modification of biodegradable polyurethanes by

48
VYSOKÉ UČENÍ TECHNICKÉ V BRNĚ BRNO UNIVERSITY OF TECHNOLOGY FAKULTA CHEMICKÁ ÚSTAV CHEMIE MATERIÁLŮ FACULTY OF CHEMISTRY INSTITUTE OF MATERIALS SCIENCE MODIFICATION OF BIODEGRADABLE POLYURETHANES BY BIOLOGICALLY ACTIVE SUBSTANCES MODIFIKACE BIODEGRADABILNÍCH POLYURETHANŮ BIOLOGICKY AKTIVNÍMI LÁTKAMI DIZERTAČNÍ PRÁCE DOCTORAL THESIS AUTOR PRÁCE Ing. VOJTĚCH KUPKA AUTHOR VEDOUCÍ PRÁCE doc. Ing. LUCY VOJTOVÁ, Ph.D. SUPERVISOR BRNO 2015

Upload: others

Post on 30-Dec-2021

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

VYSOKÉ UČENÍ TECHNICKÉ V BRNĚBRNO UNIVERSITY OF TECHNOLOGY

FAKULTA CHEMICKÁÚSTAV CHEMIE MATERIÁLŮ

FACULTY OF CHEMISTRYINSTITUTE OF MATERIALS SCIENCE

MODIFICATION OF BIODEGRADABLEPOLYURETHANES BY BIOLOGICALLY ACTIVESUBSTANCES

MODIFIKACE BIODEGRADABILNÍCH POLYURETHANŮ BIOLOGICKY AKTIVNÍMI LÁTKAMI

DIZERTAČNÍ PRÁCEDOCTORAL THESIS

AUTOR PRÁCE Ing. VOJTĚCH KUPKAAUTHOR

VEDOUCÍ PRÁCE doc. Ing. LUCY VOJTOVÁ, Ph.D.SUPERVISOR

BRNO 2015

Page 2: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

Vysoké učení technické v BrněFakulta chemická

Purkyňova 464/118, 61200 Brno 12

Zadání dizertační práce

Číslo dizertační práce: FCH-DIZ0107/2014 Akademický rok: 2014/2015Ústav: Ústav chemie materiálůStudent(ka): Ing. Vojtěch KupkaStudijní program: Makromolekulární chemie (P1422) Studijní obor: Chemie makromolekulárních materiálů (1405V003) Vedoucí práce doc. Ing. Lucy Vojtová, Ph.D.Konzultanti:

Název dizertační práce:Modifikace biodegradabilních polyurethanů biologicky aktivními látkami

Zadání dizertační práce:1) Vypracování literární rešerše na téma biodegradabilních polyurethanů a jejich modifikací biologickyaktivními látkami2) Příprava amfifilních biodegradabilních polyurethanových elastomerních filmů s různými poměry hydrofilnía hydrofobní složky a různým izokyanátovým poměrem bez použití rozpouštědla3) Chemicko-fyzikální charakterizace připravených polyurethanových filmů4) Modifikace a charakterizace vybraných PU filmů biologicky aktivními nanočásticemi s povrchovouúpravou zajišťující dobrou distribuci v PU matrici 5) Shrnutí výsledků a závěr

Termín odevzdání dizertační práce: 20.7.2015Dizertační práce se odevzdává v děkanem stanoveném počtu exemplářů na sekretariát ústavu a velektronické formě vedoucímu dizertační práce. Toto zadání je přílohou dizertační práce.

- - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - -Ing. Vojtěch Kupka doc. Ing. Lucy Vojtová, Ph.D. prof. RNDr. Josef Jančář, CSc.

Student(ka) Vedoucí práce Ředitel ústavu

- - - - - - - - - - - - - - - - - - - - - - -V Brně, dne 20.9.2014 prof. Ing. Jaromír Havlica, DrSc.

Děkan fakulty

Page 3: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

3

Abstract:

Presented dissertation thesis is focused on novel preparation of biodegradable

polyurethanes (PUs) and their modification by biologically active cellulose nanocrystals.

Literary review deals with current state of bioresorbable PUs used in tissue engineering.

Examples of prepared PU elastomers, scaffolds and injectable PUs, together with

biodegradation pathways to non-toxic products are summarized. The last part of the literary

review is targeting on nanocellulose, which has gained much attention for the use as

biomedical material due to its remarkable physical (high specific surface area, mechanical

reinforcement) and biological (biocompatibility, biodegradability and low toxicity) properties.

Experimental part presents characterization of biodegradable amphiphilic polyurethane

films (bio-PUs) synthesized by solvent free polyaddition reaction of hydrophilic

poly(ethylene glycol) (PEG) and hydrophobic poly(-caprolactone) (PCL) as macrodiols with

hexamethylene diisocyanate. Prepared bio-PUs were characterized on one hand by means of

different PEG/PCL ratio and on the other hand by changing the isocyanate ratio between

NCO/OH groups. Abrupt enhancement of mechanical properties was observed when

PEG/PCL weight ratio was equal to or less than 20/80 and was ascribed to the PCL ability to

form crystalline domains. The increasing amount of PEG promoted the ability of bio-PUs to

absorb water and enhance the rate of hydrolytic degradation. Whereas, reducing the ability of

bio-PUs to absorb water and prolonged time of hydrolytic degradation was achieved with

increasing the crosslink density by enhancing the isocyanate ratio. The last part deals with

novel solvent free preparation of nanocomposite utilizing bio-PU as a matrix and cellulose

nanocrystals either neat or surface grafted by PEG. Structural analysis demonstrated that the

presence of rod-like nanoparticles causes the immobilization of the PU chains in matrix

resulting in increased stiffness and rigidity of bio-PU/cellulose nanocomposite.

By adjusting the PEG/PCL ratio, the amount of isocyanate or the presence of nanofiller,

the novel bio-PU material with desirable mechanical (toughness, flexibility) and physical

(swelling, degradation) properties can be obtained. Prepared solvent free bio-PUs may

advantageously be used in regenerative medicine for soft tissue regeneration (e.g. as vascular

grafts).

Keywords:

Polyurethane, poly(ethylene glycol), poly(-caprolactone), nanocellulose, nanocomposite,

tissue engineering

Page 4: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

4

Abstrakt:

Předkládaná dizertační práce se zabývá novým způsobem přípravy biodegradabilních

polyuretanů (PU) a jejich modifikací biologicky aktivními celulózovými nanokrystaly.

Literární rešerše se zaměřuje na bioresorbovatelné PU v tkáňovém inženýrství. Shrnuje

příklady těchto PU elastomerů, skafoldů (nosičů buněk) i injektovatelných PU společně se

způsoby biodegradace na netoxické produkty. Poslední část je zaměřena na nanocelulózu,

která si získala pozornost díky svým pozoruhodným fyzikálním (velký specifický povrch,

mechanické vlastnosti) a biologickým (biokompatibilita, biodegradabilita a nízký toxicita)

vlastnostem jako materiál pro biomedicínu.

V experimentální části byly charakterizovány amfifilní biodegradovatelné polyuretanové

filmy (bio-PU) syntetizované bez použití rozpouštědla polyadiční reakcí z hydrofilního

poly(ethylenglykolu) (PEG) a hydrofobního poly(-kaprolaktonu) (PCL) jako makrodiolů

společně s hexamethylen diizokyanátem. Připravené bio-PU filmy byly charakterizovány pro

různé poměry jak mezi PEG/PCL, tak i mezi NCO/OH reagujícími skupinami (izokyanátový

poměr). Bio-PU filmy projevily markantní nárůst mechanických vlastností při hmotnostním

poměru PEG/PCL rovnému nebo menšímu než 20/80 díky vzniku krystalických domén PCL.

Přítomnost PEGu zvyšovala schopnost bio-PU filmu absorbovat vodu i urychlila jeho

hydrolytickou degradaci. Oproti tomu nižší absorpční schopnost a delší čas hydrolytické

degradace materiálu způsobil vyšší izokyanátový poměr, a tedy i vyšší síťová hustota. Třetí

část práce se zabývá přípravou polyuretanových nanokompozitů unikátní metodou bez použití

rozpouštědla za využití bio-PU matrice a celulózových nanokrystalů buď nemodifikovaných,

nebo povrchově roubovaných PEGem. Strukturní analýza prokázala, že přítomnost

tyčinkovitých nanočástic způsobuje imobilizaci polymerních segmentů, v důsledku čehož se

zvýšila tuhost a křehkost materiálu.

Nastavením vhodného poměru mezi PEG/PCL, množstvím izokyanátu, či přídavkem

modifikovaného nanoplniva může být bio-PU materiál „ušit na míru“ s vhodnými

mechanickými (houževnatost, tažnost) a fyzikálními (botnání, degradace) vlastnostmi. Díky

přípravě bez použití rozpouštědla by mohly být připravené materiály využity v regenerativní

medicíně jako např. cévní štěpy.

Klíčová slova:

polyuretan, polyethylenglykol, poly(-kaprolakton), nanocelulóza, nanokompozit, tkáňové

inženýrství

Page 5: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

5

KUPKA, V. Modification of Polyurethanes by Biologically Active Substances. Brno: Brno

University of Technology, Faculty of Chemistry, 2015. 98 p.

