] Boyce JD, Adler B. (2000) The capsule is a virulence determinant in the pathogenesis of Pasteurella multocida M1404 (B:2).

Infection and Immunity, 68, 3463-3468. [73] DeAngelis PL, Gunay NS, Toida T, Mao WJ, Linhardt RJ. (2002) Identification of the capsular polysaccharides of Type D and F

Pasteurella multocida as unmodified heparin and chondroitin, respectively. Charbohydrate Research, 337, 1547-1552. [74] Ewers C, Lubke-Becker A, Bethe A, Kiebling S, Filter M, Wieler LH. (2006) Virulence genotype of Pasteurella multocida strains

isolated from different hosts with various disease status. Veterinary Microbiology, 114, 304-317.

[75] Nozoe T, Kohnoe S, Ezaki T, Kabashima A, Maehara Y. (2004) Significance of immunohistochemical over-expression of CD44v6 as an indicator of malignant potential in esophageal squamous cell carcinoma. Journal of cancer research and Clinical Oncology,

130, 334-338.

Review on hyaluronan Natural Product Communications Vol. 6 (4) 2011 569

[76] Udabage L, Brownlee GR, Nilsson SK, Brown TJ. (2005) The over-expression of HAS2, Hyal-2 and CD44 is implicated in the invasiveness of breast cancer. Experimental Cell Research, 310, 205-217.

[77] Van Etten JL. (2003) Unusual life style of giant chlorella viruses. Annual Review of Genetics, 37, 153-195. [78] Fitzgerald LA, Graves MV, Li X, Feldblyum T, Nierman WC, Van Etten JL. (2007) Sequence and annotation of the 369-kb NY-

2A and the 345-kb AR158 viruses that infect Chlorella NC64A. Virology, 358, 472-484. [79] Rinaudo M. (2006) Chitin and chitosan; properties and applications. Progress in Polymer Science, 31, 603-632.

[80] DeAngelis PL, Jing W, Graves MV, Burbank DE, Van Etten JL. (1997) Hyaluronan synthase of chlorella virus PBCV-1. Science 278, 1800-1803

[81] Mohammed Ali AM, Kawasaki T, Yamada T. (2005) Genetic rearrangements on the Chlorovirus genome that switch between

hyaluronan synthesis and chitin synthesis. Virology, 342, 102-110. [82] Wilson WH, Van Etten JL, Allen MJ. (2009) The Phycodnaviridae: the story of how tiny giants rule the world. Current Topics in

Microbiology and Immunology, 328, 1-42. [83] Kim J-H, Yoo S-J, Oh D-K, Kweon Y-G, Park D-W, Lee C-H, Gil G-H. (1996) Selection of a Streptococcus equi mutant and

optimization of culture conditions for the production of high molecular weight hyaluronic acid. Enzyme and Microbial Technology,

19, 440-445. [84] Stangohl S. (2000) Methods and means for the production of hyaluronic acid, USA Patent 6090596

[85] Stangohl S. (2003) Methods and means for the production of hyaluronic acid, USA Patent 6537795 [86] Johns MR, Goh L-T, Oeggerli A. (1994) Effect of pH, agitation and aeration on hyaluronic acid production by Streptococcus

zooepidemicus. Biotechnology Letters, 16, 507-512. [87] Armstrong DC, Johns MR. (1997) Culture Conditions Affect the Molecular Weight Properties of Hyaluronic Acid Produced by

Streptococcus zooepidemicus. Applied and Environmental Microbiology, 63, 2759-2764.

[88] Fong Chong B, Nielsen LK. (2003) Aerobic cultivation of Streptococcus zooepidemicus and the role of NADH oxidase. Biochemical Engineering Journal, 16, 153-162.

[89] Fong Chong B, Nielsen LK. (2003) Amplifying the cellular reduction potential of Streptococcus zooepidemicus. Journal of Biotechnology, 100, 33-41.

[90] Beres SB, Sesso R, Pinto SW, Hoe NP, Porcella SF, Deleo FR, Musser JM. (2008) Genome sequence of a Lancefield group C

Streptococcus zooepidemicus strain causing epidemic nephritis: new information about an old disease. PLoS One, 3, e3026. [91] Holden MT, Heather Z, Paillot R, Steward KF, Webb K, Ainslie F, Jourdan T, Bason NC, Holroyd NE, Mungall K, Quail MA,

Sanders M, Simmonds M, Willey D, Brooks K, Aanensen DM, Spratt BG, Jolley KA, Maiden MC, Kehoe M, Chanter N, Bentley SD, Robinson C, Maskell DJ, Parkhill J, Waller AS. (2009) Genomic evidence for the evolution of Streptococcus equi: host

restriction, increased virulence, and genetic exchange with human pathogens. PLoS Pathogens, 5, e1000346. [92] Ferretti JJ, McShan WM, Ajdic D, Savic DJ, Savic G, Lyon K, Primeaux C, Sezate S, Suvorov AN, Kenton S, Lai HS, Lin SP,

Qian Y, Jia HG, Najar FZ, Ren Q, Zhu H, Song L, White J, Yuan X, Clifton SW, Roe BA, McLaughlin R. (2001) Complete

genome sequence of an M1 strain of Streptococcus pyogenes. Proceedings of the National Academy of Sciences USA, 98, 4658-4663.

[93] Beres SB, Sylva GL, Barbian KD, Lei B, Hoff JS, Mammarella ND, Liu MY, Smoot JC, Porcella SF, Parkins LD, Campbell DS, Smith TM, McCormick JK, Leung DY, Schlievert PM, Musser JM. (2002) Genome sequence of a serotype M3 strain of group A Streptococcus: phage-encoded toxins, the high-virulence phenotype, and clone emergence. Proceedings of the National Academy of

Sciences USA, 99, 10078-10083. [94] May BJ, Zhang Q, Li LL, Paustian ML, Whittam TS, Kapur V. (2001) Complete genomic sequence of Pasteurella multocida,

Pm70. Proceedings of the National Academy of Sciences USA, 98, 3460-3465. [95] DeAngelis PL, Papaconstantinou J, Weigel PH. (1993) Molecular cloning, identification, and sequence of the hyaluronan synthase

gene from group A Streptococcus pyogenes. Journal of Biological Chemistry, 268, 19181-19184. [96] Dougherty BA, van de Rijn I. (1993) Molecular characterization of hasB from an operon required for hyaluronic acid synthesis in

group A streptococci. Demonstration of UDP-glucose dehydrogenase activity. Journal of Biological Chemistry, 268, 7118-7124.

[97] Crater DL, Dougherty BA, van de Rijn I. (1995) Molecular characterization of hasC from an operon required for hyaluronic acid synthesis in group A streptococci. Demonstration of UDP-glucose pyrophosphorylase activity. Journal of Biological Chemistry,

270, 28676-28680. [98] Weigel PH. (2002) Functional characteristics and catalytic mechanisms of the bacterial hyaluronan synthases. IUBMB Life, 54, 201-211. [99] Yu H, Stephanopoulos G. (2008) Metabolic engineering of Escherichia coli for biosynthesis of hyaluronic acid. Metabolic

Engineering, 10, 24-32. [100] Mao Z, Shin HD, Chen R. (2009) A recombinant E. coli bioprocess for hyaluronan synthesis. Applied Microbiology and

Biotechnology, 84, 63-69. [101] Chien LJ, Lee CK. (2007) Hyaluronic acid production by recombinant Lactococcus lactis. Applied Microbiology and

Biotechnology, 77, 339-346. [102] Mao Z, Chen RR. (2007) Recombinant synthesis of hyaluronan by Agrobacterium sp. Biotechnology Progress, 23, 1038-1042 [103] Sloma A, Behr R, Widner W, Tang M, Sternberg D, Brown S. (2003) Methods for producing hyaluronan in a recombinant host

cell. Novozymes Biopolymer, USA Patent WO/2003/054163 [104] Widner B, Behr R, Von Dollen S, Tang M, Heu T, Sloma A, Sternberg D, Deangelis PL, Weigel PH, Brown S. (2005) Hyaluronic

acid production in Bacillus subtilis. Applied and Environmental Microbiology, 71, 3747-3752. [105] Brown SH, Pummill PE. (2008) Recombinant production of hyaluronic acid. Current Pharmaceutical Biotechnology, 9, 239-241 [106] Graves MV, Burbank DE, Roth R, Heuser J, DeAngelis PL, Van Etten JL. (1999) Hyaluronan synthesis in virus PBCV-1-infected

chlorella-like green algae. Virology 257, 15-23 [107] Yamada T. (2004) Method for separating/purifyng hyaluronic acid and/or chitin, JP Patent 2004-283096.

[108] Takeo S, Fujise M, Akiyama T, Habuchi H, Itano N, Matsuo T, Aigaki T, Kimata K, Nakato H. (2004) In vivo hyaluronan synthesis upon expression of the mammalian hyaluronan synthase gene in Drosophila. Journal of Biological Chemistry, 279, 18920-18925.