Supervisor doc. Ing. Lucy Vojtová, Ph.D.

Declaration

I declare that my Doctoral Thesis was worked out by myself and that used references

are quoted correctly and fully. The content of the above mentioned thesis is the property of

BUT Faculty of Chemistry and can be used for commercial purposes only with the

supervisor’s and dean’s consents.

……………………….

autograph of author

Acknowledgement:

I would like to thank my supervisor doc. Ing. Lucy Vojtová, Ph.D. for her professional

guidance and prof. RNDr. Josef Jančář, CSc. for providing me the working conditions. I

would also like to thank Assoc. Prof. Qi Zhou for supplying cellulose nanocrystals, Mohd

Farhan Ansari for DMA measurements, doc. RNDr. Miroslav Šlouf, Ph.D. for acquiring TEM

images, Mgr. Zdenka Fohlerová, Ph.D. for cell response analysis and Ing. Radka Bálková,

Ph.D. for discussions regarding structural analysis.

I gratefully appreciate the help of all my colleagues, present and past, at Faculty of

Chemistry, BUT, at the Department of Materials Science for making my stay at University a

pleasant one.

Finally, yet importantly I would also like to thank my parents for their lifelong support and

my dear wife Hanča.

Page 6: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

6

List of Contents:

1. INTRODUCTION .......................................................................................................... 8

2. THEORETICAL PART ............................................................................................... 10

2.1. Chemical structure of polyurethanes ...................................................................... 10

2.2. Bioresorbable polyurethanes .................................................................................. 12

2.3. Feedstocks for synthesis of bioresorbable polyurethanes ...................................... 13

Hard segments .................................................................................................. 14 2.3.1.

Soft segments ................................................................................................... 16 2.3.2.

2.4. Biodegradation of polyurethanes ........................................................................... 19

2.5. Polyurethane scaffolds ........................................................................................... 21

2.6. Injectable polyurethanes ......................................................................................... 23

2.7. Polyurethanes modified by biologically active substances .................................... 24

Nanocellulose - general properties ................................................................... 24 2.7.1.

Preparation of CNC .......................................................................................... 26 2.7.2.

Nanocellulose-polyurethane composites .......................................................... 26 2.7.3.

Nanocellulose in medicine ............................................................................... 27 2.7.4.

3. MAIN AIMS ................................................................................................................ 29

4. EXPERIMENTAL PART ............................................................................................ 30

4.1. Chemicals ............................................................................................................... 30

4.2. Solvent free synthesis of bio-PU ............................................................................ 30

4.3. Synthesis of bio-PU nanocomposites ..................................................................... 30

4.4. Characterization ..................................................................................................... 30

UV-VIS ............................................................................................................ 30 4.4.1.

Attenuated total reflection infrared spectroscopy (ATR-IR) ........................... 30 4.4.2.

Thermogravimetric analysis (TGA) ................................................................. 30 4.4.3.

Differential scanning calorimetry (DSC) ......................................................... 30 4.4.4.

Wide-angle X-ray scattering (WAXS) ............................................................. 31 4.4.5.

Swelling ............................................................................................................ 31 4.4.6.

Hydrolytic degradation ..................................................................................... 31 4.4.7.

Extraction ......................................................................................................... 31 4.4.8.

Tensile measurement ........................................................................................ 32 4.4.9.

Dynamic light scattering .................................................................................. 32 4.4.10.

Dynamic mechanical analysis (DMA) ............................................................. 32 4.4.11.

Page 7: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

7

Transmision electron microscopy (TEM) ........................................................ 32 4.4.12.

Cytotoxicity evaluation .................................................................................... 32 4.4.13.

5. RESULTS AND DISCUSSION .................................................................................. 34

6. CONCLUSIONS .......................................................................................................... 35

7. REFERENCES ............................................................................................................. 36

8. LIST OF SHORTCUTS AND SYMBOLS ................................................................. 45

9. APPENDIX .................................................................................................................. 47

10. LIST OF FIGURES ...................................................................................................... 47

11. LIST OF TABLES ....................................................................................................... 47

Page 8: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

8

1. INTRODUCTION

Polyurethanes (PUs) form diverse class of elastomers covering broad range of chemical

and physical properties. In addition to industrial applications, their structure can also be

designed to form bioresorbable materials for use in human medicine [1–3]. Important features

of PUs for the use in tissue engineering are tissue-specific biocompatibility, biodegradability,

mechanical flexibility and moderate blood compatibility [4]. PUs have been available in

medicine since the 1960s as bio-inert, hydrolytically stable materials (e.g. ventricle assist

devices or heart valves [5]). Later on, bioresorbable PUs were introduced for various tissue

constructs such as nerve conduits [6], vascular grafts [7], cartilage [8], cancellous bone graft

substitutes [9] or as grafts for small diameter vascular replacement [10].

Moreover, bioresorbable PUs can be tailored to possess a broad range of mechanical

properties by selections and content of soft and hard segments. Soft segments are commonly

oligomeric macrodiols represented by linear polyesters such as poly(-caprolactone),

poly(lactic acid) and poly(glycolic acid) or polyethers like poly(ethylene glycol) and

poly(propylene glycol) having a low glass transition temperature (lower than 25 °C). The hard

segments are provided by the combination of the chain extender and the diisocyanate

component. The nature of hydrogen bonding in the hard segment causes a mutual attraction

leading to a formation of hard and soft segment domains [11]. To eliminate the concern of

aromatic amine by-products, the utilization of aliphatic diisocyanates (as alternatives to

aromatic ones) provide a route to synthetize biodegradable polyurethanes yielding non-toxic

degradation products [3].

Generally, current bioresorbable PUs are prepared in dimethyl sulfoxide or

dimethylformamide (DMF) solvent followed by casting into a mold. This overcomes the

problem with hydrophilic PEG component in PU, which is grabbing easily the water from air

moisture within polymerization. The water reacts fast with isocyanate resulting in bubbles of

carbon dioxide. The same strategy is commonly used for preparation of PU nanocomposites

with cellulose nanocrystals (CNC). PU matrix is dissolved in DMF and CNC are dispersed in

similar solvent. Mixing of those low viscous dispersions is enabling preparation of material

with fine dispersion of nanoparticles. However, avoiding the use of organic solvent is

favorable parameter in the material processing with the potential use in medicine. Therefore,

preparation of either PU itself or PU-CNC nanocomposite in bulk is challenging.

The aim of the work was to develop solvent free procedure eliminating organic solvents

routinely used in PU synthesis. Polymerization conditions yielding compact biodegradable

films were optimized. The experimental system was based on bioresorbable macrodiols from

PEG and PCL and aliphatic 1,6-diisocyanato hexane (HDI), which is claimed as a non-toxic

amine producer during polyurethane degradation [3]. For preparation of nanocomposites was

chosen PU made of only PEG and HDI due to its amorphous nature. Effect of nanofiller on

the matrix is then not shielded by the effects associated with crystallization.

Page 9: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

9

A range of bioresorbable PU systems with similar molecular structure [12-17] or

nanocomposites made of PU and CNC [18, 19], has already been described in literature. Thus,

the approach employed in this work to synthesize thermally crosslinked bio-PUs and PU-

CNC nanocomposites without the use of organic solvent is novel and original.

Page 10: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

10

2. THEORETICAL PART

2.1. Chemical structure of polyurethanes

Polyurethanes (PU)s are a group of polymers prepared according to the diisocyanate-

polyaddition principle. The name polyurethane is derived from ethylcarbamate, known as

urethane. The reaction of polyisocyanate with polyol leads to the formation of urethane bond,

as illustrated in Figure 1.

OCN R NCO +n

polyaddition

C NH R NH C O

O

CH2 O

O

diisocyanate

n

n

OH O OH

or polyether

polyol

polyester

urethane bond

or polyether

polyester

Figure 1: General formation of polyurethane.

Compared with other functional groups like ether-, ester-, and also urea-groups, the

urethane groups represent often only a minority of the total composition (e.g. 4–6 % in

flexible foams) [20]. Therefore, the properties are not significantly affected by the urethane

groups. Most polyurethanes are composed of at least three components – long chain polyol,

diisocyanate and chain extender.

PU elastomers are characterized by a segment structure (alternating block copolymers) of

the primary chain as showed in Figure 2. The secondary and tertiary structure and

consequently, the morphology of these polyurethanes depend on the chemical composition

and on the length of the segments (blocks). PU elastomers can be microphase separated to

form hard and soft domains due to differences in polarity between the hard (polar) and soft

(nonpolar) segments [11]. The soft segments typically have a glass transition temperature

lower than 0 °C and are therefore rubbery at the room temperature.

Page 11: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

11

Figure 2: Modelled primary structure of segmented polyurethane [20].

PUs can be either physically or chemically cross-linked by using the components with

more than two functional groups, e.g., triisocyanate or a multihydroxyl polyol [21].