570 Natural Product Communications Vol. 6 (4) 2011 Murano et al.

[109] Capodilupo AL, Crescenzi V, Francescangeli A, Joudioux R, Leonelli F, Marini Bettolo R, Migneco LM, Quagliariello M, Bellato B, De Luca G, Galbiati E, Renier D, Banzato A, Rosato A. (2005) Hyaluronan Structure, Metabolism, Biological Activities,

Therapeutic Applications, Hyaluronan 2003 Endre A. Balazs and Vincent C. Hascall Editors Matrix Biology Institute (New Jersey) Vol. II, 391-395.

[110] Khan R, Bella J, Konowicz PA, Paoletti S, Vesnaver R, Linda P. (1998) Selective acetylation reactions of hyaluronic acid benzyl ester derivative Carbohydrate Research, 306, 137-146.

[111] Sorbi C, Bergamin M, Bos iS, Dinon F, Aroulmoji V, Khan R, Murano E, Norbedo S. (2009) Synthesis of 6-O-methotrexylhyaluronan as a drug delivery system. Carbohydrate Research, 344, 91-97.

[112] Khan R, Vesnaver R, Stucchi L, Bosco M. (2002) Carboxylated polysaccharides 6-substituted, USA Patent 6482941 B1

[113] Lapcík LJ, Lapcík L, De Smedt S, Demeester J, Chabrecek P. (1998) Hyaluronan: Preparation, Structure, Properties, and Applications. Chemical Reviews, 98, 2663-2684.

[114] Mrácek A, Benesová K, Minarík A, Urban P, Lapcík L. (2007) The diffusion process of sodium hyaluronate (Na-Ha) and Na-Ha-n-alkyl derivatives films swelling. Journal of Biomedical Materials Research Part A 83, 184-190.

[115] Vesnaver R. (1999) New Derivatives of Hyaluronic Acid: Synthesis and Characterisation, University of Trieste, Italy.

[116] Norbedo S, Dinon F, Bergamin M, Bosi S, Aroulmoji V, Khan R, Murano E. (2009) Synthesis of 6-amino-6-deoxyhyaluronan as an intermediate for conjugation with carboxylate-containing compounds: application to hyaluronan-camptothecin conjugates.

Carbohydrate Research, 344, 98-104. [117] Luo Y, Prestwich GD. (1999) Synthesis and Selective Cytotoxicity of a Hyaluronic Acid−Antitumor Bioconjugate. Bioconjugate

Chemistry, 10, 755-763. [118] Lee H, Lee K, Park TG. (2008) Hyaluronic acid-paclitaxel conjugate micelles: synthesis, characterization, and antitumor activity.

Bioconjugate Chemistry, 19, 1319-1325

[119] Akima K, Iwata Y, Matsuo K, Watar N. (1998) Compound for medicinal ingredient and hyaluronic acid and process for producing the same, USA Patent 5733891

[120] Luo Y, Prestwich GD. (2001) Hyaluronic acid-N-hydroxysuccinimide: a useful intermediate for bioconjugation. Bioconjugate Chemistry, 12, 1085-1088.

[121] Luo Y, Bernshaw NJ, Lu ZR, Kopecek J, Prestwich GD. (2002) Targeted delivery of doxorubicin by HPMA copolymer hyaluronan

bioconjugates. Pharmaceutical Research, 19, 396-402. [122] Leardini G, Mattara L, Franceschini M, Perbellini A. (1991) Intra-articular treatment of knee osteoarthritis. A comparative study

between hyaluronic acid and 6-methyl prednisolone acetate, Clinical and Experimental Rheumatology. Clinical and Experimental Rheumatology, 9, 375-381.

[123] Kyyrönen K, Hume L, Benedetti L, Urtti A, Topp E, Stella V. (1991) Dosage Forms From Polymeric Prodrugs: Hydrocortisone Esters of Hyaluronic Acid. International Journal of Pharmaceutics, 3, 41-48

[124] Khan R, Konowicz PA. (1996) Patent WO 96/35721

[125] Soltes L, Mendichi R, Kogan G, Schiller J, Stankovska M, Arnhold J. (2006) Degradative action of reactive oxygen species on hyaluronan. Biomacromolecules, 7, 659-668.

[126] Puig-Parellada P, Planas JM. (1978) Synovial fluid degradation induced by free radicals. In vitro action of several free radical scavengers and anti-inflammatory drugs. Biochemical Pharmacology, 27, 535-537.