Hydrogen bonding (Figure 3) between urethane and urea groups in the hard segments of

adjacent polymer chains induces the formation of ordered hard domains, which function as

physical crosslinks that resist flow when stress is applied to the material. To process the

polymer, the physical crosslinks are broken by heating the material above the hard segment

melting transition or by dissolving the material in an aprotic solvent, such as

dimethylformamide (DMF).

Figure 3: Hydrogen bonding between the chains of a polyurethane based on 1,6 – hexane

diisocyanate and 1,4 – butane diol depicted according to [20].

Page 12: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

12

2.2. Bioresorbable polyurethanes

It is important to distinguish between biodegradable and bioresorbable materials.

Biodegradability means that the material degrades and moves away from the site of its action

in vivo, but is not necessarily removed from the body. Bioresorbability then means total

elimination of the initial foreign material and degradation of all low molecular weight

compounds with no residual side effects [21].

Important features of PUs for tissue engineering are tissue-specific biocompatibility,

biodegradability, mechanical flexibility and moderate blood compatibility. PUs were initially

used in medicine as biostable materials. Various implantation sites for biostable PUs occurred

in areas such as cardiovascular system (artificial heart [24], vascular grafts [25, 26] and stents

[3, 27, 28]), the middle ear (artificial tympanic membrane [29]), the eye (artificial intraocular

lenses [30]), the digestive tract (stent-like extensions for the oesophagus [31] or the

replacement of biliary ducts [32]).

Applications of bioresorbable polyurethanes appeared later on. As scaffolds they have

already been used for various tissue constructs such as nerve conduits [6], vascular grafts [7],

cartilage [8] or cancellous bone graft substitutes [9]. The usage of polyurethanes in medicine

divided by standards of United States Food and Drug Administration (U.S. FDA) is

summarized in Table 1.

Table 1: Applications of polyurethanes in tissue engineering divided by U.S. FDA standards

[11].

U.S. FDA class I or II: U.S. FDA class III:

• Catheters, drains, medical tubing

• Blood bags

• Transdermal drug delivery patches

• Surgical drapes and gowns

• Transient cardiovascular devices

• Intra-aortic balloon pumps

• Temporary left ventricular assist devices

and biventricular assist devices

• Interim cardiovascular devices

• Ventricular assist devices

• Total artificial hearts used as temporary

measure prior to organ transplantation

(generally intended for less than a month)

• Permanent cardiovascular devices

(e.g. totally implantable ventricles,

artificial hearts, pacemakers and leads,

artificial heart valves)

• Blood conduits and access devices

• Wound dressings and barrier scaffolds

• Contraceptives

Note: Device FDA classification depends on the intended use of the device and also on the indications for

use. In addition, classification is risk based. This means the risk the device poses to the patient and/or the user is

a major factor in the class it is assigned. Class I includes devices with the lowest risk and Class III includes those

with the greatest risk [33].

Page 13: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

13

2.3. Feedstocks for synthesis of bioresorbable polyurethanes

The stability of PUs in the body depends on a number of factors including structure and

composition. The synthesis of PUs requires at least two components, the first one is isocyanate

and the second one is a bi- or multi-functional polyol.

The direct reaction of diisocyanate with a long-chain diol usually yields a soft polymer

with low mechanical strength. This property is usually changed by the addition of a chain

extender, which is low molecular weight diol or diamine. Diamine chain extenders yield

poly(urethane urea)s, which have higher hard segment melting temperatures and therefore

harder mechanical properties compared to PUs obtained from diol chain extenders [34]. The

properties of the final PU are primarily dependent on the chemical nature of mentioned three

building blocks and the relative proportions between them used during synthesis.

To increase the reaction rate, urethane catalysts such as tertiary amines or compounds as

organotin and/or elevated temperatures (60–90 °C) may be used. Dibutyltin dilaurate and

stannous octoate prevail in the articles about biodegradable PUs. Stannous octoate is more

often used as catalyst due to its lower cytotoxicity compared to dibutyltin dilaurate [35]. The

structure of both of them is shown in Figure 4.

a)

H3C(H2C)9H2C

O

O

Sn

O CH2(CH2)9CH3

O

CH3

CH3

b)

O

O-

O

O-

Sn2+

Figure 4: Chemical structure of a) dibutyltin dilaurate and b) stannous octoate.

Page 14: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

14

Hard segments 2.3.1.

Hard segments are in PU structure represented by isocyanates and chain extenders.

Traditional aromatic diisocyanates are replaced by linear ones to avoid toxic or carcinogenic

degradation products. The structures of polyisocyanates used to synthesize biodegradable PU

biomaterials are showed in Table 2. Important aliphatic isocyanates used are 1,4-

diisocyanatobutane (BDI), hexamethylene diisocyanate (HDI), dicyclohexylmethane

diisocyanate (H12MDI). Lysine-derived polyisocyanates, including lysine methyl (or ethyl)

ester diisocyanate (LDI) and lysine triisocyanate have been extensively investigated, as they

did not show any harmful effects in vivo [36]. As published by Zhang et al. [37] in their tests

on rabbit bone marrow stromal cells, LDI-glucose synthesized polymers induced no

immunogenic reaction and no antibody responses in the host. Their PU did not cause any

accumulation of macrophages (foreign body giant cells) or tissue necrosis around the material.

Table 2: Isocyanates used in polyurethane chemistry for tissue engineering.

Chemical name Structure

1,4-Diisocyanatobutane (BDI)

N C O

NCO

1,6-Diisocyanatehexane

or hexamethylene diisocyanate (HDI)

N C O

NCO

Dicyclohexylmethane diisocyanate

(H12MDI)

N C ONCO

Lysine methyl ester diisocyanate

(LDI)

NCO

OO

N C O

CH3

Reactions of isocyanate are summarized in Figure 5a-e. Water reacts with isocyanates to

form carbamic acid which is an unstable compound and decomposes immediately to an amine

and carbon dioxide gas. The water reaction is used to synthesize PU foams (called in tissue

engineering scaffolds), where the carbon dioxide gas acts as a blowing agent. Isocyanates

react with hydroxyl- and amino-functional molecules by addition to the carbon-nitrogen

Page 15: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

15

double bond. Urethane bonds are formed by reaction of isocyanates with hydroxyl-functional

molecules and urea linkages are formed by reaction with amines. The excess of isocyanate is

used to introduce certain degree of crosslinking, which is desirable for improved mechanical

strength, since it leads to the formation of allophanate and biuret branch points [34].

a) With water: amine

R N C O + H2O R NH2C OH

O

R N

H

+ CO2

b) With alcohol: urethane

R N C O OH R'+R

NH

CO

O

R'

c) With amine: urea

R N C O NH2 R'+ R

NH

CO

NH

R'

d) With urethane group: allophanate

R N C OR'

NH

CO

O

R''+

RNH

CON

R'

COO

R''

e) With urea group: biuret

R N C O + R'

NH

CO

NH

R'' RNH

CON

R'

CONH

R''

Figure 5: Reactions of isocyanate with other functional groups.

The relative fractions of the hard and soft segments affect the mechanical properties of

segmented PUs. Hard segment content increases with increasing chain extender to polyol

ratio. This yields harder and stiffer polymers with higher tear strength but lower elongation at

break. Gisselfaelt et al. [38] demonstrated this in PUs made of MDI and 1,3-diaminopropane

as hard segment part and PCL as a soft segment. Reducing the PCL soft segment molecular

weight from 2000 to 530 g·mol-1

increased the hard segment content from 21.9 to 51.4 wt%,

Page 16: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

16

resulting in increased stiffness (6 to 45 MPa) and decreased elongation at break (77 to 32 %).

The polymer became harder and less elastic with increasing hard segment content.

Soft segments 2.3.2.

Polyols are viscous liquids with hydroxyl end groups and a polyether, polyester,

polycarbonate, polydimethylsiloxane, or polybutadiene backbone. The majority of

biodegradable PUs are based on polyester or polyether macrodiols. Typical representatives

include polyethyleneglycol (PEG), polypropyleneglycol (PPG), polylactic acid (PLA),

polyglycolic acid (PGA) and their copolymer polylactic-co-glycolic acid (PLGA),

polycaprolactone (PCL) or polyhydroxybutyrate (PHB). Their properties and characterization

have already been reviewed many times [39, 40]. Their major properties are listed in Table 3.

Soft segments affect biostability in the PU materials. Polyester soft segments were found to

be more susceptible to hydrolytic degradation and attack by microorganisms than polyethers

[1].

2.3.2.1. Poly(ethylene glycol)

Poly(ethylene glycol) (PEG), also known as poly(ethylene oxide), poly(oxyethylene) or

polyoxirane, is a hydrophilic, non-ionic polymer. PEG is known for its biocompatibility and

low toxicity and is soluble in water via hydrogen bonding interactions. It has been approved

by the U.S. FDA for internal consumption [41]. It can make a surface highly resistant to

biological fouling and can reduce protein adsorption and resistance to bacterial and animal

cell adhesion. Most PEGs with molecular weight < 1000 g·mol-1

are removed from the body

unaltered only dissolved in the body fluids [42]. It is also apparently not readily recognized by

the immune system. Modifying proteins with PEG have been shown to reduce the

immunogenicity and antigenicity of these proteins and to increase circulation times [43].

PEGs having molecular weight less than 1000 g·mol-1

are viscous, colorless liquids. Higher

molecular weight PEGs are waxy, white solids. The melting point of the solid is proportional

to molecular weight, approaching a plateau about 67 °C. The molecular weights commonly

used in biomedical applications range from a few hundreds to 20 000 g·mol-1

[42].

Block copolymer of poly(ethylene-propylene-ethylene glycol) is sold under the trademark

Pluronic®[45] and is often used in synthesis of bioresorbable PUs. Gorna and Gogolewski

[46] synthesized linear biodegradable polyurethanes with varying ratios of the hydrophilic-to-

hydrophobic segment. The hydrophilic segment was based on Pluronic® and the hydrophobic

segment was based on PCL diol. The polymers absorbed up 3.9 % of water depending on the

chemical composition. Tensile strength, Young's modulus and elongation at break of the

polymers were in the range of 11–46 MPa, 4.5–91 MPa and 370–960 %. Degradation in vitro

caused 2 % of mass loss in 48 weeks. The extent of degradation was dependent on the

polymer composition and the hydrophilic segment content.

Page 17: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

17

Table 3: Physical properties of the biodegradable polymers used as polyols in formation of polyurethane.

Synthetic polymer Thermal and mechanical properties Degradation properties Applications Ref.

Tm (°C) Tg (°C) Elastic

modulus (GPa)

Time

(months) Products

PEG - Upper limit

at 67 °C - Immediately

Dissolves in body

fluids

Laxatives, bowel preparation

before surgery, carrier for active

ingredients

42

PCL 58-63 -60 0.4 >24 Caproic acid Sutures, dental orthopaedic

implants 47

PGA 225-230 35-40 5-7 3-4 Glycolic acid Suture anchors, meniscus repair,

medical devices, drug delivery 50

PLA 170 56 8.5 12-18 L-lactic acid Fracture fixation, suture anchors,

meniscus repair 40, 50

PLGA (50/50) Amorphous - 2.0 1-2 weeks D,L-lactic acid and

glycolic acid - 43

PLGA (85/15) Amorphous - 2.0 5-6 weeks D,L-lactic acid and

glycolic acid

Interference screws, suture

anchors, ACL reconstruction 43

PHB 177 2 3.5 >17 D-3-hydroxybutyric

acid Screws 55, 57

Page 18: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

18

2.3.2.2. Poly(-caprolactone)

Poly(-caprolactone) (PCL) is a semicrystalline, bioresorbable polymer belonging to the

aliphatic polyesters. PCL has melting point ranging between 59 and 64 °C and a glass

transition temperature of about −60 °C. PCL can be prepared either by ring-opening

polymerization of -caprolactone using a variety of anionic, cationic and co-ordination

catalysts or via free radical ring-opening polymerization of 2-methylene-1-3-dioxepane [47].

It is regarded as a soft and hard tissue compatible bioresorbable material and has been used in

biomedicine e.g. as a scaffold, sutures or fixation devices [48].

The PCL homopolymer has a total degradation of 2–4 years depending of the starting

molecular weight used for the implant. Its degradation pathway undergoes a two-stage

degradation process. First, the non-enzymatic hydrolytic cleavage of ester groups, and second,

when the polymer has lower molecular weight (less than 3000) it undergoes intracellular

degradation as evidenced by observation of PCL fragments uptake in phagosomes of

macrophages and giant cells and within fibroblasts [47]. Local decrease of pH during the first

stage has been observed same as in other polyesters. The combination of PCL with other

materials such as bioceramics can suppress the local decrease of pH during degradation [49].

2.3.2.3. Polyglycolic acid

Polyglycolic acid (PGA) is a semicrystalline biodegradable polymer known to lose its

strength in 1–2 months when hydrolyzed and losses mass within 6–12 months. PGA sutures

(Dexon®) have been commercially available since 1970 [50]. The hydrolytic degradation in

vivo may take place via nonspecific enzymes such as esterases and carboxyl peptidases that

produce glycolic acid monomers. Glycolic acid monomers are then converted enzymatically

either into glycine, which can be used in protein synthesis, or into pyruvate, that enters the

tricarboxylic acid cycle and yielding energy, CO2 and water [51]. Glycolic acid is partially

excreted in urine. Hydrolysis is affected by the initial molecular weight, surface area/weight

ratio, porosity and monomer concentration, geometric isomerism, conformation and

crystallinity. Increased pH accelerates PGA degradation.

2.3.2.4. Polylactic acid (PLA)

Polylactic acid (PLA) is a thermoplastic, high strength, high modulus polymer, which

belongs to the family of aliphatic polyesters. Poly(L-lactide) is a crystalline polymer

(approximately of 37 % crystallinity) while poly(D,L-lactide) is amorphous. Poly(L-lactide)

has a glass transition temperature between 60–65 °C and a melting temperature approximately

of 175 °C, while poly(D,L-lactide) has glass transition temperature between 44–55 °C [40].

PLA undergoes scission of chains in the host body to monomeric units of lactic acid, which is

a natural intermediate in carbohydrate metabolism. These characteristics make this polymer

suitable for use as resorbable sutures, carries for the controlled release of drugs, implants for

Page 19: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

19

orthopaedic surgery or blood vessels, which finally can be replaced by living tissues. The

attraction of PLA as a biodegradable material is its ready availability from renewable

resources such as corn starch (in the U. S.) and sugarcanes (rest of world). The PLA life cycle

starts with corn starch. The plants are first milled to separate the starch, which is converted to

lactic acid utilizing fermentation and a series of purification steps. It is fully compostable. It

can be converted back to monomer and oligomer by enzymatic degradation, or it can be

degraded into water, carbon dioxide and organic materials [52].

2.3.2.5. Poly(lactic-co-glycolic acid)

Poly (lactic-co-glycolic acid) (PLGA) copolymer is built from the units of poly(lactic acid)

and poly(glycolic acid). The rate of polymer degradation may affect many cellular processes,

including cell growth, tissue regeneration, and host response. PLGA has been shown to

undergo bulk erosion through hydrolysis of the ester bonds and the rate of degradation

depends on a variety of parameters including the PLA/PGA ratio, molecular weight, and the

shape and structure of the matrix. The degradation products are endogenous compounds

(lactic and glycolic acid) and are nontoxic. The rate of degradation can be controlled by the

ratio of PLA and PGA. The higher the content of glycolide units the faster is the degradation

[53]. PLGA has good cell adhesion and proliferation. Various studies have been performed so

far using micro- and nano-fabrication techniques to form three-dimensional scaffolds based

on PLGA [43]. Another application of biodegradable PLGA is for use in guided tissue

regeneration by providing a permeable material for space preservation or in drug delivery

systems [54].

2.3.2.6. Polyhydroxybutyrate

Poly-3-hydroxybutyrate (PHB) belongs to a group of polyesters called

polyhydroxyalkanoates (PHAs). PHAs are biodegradable plastics produced by

microorganisms. They have mechanical properties similar to some common synthetic

thermoplastics and are environmentally degradable, where the products of decomposition are

CO2 and H2O [55]. PHB has mechanical properties quite similar to commercial polypropylene

with Young’s modulus 3.5 GPa, tensile strength of 40 MPa and melting point at 179 °C.

Moreover, product of PHB degradation (3-hydroxybutyric acid) belongs to short-chain fatty

acids and reveals antibacterial activity [56].

2.4. Biodegradation of polyurethanes

PUs were firstly used as a biostable materials in medicine and since that time have been

applied in a range of medical devices [58, 59] In late 1980s a failure on functions in the

applications such as the pacemaker leads and breast implant coatings containing PUs was

found [3]. Aromatic diisocyanates (e.g. methylene diphenyl diisocyanate (MDI) and toluene

diisocyanate) have been recognized as potentially carcinogenic by-products of degradation.

Page 20: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

20

To solve this problem, aliphatic diisocyanates such as 1,4-diisocyanatobutane (butyl

diisocyanate, BDI) and lysine diisocyanates (LDI) started to be used instead of aromatic ones.

These polymers exhibit better blood and tissue compatibility both, in vitro and in vivo tests, in

comparison to materials where aromatic reactants were used [39].

PU elastomers with hydrophilic soft segments have been reported to exhibit increased

water uptake, which has also been suggested to increase the degradation rate [38]. Mechanism

of hydrolytic degradation that has been suggested by Hafeman et. al [63] is shown in Figure 6.

Above mentioned phenomenons can be used to design PU materials with targeted physical

properties.

Figure 6: Schematic showing hypothesized mechanisms of PU scaffold degradation and

soluble breakdown products. (A) Hydrolysis of ester bonds to yield -hydroxy acids (dotted

line). (B) Abstraction of the -hydrogen atom in the R group adjacent to the urethane bonds

by reaction with hydroxyl radicals to yield lysine or ethanolamine and carbon dioxide. (C)

Abstraction of the -hydrogen atom in lysine by reaction with hydroxyl radicals to yield

multiple degradation products, including NH4, -ketoacids, oximes, CO2, and carboxylic

acids (degradation products not shown to simplify the diagram) [63].