[127] Parenti C, Costantino L, Di Bella M. (1990) Heteroarylalkanoic acids with possible antiinflammatory activities. Part 8: The

scavenging of the oxygen-free radicals. Pharmazie, 45, 680-681. [128] Mendoza G, Alvarez AI, Pulido MM, Molina AJ, Merino G, Real R, Fernandes P, Prieto JG. (2007) Inhibitory effects of different

antioxidants on hyaluronan depolymerization. Carbohydrate Research, 342, 96-102. [129] Kvam C, Granese D, Flaibani A, Pollesello P, Paoletti S. (1993) Hyaluronan can be protected from free-radical depolymerisation

by 2,6-diisopropylphenol, a novel radical scavenger. Biochemical and Biophysical Research Communications, 193, 927-933. [130] McKee CM, Penno MB, Cowman M, Burdick MD, Strieter RM, Bao C, Noble PW. (1996) Hyaluronan (HA) fragments induce

chemokine gene expression in alveolar macrophages. The role of HA size and CD44. The Journal of Clinical Investigation, 98,

2403-2413. [131] Berbis P. (2010) [Hyaluronan in inflammation, auto-immunity and cardio-vascular diseases]. Annales de Dermatologie et de

Venereologie, 137 Suppl 1: S40-43. [132] El-Safory NS, Fazary AE, Lee C-K. (2010) Hyaluronidases, a group of glycosidases: Current and future perspectives.

Carbohydrate Polymers, 81, 165-181.

[133] Ossipov DA, Piskounova S, Varghese OP, Hilborn J. (2010) Functionalization of hyaluronic acid with chemoselective groups via a disulfide-based protection strategy for in situ formation of mechanically stable hydrogels. Biomacromolecules, 11, 2247-2254.

[134] Li F, Bae B-c, Na K. (2010) Acetylated Hyaluronic Acid/Photosensitizer Conjugate for the Preparation of Nanogels with Controllable Phototoxicity: Synthesis, Characterization, Autophotoquenching Properties, and in vitro Phototoxicity against HeLa

Cells. Bioconjugate Chemistry, 21, 1312-1320. [135] Kim KS, Hur W, Park SJ, Hong SW, Choi JE, Goh EJ, Yoon SK, Hahn SK. (2010) Bioimaging for targeted delivery of hyaluronic

Acid derivatives to the livers in cirrhotic mice using quantum dots. ACS Nano, 4, 3005-3014.

[136] Kadi S, Cui D, Bayma E, Boudou T, Nicolas C, Glinel K, Picart C, Auzely-Velty R. (2009) Alkylamino hydrazide derivatives of hyaluronic acid: synthesis, characterization in semidilute aqueous solutions, and assembly into thin multilayer films.

Biomacromolecules, 10, 2875-2884. [137] Bencherif SA, Washburn NR, Matyjaszewski K. (2009) Synthesis by AGET ATRP of Degradable Nanogel Precursors for In Situ

Formation of Nanostructured Hyaluronic Acid Hydrogel. Biomacromolecules, 10, 2499-2507.

[138] Ruhela D, Riviere K, Szoka FC. (2006) Efficient Synthesis of an Aldehyde Functionalized Hyaluronic Acid and Its Application in the Preparation of Hyaluronan−Lipid Conjugates. Bioconjugate Chemistry, 17, 1360-1363.

[139] Ilgin P, Avci G, Silan C, Ekici S, Aktas N, Ayyala RS, John VT, Sahiner N. (2010) Colloidal drug carries from (sub)micron hyaluronic acid hydrogel particles with tunable properties for biomedical applications. Carbohydrate Polymers, 82, 997-1003.

Review on hyaluronan Natural Product Communications Vol. 6 (4) 2011 571

[140] Yeom J, Bhang SH, Kim BS, Seo MS, Hwang EJ, Cho IH, Park JK, Hahn SK. (2010) Effect of cross-linking reagents for hyaluronic acid hydrogel dermal fillers on tissue augmentation and regeneration. Bioconjugate Chemistry, 21, 240-247.

[141] Inukai M, Jin Y, Yomota C, Yonese M. (2000) Preparation and characterization of hyaluronate-hydroxyethyl acrylate blend hydrogel for controlled release device. Chemical & Pharmaceutical Bulletin, 48, 850-854.

[142] Leach JB, Bivens KA, Collins CN, Schmidt CE. (2004) Development of photocrosslinkable hyaluronic acid-polyethylene glycol-peptide composite hydrogels for soft tissue engineering. Journal of Biomedical Materials Research Part A 70, 74-82.

[143] Su WY, Chen KH, Chen YC, Lee YH, Tseng CL, Lin FH. (2010) An Injectable Oxidated Hyaluronic Acid/Adipic Acid Dihydrazide Hydrogel as a Vitreous Substitute. Journal of Biomaterials Science, Polymer Edition, in press.

[144] Oldinski RA, Cranson CN, James SP. (2010) Synthesis and characterization of a Hyaluronan-polyethylene copolymer for

biomedical applications. Journal of Biomedical Materials Research Part B: Applied Biomaterials, 94, 441-446. [145] Bhattacharyya S, Guillot S, Dabboue H, Tranchant JF, Salvetat JP. (2008) Carbon nanotubes as structural nanofibers for hyaluronic

acid hydrogel scaffolds. Biomacromolecules, 9, 505-509. [146] Mendes RM, Silva GA, Caliari MV, Silva EE, Ladeira LO, Ferreira AJ. (2010) Effects of single wall carbon nanotubes and its

functionalization with sodium hyaluronate on bone repair. Life Sciences, 87, 215-222.

[147] Mok H, Park JW, Park TG. (2007) Antisense oligodeoxynucleotide-conjugated hyaluronic acid/protamine nanocomplexes for intracellular gene inhibition. Bioconjugate Chemistry, 18, 1483-1489.

[148] Jeong JH, Mok H, Oh YK, Park TG. (2009) siRNA conjugate delivery systems. Bioconjugate Chemistry, 20, 5-14. [149] Jiang G, Park K, Kim J, Kim KS, Oh EJ, Kang H, Han SE, Oh YK, Park TG, Kwang Hahn S. (2008) Hyaluronic acid-

polyethyleneimine conjugate for target specific intracellular delivery of siRNA. Biopolymers, 89, 635-642. [150] Han SE, Kang H, Shim GY, Kim SJ, Choi HG, Kim J, Hahn SK, Oh YK. (2009) Cationic derivatives of biocompatible hyaluronic

acids for delivery of siRNA and antisense oligonucleotides. Journal of Drug Targeting, 17, 123-132.

[151] Jiang G, Park K, Kim J, Kim KS, Hahn SK. (2009) Target specific intracellular delivery of siRNA/PEI-HA complex by receptor mediated endocytosis. Molecular Pharmacology, 6, 727-737.

[152] Millenium Research Group. Global market for hyaluronic acid 2007. [153] Tezel A, Fredrickson GH. (2008) The science of hyaluronic acid dermal fillers. Journal of Cosmetic Laser Therapy, 10, 35-42. [154] Beasley KL, Weiss MA, Weiss RA. (2009) Hyaluronic acid fillers: a comprehensive review. Facial Plastic Surgery, 25, 86-94.

[155] Monheit GD, Coleman KM. (2006) Hyaluronic acid fillers. Dermatologic Therapy, 19, 141-150. [156] Gold MH. (2007) Use of hyaluronic acid fillers for the treatment of the aging face. Journal of Clinical Interventions in Aging, 2,

369-376. [157] Mercer SE, Kleinerman R, Goldenberg G, Emanuel PO. (2010) Histopathologic identification of dermal filler agents. Journal of

Drugs in Dermatology, 9, 1072-1078. [158] Falcone SJ, Berg RA. (2008) Crosslinked hyaluronic acid dermal fillers: a comparison of rheological properties. Journal of

Biomedical Materials Research, Part A 87, 264-271.

[159] Kablik J, Monheit GD, Yu L, Chang G, Gershkovich J. (2009) Comparative physical properties of hyaluronic acid dermal fillers. Dermatologic Surgery, 35 Suppl 1, 302-312.

[160] Wang F, Garza LA, Kang S, Varani J, Orringer JS, Fisher GJ, Voorhees JJ. (2007) In vivo stimulation of de novo collagen production caused by cross-linked hyaluronic acid dermal filler injections in photodamaged human skin. Archives of Dermatology, 143, 155-163.

[161] Benke M, Shaffer B. (2009) Viscosupplementation treatment of arthritis pain. Current Pain Headache Reports, 13, 440-446. [162] Kenner JR. (2010) Hyaluronic acid filler and botulinum Neurotoxin delivered simultaneously in the same syringe for effective and

convenient combination aesthetic rejuvenation therapy. Journal of Drugs in Dermatology, 9, 1135-1138. [163] Hasson A, Romero WA. (2010) Treatment of Facial Atrophic Scars With Esthelis, a Hyaluronic Acid Filler With Polydense

Cohesive Matrix (CPM). Journal of Drugs in Dermatology, 9, 1507-1509. [164] Monheit GD, Baumann LS, Gold MH, Goldberg DJ, Goldman MP, Narins RS, Bachtell N, Garcia E, Kablik J, Gershkovich J,

Burkholder D. (2010) Novel hyaluronic acid dermal filler: dermal gel extra physical properties and clinical outcomes.