The stability of segmented (block) PUs in the body depends not only on the composition

and structure. It also depends on the presence of stabilizing additives, the chosen implant site

and the sample’s purity and processing history. Fabrication technique, handling, residual

solvent, sterilization method and device design are also important factors [23]. The common

degradation pathway of poly(ester urethanes) is the hydrolysis of ester linkages. Through this

way the degradation products are hydroxy acids as well as urethane and urea fragments with

terminal acid groups. The hydrolysis of urethane and urea linkages to amine has been also

Page 21: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

21

reported [60]. Hydrolysis of ester bonds is taking control of the degradation rate. The

formation of acidic degradation products is known to autocatalyze polymer degradation and

may cause in vivo inflammation [60].

By incorporating a chain extender that is recognizable by an enzyme, PU elastomers with

degradable hard segments have been also synthesized [61]. Guan et al. [62] synthesized PUs

from BDI, PCL-PEG-PCL macrodiol triblock copolymers and the peptide Alanine-Alanine-

Lysine (AAL) as a chain extender. The hard segment of this polymer was designed to be

enzymatically degradable. Polymers synthesized from the AAL chain extender degraded

faster than those synthesized from tetramethylenediamine (putrescin®) chain extender and the

degradation products were observed to be non-cytotoxic. Further, materials synthesized from

the AAL chain extender displayed mechanical properties comparable to those synthesized

from usually used chain extender putrescin®. Obtained tensile strengths ranged from 15 to

28 MPa, breaking strains 670–890 %.

2.5. Polyurethane scaffolds

Many techniques/methods are employed to process biomaterials into scaffolds. The reason

why to make scaffolds rather porous than solid, is that cell colonization, in-growth and

proliferation can proceed easier. Porosity leads to a weaker structure and it can also accelerate

degradation due to the larger surface area. Having a porous structure allows nutrients to flow

into the cells in the inner regions of the scaffold and for waste to be removed. The ideal pore

size and pore morphology of the scaffold depends on the intended purpose of the scaffold. It

has been claimed that the optimal pore size is between five and ten times the diameter of the

cell i.e. 100–300 μm [65].

Techniques used for processing of scaffolds have been reviewed many times [65, 66].

Because the dissertation thesis is not directly focused on preparation of scaffold, there is just a

list of them. Techniques involve solvent casting and particulate leaching, gas foaming, non-

woven fibers, fiber knitting and phase separation/emulsion freeze drying. Emerging

fabrication techniques under development include solid free forming techniques, including 3D

printing and fused deposition. Characteristics differentiating the techniques are the use of

solvents, heat, pressure, or pore creating agents.

The scaffold should be non-immunogenic, non-toxic, biocompatible, biodegradable and

easily manufactured. The cells survival and growth is affected by macro and micro-structural

properties. Scaffold should possess an interconnected and spread porosity (usually exceeding

90 %) with a highly porous surface and microstructure. This would allow in vitro cell

adhesion and would provide the necessary space for neovascularization in vivo. Pore

interconnectivity directly influences the diffusion of physiological nutrients and gases to cells

as well as the removal of metabolic waste and by-products from cells.

Page 22: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

22

Grenier et al. [67] examined two medical grade polyurethanes for their potential use as

scaffold for vascular tissue engineering applications. Those two polyurethanes were

commercially available Bionate 55D (polycarbonate-urethane – thermoplastic elastomer

formed from hydroxyl terminated polycarbonate, an aromatic diisocyanate, and a low

molecular weight glycol as a chain extender [68]) and Elasthane 75D (polyether-urethane –

made of poly(tetramethylene oxide) and an aromatic diisocyanate and a low molecular weight

glycol chain extender [68]). Scaffolds were prepared via pressure differential/particulate

leaching technique, which improved the pore interconnectivity of the scaffolds. Two types of

porogen were used for fabrication of the scaffolds, NH4Cl particles, and paraffin spheres. In

the preparation procedure, they firstly dissolve the polyurethane in dimethylformamide. Then

the cylindrical mold was filled with porogen and using the pressure dissolved polymer was

pushed into the mold. After a certain time the mold was removed from the apparatus and

placed in a fume hood at ambient temperature for 2 days to evaporate the solvent. The

polymer matrix was then pushed out of the mold and immersed in deionized water for NH4Cl

particles or hexane for paraffin spheres, to leach out the porogen.

Sharifpoor et al. [69] synthesized degradable polar hydrophobic ionic polyurethane (D-

PHI) porous scaffold. D-PHI scaffold properties were adjusted through the introduction of a

lysine-based cross-linker. Increasing lysine-based cross-linker concentration resulted in an

increase of the elastic modulus (from 0.5 to 21 MPa), a decrease of the elongation-at-yield

(from 45 to 5 %) and a reduction of scaffold swelling (from 170 to 100 %). Based on a

preliminary study with vascular smooth muscle cells, D-PHI scaffolds demonstrated the

ability to support cell adhesion and growth during 2 weeks of culture, suggesting their

potential suitability for longer term vascular tissue engineering. The versatility of the D-PHI

properties may allow for the tailoring of cell-material interaction and ultimately functional

tissue regeneration.

Page 23: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

23

2.6. Injectable polyurethanes

Synthetic polymer systems as injectable liquids, gels or pastes have the advantage for the

surgical operations, because they minimalize the invasive procedures. The way how to deliver

the gel to the proper place in the body is for example arthroscopic delivery. Injectable

polymer systems could help in complex fracture and bone defect repair, where substantial

amount of bone is lost due to trauma or disease. Further, they can be used to stabilize and

reinforce the fixation of implants such as plates and screws, particularly in patients with

osteoporotic bone [70]. Several biodegradable injectable hydrogel systems made of

polyurethanes have been suggested as described below.

Thermoresponsive polymers showing enhanced rheological properties were published by

Cohn et al. [71]. PUs were synthesized from PEO-PPO-PEO triblock macrodiols end capped

with lactid acid or -caprolactone oligoesters and chain extended with HDI. The length and

composition of the ester blocks affected the viscosity response with temperature. Gels with

viscosities up to 193 kPa∙s were prepared. Degradation times ranging from days to months

were achieved by varying the composition of the backbone. The improved rheological

properties of these materials were suggested as potential injectable drug delivery system.

Guelcher et al. [72] introduced two-component injectable polyurethane hydrogels as useful

biodegradable material for fracture healing. PU network consisted of reactive liquid molding

low-viscosity prepolymers derived from lysine polyisocyanates and poly(caprolactone-co-

D,L-lactide-co-glycolide) triols. The values of Young’s modulus ranged from 1.20

to 1.43 GPa, and the compressive yield strength varied from 82 to 111 MPa, which is

comparable to the strength of poly(methyl methacrylate) bone cements. They studied the

biodegradation in vitro, where the materials decomposed to non-cytotoxic products. As well,

the material supported the attachment and proliferation of human osteoblasts.

An injectable, two component LDI-based polyurethane system with commercial name

PolyNovo® [73] was developed for orthopaedic applications with curing in situ. This self-

setting system can be inserted to the body arthroscopically in liquid form. It polymerizes at

physiological temperature in situ to provide appropriate bonding strength and mechanical

support comparable to or superior to widely used bone cements. This material has also been

shown to promote favorable cell adhesion and proliferation [74].

Although quite a lot of articles dealing with injectable PU concept have been published,

the injectable PUs have still basic problems that should be overcome. The first one is the

toxicity and ultimate fate of reactive components that are not incorporated in the final cured

product [75]. The nature of the reaction or reactivity of the NCO-terminated prepolymer is

exothermic, which may destroy the surrounding tissue. And finally, diffusion of water from

the wound bed into reactive PU can result in over-expansion of the formed scaffold or in

forming large voids in the scaffold.

Page 24: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

24

2.7. Polyurethanes modified by biologically active substances

Each group, either natural or synthetic polymers, has its advantages and disadvantages for

the use in tissue engineering. Synthetic polymer-derived materials lack cell recognition

signals and the surfaces are hydrophobic, which affect the seeding of cells. Naturally derived

polymers such as collagen have the advantage of good cell interaction and hydrophilicity, but

generally lack adequate mechanical properties and batch-to-batch consistency [76]. To

overcome the drawback of the synthetic materials, naturally occurring polymers have been

used to modify the synthetic materials. Polyurethanes have been used for a long time as

potential candidates in various fields of biomedicine. Thus, different natural polymers were

already used for their modification, such as collagen [77, 78], elastin, [79-82], hyaluronic acid

[83-86], silk fibroin [87, 88], chitosan [89-91], bioactive glasses [92-94] or nanocellulose.

The last mentioned natural polymer was used in the presented work in the form of cellulose

nanocrystals, and therefore is reviewed more into detail in the following section.

Nanocellulose - general properties 2.7.1.

As natural nanoscaled material, nanocellulose provides unique properties different from

other nanomaterials, including special morphology and geometrical dimensions, crystallinity,

liquid crystalline behavior, alignment and orientation, mechanical reinforcement,

biocompatibility, biodegradability and lack of toxicity [95]. Nanocellulose can be divided into

three different types such as cellulose nanocrystals (CNC), cellulose nanofibrils (CNF) and

bacterial cellulose (BC) also called microbial cellulose [96].