Dermatologic Surgery, 36 Suppl 3, 1833-1841. [165] Figueiredo ES, Macedo AC, Figueiredo PF, Figueiredo RS. (2010) [Use of hyaluronic acid in Ophthalmology]. Arquivos

Brasileiros de Oftalmologia, 73, 92-95. [166] Van den Bruel A, Gailly J, Devriese S, Welton N, Shortt A, Vrijens F. (2009) The protective effect of ophthalmic viscoelastic

devices on endothelial cell loss during cataract surgery: a meta-analysis using mixed treatment comparisons. British Journal of

Ophthalmology DOI: 10.1136/bjo.2009.158360. [167] Balazs EA. (2008) Hyaluronan as an ophthalmic viscoelastic device. Current Pharmaceutical Biotechnology, 9, 236-238.

[168] Jui-Yang L, Hui-Kang MD, Hsiao-Yun C, Chi-Chin S, Shu-Jung H, Ya-Ting L, Ging-Ho L. (2010) Ocular Biocompatibility of Carbodiimide Cross-Linked Hyaluronic Acid Hydrogels for Cell Sheet Delivery Carriers. Journal of Biomaterials Science,

Polymer Edition, 21, 359-376. [169] Gwon A, Gruber L. (2010) Engineering the crystalline lens with a biodegradable or non-degradable scaffold. Experimental Eye

Research, 91, 220-228.

[170] Morley AMS, Malhotra R. (2010) Non-surgical Volume Enhancement with Fillers in the Orbit and Periorbital Tissues: Cosmetic and Functional Considerations. Essentials in Ophthalmology, Oculoplastics and Orbit, 221-229.

[171] Migliore A, Giovannangeli F, Bizzi E, Massafra U, Alimonti A, Lagana B, Diamanti Picchianti A, Germano V, Granata M, Piscitelli P. (2010) Viscosupplementation in the management of ankle osteoarthritis: a review. Archives of Orthopaedic and Trauma Surgery DOI: 10.1007/s00402-010-1165-5.

[172] Witteveen AG, Sierevelt IN, Blankevoort L, Kerkhoffs GM, van Dijk CN. (2010) Intra-articular sodium hyaluronate injections in the osteoarthritic ankle joint: effects, safety and dose dependency. Foot and Ankle Surgery, 16, 159-163.

[173] Abate M, Pulcini D, Di Iorio A, Schiavone C. (2010) Viscosupplementation with intra-articular hyaluronic acid for treatment of osteoarthritis in the elderly. Current pharmaceutical design, 16, 631-640.

572 Natural Product Communications Vol. 6 (4) 2011 Murano et al.

[174] Waddell DD, Bert JM. (2010) The use of hyaluronan after arthroscopic surgery of the knee. Arthroscopy, 26, 105-111. [175] Strauss EJ, Hart JA, Miller MD, Altman RD, Rosen JE. (2009) Hyaluronic acid viscosupplementation and osteoarthritis: current

uses and future directions. The American Journal of Sports Medicine, 37, 1636-1644. [176] Sun SF, Chou YJ, Hsu CW, Chen WL. (2009) Hyaluronic acid as a treatment for ankle osteoarthritis. Current Reviews in

Musculoskeletal Medicine, 2, 78-82. [177] Benke M, Shaffer B. (2009) Viscosupplementation treatment of arthritis pain. Current Pain and Headache Reports, 13, 440-446.

[178] Mathieu P, Conrozier T, Vignon E, Rozand Y, Rinaudo M. (2009) Rheologic behavior of osteoarthritic synovial fluid after addition of hyaluronic acid: a pilot study. Clinincal Orthopaedics and Related Research, 467, 3002-3009.

[179] Allison DD, Grande-Allen KJ. (2006) Review. Hyaluronan: a powerful tissue engineering tool. Tissue Engineering, 12, 2131-2140.

[180] Brusselaers N, Pirayesh A, Hoeksema H, Richters CD, Verbelen J, Beele H, Blot SI, Monstrey S. (2010) Skin replacement in burn wounds. Journal of Trauma, 68, 490-501.

[181] Hunt NC, Grover LM. (2010) Cell encapsulation using biopolymer gels for regenerative medicine. Biotechnology Letters, 32, 733-742.

[182] Fan J, Shang Y, Yuan Y, Yang J. (2010) Preparation and characterization of chitosan/galactosylated hyaluronic acid scaffolds for

primary hepatocytes culture. Journal of Material Science: Materials in Medicines, 21, 319-327. [183] Kasahara Y, Iwasaki N, Yamane S, Igarashi T, Majima T, Nonaka S, Harada K, Nishimura S, Minami A. (2008) Development of

mature cartilage constructs using novel three-dimensional porous scaffolds for enhanced repair of osteochondral defects. Journal of Biomedical Materials Research Part A, 86, 127-136.

[184] Lee SJ, Kim SY, Lee YM. (2007) Preparation of porous collagen/hyaluronic acid hybrid scaffolds for biomimetic functionalization through biochemical binding affinity. Journal of Biomedical Materials Research Part B: Applied Biomaterials, 82, 506-518.

[185] Enrione J, Osorio F, López D, Weinstein-Oppenheimer C, Fuentes MA, Ceriani R, Brown DI, Albornoz F, Elizabeth Sánchez E,

Patricio Villalobos P, Somoza RA, Young ME, Acevedo CA. (2010) Characterization of a Gelatin/Chitosan/Hyaluronan scaffold- polymer. Electronic Journal of Biotecnology, 13, 1-11.

[186] Chou CH, Cheng WT, Kuo TF, Sun JS, Lin FH, Tsai JC. (2007) Fibrin glue mixed with gelatin/hyaluronic acid/chondroitin-6-sulfate tri-copolymer for articular cartilage tissue engineering: the results of real-time polymerase chain reaction. Journal of Biomedical Materials Research Part A, 82, 757-767.

[187] Hu X, Lu Q, Sun L, Cebe P, Wang X, Zhang X, Kaplan DL. (2010) Biomaterials from Ultrasonication-Induced Silk Fibroin-Hyaluronic Acid Hydrogels. Biomacromolecules, in press.

[188] Diamond MP, Wexner SD, diZereg GS, Korell M, Zmora O, Van Goor H, Kamar M. (2010) Adhesion prevention and reduction: current status and future recommendations of a multinational interdisciplinary consensus conference. Surgical Innovation, 17, 183-188.

[189] Robertson D, Lefebvre G, Leyland N, Wolfman W, Allaire C, Awadalla A, Best C, Contestabile E, Dunn S, Heywood M, Leroux N, Potestio F, Rittenberg D, Senikas V, Soucy R, Singh S. (2010) Adhesion prevention in gynaecological surgery. Journal of Obstetrics and Gynaecology Canada, 32, 598-608.

[190] Tabata T, Kihira T, Shiozaki T, Tanida K, Kondo E, Nagao K, Okugawa T, Sagawa N. (2010) Efficacy of a sodium hyaluronate-carboxycellulose membrane (seprafilm) for reducing the risk of early postoperative small bowel obstruction in patients with

gynecologic malignancies. International Journal of Gynecological Cancer, 20, 188-193. [191] Yilmaz E, Avci M, Bulut M, Kelestimur H, Karakurt L, Ozercan I. (2010) The Effect of Seprafilm on Adhesion Formation and

Tendon Healing After Flexor Tendon Repair in Chicken. Orthopedics: 164-170. DOI:10.3928/01477447-20100129-16.

[192] Tsuji S, Takahashi K, Yomo H, Fujiwara M, Kita N, Takebayashi K, Miyazaki K, Noda Y. (2005) Effectiveness of antiadhesion barriers in preventing adhesion after myomectomy in patients with uterine leiomyoma. Europena Journal of Obstetrics &

Gynecology an Reproductive Biology, 123, 244-248. [193] Yeo Y, Adil M, Bellas E, Astashkina A, Chaudhary N, Kohane DS. (2007) Prevention of peritoneal adhesions with an in situ cross-

linkable hyaluronan hydrogel delivering budesonide. Journal of Controlled Release, 120, 178-185. [194] Yeo Y, Ito T, Bellas E, Highley CB, Marini R, Kohane DS. (2007) In situ cross-linkable hyaluronan hydrogels containing

polymeric nanoparticles for preventing postsurgical adhesions. Annals of Surgery, 245, 819-824.

[195] Lim R, Stucchi AF, Morrill JM, Reed KL, Lynch R, Becker JM. (2010) The efficacy of a hyaluronate-carboxymethylcellulose bioresorbable membrane that reduces postoperative adhesions is increased by the intra-operative co-administration of a neurokinin

1 receptor antagonist in a rat model. Surgery, 148, 991-999. [196] Zhao XH, He XW, Xie SQ, Yang LS. (2010) Preparation and Properties of Sodium Carboxymethyl Cellulose-Hyaluronic Acid-

Carboxymethyl Chitosan Blend. Applied Mechanics and Materials, 20-23, 1157-1161.

[197] Rizzi SC, Upton Z, Bott K, Dargaville TR. (2010) Recent advances in dermal wound healing: biomedical device approaches. Expert review in Medical Devices, 7, 143-154.

[198] Price RD, Myers S, Leigh IM, Navsaria HA. (2005) The role of hyaluronic acid in wound healing: assessment of clinical evidence. American Journal of Clinical Dermatology, 6, 393-402.

[199] Lin YK, Matsumoto Y, Kuroyanagi Y, Kagawa Y. (2009) A Bilayer Hyaluronic Acid Wound Dressing to Promote Wound Healing in Diabetic Ulcer. Journal of Bioactive and Compatible Polymers, 24, 424-443.