Cellulose nanocrystals with other designations such as nanocrystalline cellulose, cellulose

(nano) whiskers or rod-like cellulose microcrystals are nanoparticles with diameter in a range

of 5–30 nm, and length between 100–500 nm derived from plant cellulose, or length of 100

nm to several micrometers from tunicate and algae celluloses. Acid hydrolysis is commonly

performed for the extraction of CNC through the removal of amorphous regions and

preservation of highly-crystalline structure. Scheme of their structure is shown in Figure 7.

Page 25: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

25

O

C1

C2

C3

C4C5

O

C4OHOH

C6OH

C5

C3

C2

C1

O

O

C6OH

OHOH

n

1 4

cellulose chainsdiordered region

crystalline regions

cellulose nanocrystals

100 nm

a)

b)

c)

Figure 7: Schematics of (a) single cellulose chain repeat unit, showing the directionality of

the 1 -> 4 linkage and intrachain hydrogen bonding (dotted line), (b) idealized cellulose

microfibril showing one of the suggested configurations of the crystalline and amorphous

regions, and (c) cellulose nanocrystals after acid hydrolysis dissolved the disordered regions.

Figure depicted according to [97].

Different from rigid CNC, cellulose nanofibrils (CNF) consist of both individual and

aggregated nanofibrils made of alternating crystalline and amorphous cellulose domains,

which attributes to the morphology of CNF with soft and long chains. Due to the

entanglement of long cellulosic chains, it is not so easy to determine the length of CNF with

microscopic techniques. Therefore, only the information of fibril width for CNF is provided

in the studies, which varies from 10 to 100 nm depending on the source of cellulose,

defibrillation process and pretreatment. Regarding the preparation of CNF, mechanically

induced destruction processes are mainly applied, which involves high-pressure

homogenization and grinding before or after chemical or enzymatic treatment. Multiple

mechanical shearing effectively delaminate individual microfibrils from cellulosic fibers.

Contrary to CNC and CNF, bacterial cellulose (BC) is produced by biosynthesis typically

by bacteria such as Acetobacter xylinum [98]. The major advantage is that the product is in a

pure form and doesn’t require removing of unwanted impurities or contaminants such as

lignin, pectin and hemicellulose. During the biosynthesis of BC, the glucose chains are

produced inside the bacterial body and extruded out through pores present on the cell

envelope. With the combination of glucose chains, microfibrils are formed and further

Page 26: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

26

aggregate as ribbons (nanofibers). These ribbons subsequently generate a web-shaped

network structure with cellulosic fibers (BC), which has a diameter of 20–100 nm with

different types of nanofiber networks.

Preparation of CNC 2.7.2.

Considerable academic and industrial research effort have been devoted to developing

cellulose nanocrystals and nanofibers. The reasons for this are e.g. that cellulose is one of the

most abundant polymeric material in nature, is available everywhere, can be obtained from

different resources, is biodegradable and can be modified to generate whiskers of high aspect

ratio, high modulus and low density [99]. To obtain these nanofibers, cellulose must be

separated from other components of the plant and exposed to different treatments that

decompose its hierarchical structure down to nanoparticles. One frequently reported method is

the acidic hydrolysis of cellulose, which produces cellulose nanocrystals with diameter of 5–

20 nm and length ranging from 100 nm to approximately 500 nm, depending on the cellulose

source and the preparation process [100, 101] Although the high OH concentration on the

surface of the crystals suggests high attraction between them, the use of sulfuric acid during

hydrolysis is known to leave sulfate groups on the crystal surface, which produce repulsion

and help to produce stable suspensions in aqueous media and highly polar solvents [102].

The surface hydroxyl groups on the cellulose can be reacted to produce surface-modified

cellulose crystals [101]. Coreaction with a polymerizable mixture has also been reported, so

that the nanofibers became covalently attached to the polymer [102].

Nanocellulose-polyurethane composites 2.7.3.

A couple of systems suggesting PU-CNC composites have been introduced in the

literature. Those are usually using CNC dispersed in N,N-dimethylformamide (DMF) and PU

matrix dispersed in the same solvent. PU nanocomposite films are then prepared by casting

the mixture of those two components into the mold and after removing the solvent the films

are obtained with thickness ranging from 0.1 mm to 0.5 mm.

Pei et al. [18] observed a strong reinforcement effect by adding CNC into PU matrix made

of 4,4’-diphenylmethane diisocyanate (MDI) and 1.4-butanediol as hard segment and

poly(tetramethylene glycol) as soft segment. CNC were grafted by MDI before the synthesis

proceeded. With only 1 wt.% of cellulose nanocrystals incorporated, an 8-fold increase in

tensile strength and 1.3-fold increase in strain-to-failure were achieved. It is unusual to

improve the modulus while at the same time significantly improve the strength and toughness.

Such enhancement of mechanical properties was explained by CNC reinforcement in the hard

domains and also by increased effective cross-link density of the elastomer network due to

CNC-PU molecular interaction. Preferential reinforcement of CNC in the hard microdomains

rather than in the soft segments of PU avoided the undesired stiffening of the soft domain and

maintained the large strain-to-failure.

Page 27: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

27

The similar strategy with different PU system performed Rueda et al. [19]. The surface of

CNC was grafted by hexamethylene diisocyanate (HDI) and the PU nanocomposite was

processed via similar solvent casting procedure as Pei et al. Successful grafting was

confirmed by solid state NMR and it was claimed that half of the C6-OH groups in glucose

units were grafted by HDI. Comparison between nanocomposite material with unmodified

CNC and HDI modified CNC, both in 1.5 wt %, showed more tough material when used

unmodified CNC without impeding the extension of the soft segment and provoked a

positively impact on the mechanical properties. The use of HDI modified CNC resulted in

preferential interaction of HDI anchored chains with hard segment which provided fairly good

Young’s modulus however showed decrease to more than half of values in ductility and strain

at break. The same group published later the cell evaluation tests of those PU nanocomposites

[103]. Cells seeded on top of the materials showed that L-929 fibroblasts colonized the

materials surface giving rise to good substrates for cell adhesion and proliferation.

Saralegi et al. [104] used renewable resources, such as castor oil based highly crystalline

polyol and corn-sugar based chain extender. CNC PU nanocomposites were prepared where

PU matrix was partially crystalline. They observed increase in Young’s modulus and yield

strength by increasing CNC content and at the same time decrease in strain at break and

tensile strength. Such behavior of the material was explained by CNCs influencing the

mobility of amorphous segments. However due to high crystalline nature of both soft and

hard segments, that also served as reinforcing agents, the effect of the addition of CNC on the

final properties was less pronounced.

Nanocellulose in medicine 2.7.4.

One of the targeting outcomes of that dissertation work is utilizing the materials for further

use in medicine. Nanocellulose as a unique natural material has gained much attention for its

use in medicine. The studies of blood vessel replacement are the most developed reaching the

stage of clinical research as various systems have been already tested on animals [105].

Regarding other studies on nanocellulose e.g. for soft tissue replacement and regeneration,

most reports are still in the fundamental stage, and mainly focus on the comparison of

different properties between nanocellulose-based materials and real organs. Due to the most

developed section of vessel replacement studies some of them are described more into detail

below.

One of the most common treatments of cardiovascular disease is the graft surgery, which is

performed to supply blood to the damaged tissue (e.g. heart) with a suitable blood vessel

replacement. Biosynthesized BC has already been processed by researchers to form artificial

vascular substitutes. The team of Dieter Klemm (University Jena and Polymer Jena,

Germany) was the first research organization to investigate and apply artificial vascular

substitute obtained from BC. They have evolved a clinical product named BActerial

SYnthesized Cellulose (BASYC®) with high mechanical strength in wet state, enormous

Page 28: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

28

water retention property and low roughness of inner tube surface [105]. In comparison with

conventional synthetic vascular graft materials, e.g. polyester (Dacron®) or ePTFE,

biosynthetic BC tubes can be suitable for small diameter (<4 mm) vascular conduits. It has

been reported that BASYC from BC has been successfully used as the artificial blood vessel

in rats and pigs for microsurgery [106].

Different from BC biosynthetic procedure, it is impossible to directly fabricate the tubes

from CNC and CNF. Therefore, the development of CNC or CNF-based blood vessel

replacement commonly includes the use of a matrix material. Novel biomaterials from

polyurethane reinforced with CNF have been reported to be potentially used as vascular

replacement [107]. The presence of CNF in polyurethane improved the elastic properties of

the material, coupled with low thrombogenicity and improved physical and mechanical

properties. CNF/polyurethane biomaterials, with a wall thickness of 0.7–1.0 mm, were

applied as vascular prostheses between the brachiocephalic trunk and the right common

carotid artery in a 26-year-old male patient with multiple endocrine neoplasia. No further

effect of this CNF/polyurethane biomaterial in clinical study was reported yet.

Page 29: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

29

3. MAIN AIMS

The dissertation thesis is focused on synthesis of biodegradable polyurethane elastomers

and their modification by biologically active cellulose nanocrystals. Firstly, the PU material

was synthesized with tailorable mechanical and biodegradable properties. For preparation of

PU elastomers were chosen hydrophilic poly(ethylene glycol) and hydrophobic

poly(caprolactone) as diols, aliphatic hexamethylene diisocyanate as hard segment and

stannous octoate as catalyst. Characterization of synthesized materials was evaluated from the

viewpoint of changing the diol ratios (hydrophilic vs. hydrophobic nature) and changing the

crosslink density (ratio between NCO/OH groups). Both parameters possess ability to tailor

the physico-chemical properties for potential application in medicine.

Further modification by cellulose nanocrystals (CNCs) was performed to study the

nanofiller influence on material behavior and also for possible improvement of the

biocompatibility. CNCs were used either untreated or surface modified by polyethylene

glycol.

Proposed dissertation thesis can be summarized in following highlights:

solvent free synthesis of biodegradable PU material

interpretation of PU behavior with different PEG/PCL ratio

tailoring the properties of material by changing crosslink density (NCO/OH ratio)

solvent free synthesis of PU nanocomposite

structural investigation of the nanocomposite’s behavior

Page 30: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

30

4. EXPERIMENTAL PART

4.1. Chemicals

Text is not available in the public version.

4.2. Solvent free synthesis of bio-PU

Text is not available in the public version.

4.3. Synthesis of bio-PU nanocomposites

Text is not available in the public version.

4.4. Characterization

UV-VIS 4.4.1.

Absorbance spectra were obtained on Jasco V-630 UV-VIS spectrophotometer in the range

of wavelengths from 240 to 1100 nm with the resolution of 0.5 nm.

Attenuated total reflection infrared spectroscopy (ATR-IR) 4.4.2.

Attenuated total reflection infrared spectroscopy (ATR-IR) was conducted on Fourier-

transform infrared spectrometer (Bruker Tensor 27, USA) equipped with the germanium

crystal for ATR over the spectral range from 4000 to 600 cm-1

at the resolution of 4 cm-1

and

32 scans.

Thermogravimetric analysis (TGA) 4.4.3.

Thermal decompositions were investigated by performing TGA (TA Q500, USA) with

sample mass in the range of 5–10 mg. All measurements were carried out in the nitrogen

atmosphere with the nitrogen flow rate of 100 ml·min-1

. Heating rate was set as 10 °C·min-1

starting from 25 °C up to 750 °C.

Differential scanning calorimetry (DSC) 4.4.4.

The calorimetric measurement was carried out utilizing DSC (Netzsch 204 F1, Germany)

in the nitrogen atmosphere. Each sample (5–10 mg) was cooled with the rate of 5 °C·min-1

and heated with heating rate of 10 °C min-1

. Raw data were processed using the NETZSCH

Proteus® Software to obtain glass transition temperature (Tg), melting temperature (Tm) and

melting enthalpy (Hm). The crystallinity (c) was calculated from the first DSC heating run

according to the following equation:

c (%) = (∆𝐻𝑚 × 100)/(𝑤 × ∆𝐻𝑚0 ) (1)

where ∆𝐻𝑚 is heat of fusion of the bio-PU sample, ∆𝐻𝑚0 is heat of fusion of 100 % crystalline

PCL (135.44 J·g-1

) [108] and w is the PCL’s weight fraction in the bio-PU sample.

Page 31: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

31

Wide-angle X-ray scattering (WAXS) 4.4.5.

Wide-angle X-ray scattering (WAXS) was performed on the diffractometer (Rigaku

MiniFlex 600, Japan) with Cu K radiation beam ( = 1.5406 Å) operated at 40 kV and 15

mA in the scattering angle (2 range from 5° to 35° using the scan rate of 2° min-1

with step

of 0.02°. As a reference material, WAXS of a commercial PCL (Mn = 80 000 g mol-1

) was

also measured.

Swelling 4.4.6.

Swelling ratio of the 1 cm × 1 cm × 1 mm specimens cut out of the bio-PU films was

measured in ultrapure water (UPW) at laboratory temperature. The samples weight was

measured within the first hour every ten minutes, within the second hour every 20 minutes

and then every hour for the next 4 hours, with the last measurement performed after 1 day

since the beginning of the swelling experiment. Water uptake was calculated according to the

formula (2):

Water uptake (%) = (𝑤𝑠 − 𝑤𝑑) 𝑤𝑑⁄ × 100 (2)

where ws is the weight of swollen sample at the given time and wd is the weight of dry sample.

Each sample was measured 3 times.

Hydrolytic degradation 4.4.7.

Hydrolytic degradation tests employing the swelled samples were carried out in an

incubator at 37 °C in UPW for 365 days. Each measurement is an average of 3 specimens.

The specimens were removed at the given time from vials with distilled water, wiped using

filter paper and weighed to determine the weight loss. The UPW was changed every two

weeks. Mass loss was calculated according to formula (3):

Mass loss (%) = (𝑤0 − 𝑤𝑡) 𝑤0⁄ × 100 (3)

where w0 is the weight of swollen sample after 24 hours in water and wt is the weight of the

sample in the given time.

Extraction 4.4.8.

Extractions were performed using Soxhlet apparatus. Small pieces of dry films (1×1 cm

and 1 mm thick) were used for the extractions. The extractions were performed for 8 hours

with volume 200 ml of solvent, extracted samples were dried in the vacuum oven to the

constant weight. Extracts from acetone with dissolved residues were evaporated followed by

drying in the vacuum oven to be evaluated by ATR-IR. Bio-PU films with NCO/OH ratio 1.2

were extracted in acetone and in N,N-dimethylformamide (DMF). Both extractions either in

acetone or DMF were carried out via the same procedure.

Page 32: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

32

Tensile measurement 4.4.9.

Tensile tests were performed employing the tensile tester (Zwick Roell Z010, Germany)

according to the ISO 527 standard. Dog-bone specimens obtained according to ISO 527-2/5B

were 35 mm in length, 2 mm in width in the middle part, 1 mm thick and gauge length was

10 mm. The 500 N load cell was used for the measurement with the cross-head speed of

10 mm·min-1

corresponding to the 100 %·min-1

deformation rate. The 0.1 N preload was used

and all tensile measurements were performed at the laboratory temperature. Each sample was

measured at least 5 times and data were averaged to get the standard deviation.

Dynamic light scattering 4.4.10.

Dynamic light scattering (DLS, DynaPro Nanostar, Wyatt) using digital autocorrelator and

laser with wavelength of 658 nm was performed to qualitatively prove the size distribution of

nanoparticles. DLS was measured from a dilute suspension of either CNC or CNC-PEG

nanoparticles.

Dynamic mechanical analysis (DMA) 4.4.11.

Dynamic mechanical analysis (DMA) was measured on a TA Instruments Q800 in tension

mode with frequency 1 Hz and the temperature scanning rate 3 °C·min-1

. Samples with

thickness of 1 mm were cut to stripes with dimensions of 4 mm width and 15 mm length.

Transmision electron microscopy (TEM) 4.4.12.

Transmission electron microscopy (TEM) was carried out using Tecnai G2 Spirit Twin 12

microscope (FEI, Czech Republic) Thin sections (∼60 nm) were cut at –80 °C by an

ultramicrotome (Ultracut UCT, Leica, Germany) under cryogenic conditions (the sample and

the knife temperatures were –85 °C and –50 °C, respectively). The ultrathin sections were

transferred to a microscopic grid and observed in the bright field mode at the acceleration

voltage of 120 kV.

Cytotoxicity evaluation 4.4.13.

Chosen samples were tested on cytotoxicity by seeding mesenchymal stem cells (MSCs)

on bio-PU films and evaluation of spreading and proliferation after 24, 72 and 168 hours.

Chosen bio-PU or PU nanocomposite films with a diameter of 6 mm and a thickness of

1 mm were sterilized using combination of UV for 15 min and ethanol. Before seeding with

MSCs, the films were incubated in culture medium (Dulbecco's Modified Eagle's medium

with L-glutamine, 10% fetal bovine serum, 1% penicillin/streptomycin) overnight at 37 °C.

Cells were seeded on PU films at a density of 3·104 cells·cm

-2 in 48-well plate. PU films with

seeded MSCs were cultivated at 37 °C and 5% CO2 atmosphere and cell viability was

acquired in 24, 72 and 168 hours after seeding. The medium was changed every 3 days.

Fluorescence microscopy and live/dead staining (calcein-AM/ propidium iodide) were used to

Page 33: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

33

determine the cell viabillity. The mix of calcein-AM (2 μM) and propidium (1,5 μM) in

phosphate buffered saline (PBS) was added to films containing seeded cells and incubated for

15 min at 37 °C and 5% CO2 for live/dead cell detection. The cells were visualized using a

Zeiss Axio Imager 2 microscope.

Page 34: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

34

5. RESULTS AND DISCUSSION

Text is not available in the public version.