[200] Gao F, Liu Y, He Y, Yang C, Wang Y, Shi X, Wei G. (2010) Hyaluronan oligosaccharides promote excisional wound healing

through enhanced angiogenesis. Matrix Biology, 29, 107-116. [201] Yasuhiro M, Yoshimitsu K. (2010) Development of a Wound Dressing Composed of Hyaluronic Acid Sponge Containing Arginine

and Epidermal Growth Factor. Journal of Biomaterials Science, Polymer Edition, 21, 715-726. [202] Ferguson EL, Alshame AM, Thomas DW. (2010) Evaluation of hyaluronic acid-protein conjugates for polymer masked-unmasked

protein therapy. IInternational Journal of Pharmaceutics 402, 95-102.

[203] Samir I, Qian KK, Anand R. (2010) The impact of hyaluronic acid oligomer content on physical, mechanical, and biologic properties of divinyl sulfone-crosslinked hyaluronic acid hydrogels. Journal of Biomedical Materials Research Part A, 94A,

355-370.

Natural Product Communications

2011 Volume 6, Number 4

Contents

Original Paper Page

Myrtenal, a Controversial Molecule for the Proper Application of the CIP Sequence Rule for Multiple Bonds

L. Gerardo Zepeda, Eleuterio Burgueño-Tapia and Pedro Joseph-Nathan 429

A Facile and Efficient Four-Step Enantioselective Synthesis of (+)-Vernolepin from (+)-Minimolide, the

Major Germacranolide of Mikania minima

Sandra M. Bach, Fernando R. Díaz, Horacio Bach and César A. N. Catalán 433

Use of the Plant Bellardia trixago for the Enantiospecific Synthesis of Odorant Products

Alejandro F. Barrero, M. Mar Herrador, Pilar Arteaga, Alexis Castillo and Alejandro F. Arteaga 439

Botcinolide/Botcinin: Asymmetric Synthesis of the Key Fragments

Jacinto Ramírez-Fernández, José Manuel Botubol, Antonio J. Bustillo, Josefina Aleu, Isidro G. Collado and

Rosario Hernández-Galán 443

C-6 Regioselective Bromination of Methyl Indolyl-3-acetate

Oscar R. Suárez-Castillo, Myriam Meléndez-Rodríguez, Lidia Beiza-Granados, Indira C. Cano-Escudero,

Martha S. Morales-Ríos and Pedro Joseph-Nathan 451

Synthesis of Pyrrolidinoindolines from 2-(2-Oxo-3-indolyl)acetates: Scope and Limitations

Martha S. Morales-Ríos, Ernesto Rivera-Becerril, Daphne E. González-Juárez, Juan Benjamín García-Vázquez,

Joel J. Trujillo-Serrato, Angelina Hernández-Barragán, and Pedro Joseph-Nathan 457

Review/Account

Biomimetic Cyclization of Geraniol Derivatives, a Useful Tool in the Total Synthesis of Bioactive

Monocyclic Terpenoids

Valentina Merlini, Marco Luparia, Alessio Porta, Giuseppe Zanoni and Giovanni Vidari 465

Drimenol: A Versatile Synthon for Compounds with trans-Drimane Skeleton

Manuel Cortés, Virginia Delgado, Claudio Saitz and Veronica Armstrong 477

Recent Advances in the Synthesis of Sesquiterpenolides from Umbelliferae

Francisco M. Guerra, F. Javier Moreno-Dorado, Zacarías D. Jorge and Guillermo M. Massanet 491

Diterpene Lactones with Labdane, Halimane and Clerodane Frameworks

Lúcia Silva, Arlindo C. Gomes and Jesus M. L. Rodilla 497

Stereoselective Synthesis of Five Biologically Active, Naturally Occurring Medium and Large Ring Lactones

J. Alberto Marco and Miguel Carda 505

Enantioselective Synthesis of Alkaloids from Phenylglycinol-Derived Lactams

Mercedes Amat, Núria Llor, Rosa Griera, Maria Pérez and Joan Bosch 515

Aconitum lipo-alkaloids – Semisynthetic Products of the Traditional Medicine

Botond Borcsa, Dezső Csupor, Peter Forgo, Ute Widowitz, Rudolf Bauer and Judit Hohmann 527

Synthesis of Pterocarpans

Leticia Jiménez-González, Carmen Hernández-Cervantes, Míriam Álvarez-Corral, Manuel Muñoz-Dorado and

Ignacio Rodríguez-García 537

Hyaluronan: From Biomimetic to Industrial Business Strategy

Erminio Murano, Danilo Perin, Riaz Khan and Massimo Bergamin 555