Page 35: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

35

6. CONCLUSIONS

New solvent free method was successfully used for the synthesis of biodegradable

polyurethanes (bio-PUs) and also for in situ synthesis of polyurethane nanocomposites filled

by either unmodified or PEG grafted cellulose nanocrystals.

Different diol ratios between PEG/PCL resulted in materials from brittle through elastic up

to tough elastomeric films. The structural analysis revealed that crystalline PCL domains

formed below the PEG/PCL weight ratio of 30/70 are responsible for abrupt increase in the

elastic modulus, stress and deformation at break by order of magnitude. These domains

reinforce the PU network and their small size allows for more uniform distribution of strain

resulting in enhanced toughness. Extraction studies confirmed crosslinked structure of bio-

PUs although only bifunctional feedstocks were used.

Variation between NCO/OH groups (isocyanate ratio) confirmed increase in crosslink

density with increasing isocyanate ratio. Higher crosslink density reduced the ability of bio-

PUs to absorb water and followly prolonged the time of hydrolytic degradation. Systems with

very low crosslinking tend to be weak and flexible, whereas polymers with high degrees of

crosslinking were more rigid.

Polyurethane nanocomposites filled by either unmodified or PEG grafted CNCs were

successfully synthesized by evolved solvent free method. Presence of nanofiller promoted

significant effect on stiffness explained by restricted motion of the rubbery PU matrix due to

the presence of stiff and rod-like cellulose nanocrystals.

Five representatives from all the samples were chosen for preliminary tests of human

mesenchymal stem cells (MSCs) response to the material. MSCs didn’t adhere on PUs with

high PEG content although the material was modified by low amount of biologically active

CNCs. PUs with high abundance of PCL allowed cell adhesion on the surface, but promoted

with timescale of one week detachment of the MSCs from the surface.

Tailored biomaterials with adjustable functional properties are desirable for many

applications ranging from drug delivery to regenerative medicine. Presented variation of PU

materials possess great range of mechanical (toughness, flexibility) and physical (swelling,

degradation) properties. Low adherence of cells to the bio-PU surface predestines the material

for the use as an artificial vein. However for interaction with a body, further modification of

the contact side is desirable by compounds favoring material attachment e.g. by RGD

proteins.

Page 36: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

36

7. REFERENCES

1. Howard, G. 2002. Biodegradation of polyurethane. International Biodeterioration, vol.

49, issue 4, pp. 245-252. DOI: 10.1016/S0964-8305(02)00051-3.

2. Santerre, J., Woodhouse, K., Laroche, G. & Labow, R.. 2005. Understanding the

biodegradation of polyurethanes. Biomaterials, vol. 26, issue 35, pp. 7457-7470. DOI:

10.1016/j.biomaterials.2005.05.079

3. Guelcher, S. 2008. Biodegradable Polyurethanes. Tissue Engineering Part B: Reviews,

vol. 14, issue 1, pp. 3-17. DOI: 10.1089/teb.2007.0133

4. Martina, M. & Hutmacher, D.. 2007. Biodegradable polymers applied in tissue

engineering research. Polymer International, vol. 56, issue 2, pp. 145-157. DOI:

10.1002/pi.2108.

5. Vermette P, Griesse HJ, Laroche G, Guidoin R, Developments in Design and Synthesis

of Biostable Polyurethanes, in Biomedical Applications of Polyurethanes, edited by

Gunatillake PA, Meijs GF, McCarthy SJ. Eurekah.com, USA, p 160-170 (2001).

6. Sundback, C. 2003. Manufacture of porous polymer nerve conduits by a novel low-

pressure injection molding process. Biomaterials, vol. 24, issue 5, pp. 819-830. DOI:

10.1016/S0142-9612(02)00409-X.

7. Ratcliffe, A. 2000. Tissue engineering of vascular grafts. Matrix Biology, vol. 19, issue

4, pp. 353-357. DOI: 10.1016/S0945-053X(00)00080-9

8. Grad, S., Kupcsik, L., Gorna, K., Gogolewski, S. & Alini, M.. 2003. The use of

biodegradable polyurethane scaffolds for cartilage tissue engineering. Biomaterials,

vol. 24, issue 28, pp. 5163-5171.DOI: 10.1016/S0142-9612(03)00462-9.

9. Gogolewski, S. & Gorna, K.. 2007. Biodegradable polyurethane cancellous bone graft

substitutes in the treatment of iliac crest defects. Journal of Biomedical Materials

Research Part A, 80A, issue 1, pp. 94-101.DOI: 10.1002/jbm.a.30834.

10. Bergmeister, H., Seyidova, N., Schreiber, C., Strobl, M., Grasl, C., Walter, I., Messner,

B., Baudis, S., Fröhlich, S., Marchetti-Deschmann, M., Griesser, M., Di Franco, M.,

Krssak, M., Liska, R. & Schima, H.. 2015. Biodegradable, thermoplastic polyurethane

grafts for small diameter vascular replacements. Acta Biomaterialia, vol. 11, pp. 104-

113. DOI: 10.1016/j.actbio.2014.09.003

11. Chen, Q., Liang, S. & Thouas, G.. 2013. Elastomeric biomaterials for tissue

engineering. Progress in Polymer Science, vol. 38, 3-4, pp. 584-671. DOI:

10.1016/j.progpolymsci.2012.05.003

12. Kim, H., Kang, M., Knowles, J. & Gong, M.. 2014-08-18. Synthesis of highly elastic

biocompatible polyurethanes based on bio-based isosorbide and poly(tetramethylene

glycol) and their properties. Journal of Biomaterials Applications, vol. 29, issue 3, pp.

454-464. DOI: 10.1177/0885328214533737

13. Hassan, M., Mauritz, K., Storey, R. & Wiggins, J.. 2006-05-01. Biodegradable aliphatic

thermoplastic polyurethane based on poly(ɛ-caprolactone) andL-lysine diisocyanate.

Journal of Polymer Science Part A: Polymer Chemistry, vol. 44, issue 9, pp. 2990-

3000. DOI: 10.1002/pola.21373

Page 37: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

37

14. Cometa, S., Bartolozzi, I., Corti, A., Chiellini, F., De Giglio, E. & Chiellini, E.. 2010.

Hydrolytic and microbial degradation of multi-block polyurethanes based on poly(ɛ-

caprolactone)/poly(ethylene glycol) segments. Polymer Degradation and Stability,

vol. 95, issue 10, pp. 2013-2021. DOI: 10.1016/j.polymdegradstab.2010.07.007

15. Mondal, S. & Martin, D.. 2012. Hydrolytic degradation of segmented polyurethane

copolymers for biomedical applications. Polymer Degradation and Stability, vol. 97,

issue 8, pp. 1553-1561.DOI: 10.1016/j.polymdegradstab.2012.04.008

16. Sartori, S., Boffito, M., Serafini, P., Caporale, A., Silvestri, A., Bernardi, E.,

Boccafoschi, F. & Ciardelli, G.. 2013. Synthesis and structure–property relationship of

polyester-urethanes and their evaluation for the regeneration of contractile tissues.

Reactive and Functional Polymers, vol. 73, issue 10, pp. 1366-1376.DOI:

10.1016/j.reactfunctpolym.2013.01.006

17. Chan-Chan, L., Solis-Correa, R., Vargas-Coronado, R., Cervantes-Uc, J., Cauich-

Rodríguez, J., Quintana, P. & Bartolo-Pérez, P.. 2010. Degradation studies on

segmented polyurethanes prepared with HMDI, PCL and different chain extenders.

Acta Biomaterialia, vol. 6, issue 6, pp. 2035-2044. DOI: 10.1016/j.actbio.2009.12.010

18. Pei, A., Malho, J., Ruokolainen, J., Zhou, Q. & Berglund, L.. 2011-06-14. Strong

Nanocomposite Reinforcement Effects in Polyurethane Elastomer with Low Volume

Fraction of Cellulose Nanocrystals. Macromolecules, vol. 44, issue 11, pp. 4422-4427.

DOI: 10.1021/ma200318k

19. Rueda, L., Fernández d’Arlas, B., Zhou, Q., Berglund, L., Corcuera, M., Mondragon, I.

& Eceiza, A.. 2011. Isocyanate-rich cellulose nanocrystals and their selective insertion

in elastomeric polyurethane. Composites Science and Technology, vol. 71, issue 16,

pp. 1953-1960. DOI: 10.1016/j.compscitech.2011.09.014

20. Oertell, Günter. Polyurethane Handbook. 2nd edition. Munich: Hanser Publishers,

1994. 688 p. ISBN: 3-446-17198-3

21. Vert, M., Li, S., Spenlehauer, G. & Guerin, P.. 1992. Bioresorbability and

biocompatibility of aliphatic polyesters. Journal of Materials Science: Materials in

Medicine, vol. 3, issue 6, pp. 432-446. DOI: 10.1007/BF00701240

22. Ilavský, M., Ulmer, H., Te Nijenhuis, K. & Mijs, W.. 2001. Network formation in

polyurethanes based on triisocyanate and diethanolamine derivatives. European

Polymer Journal, vol. 37, issue 5, pp. 887-896. DOI: 10.1016/S0014-3057(00)00217-2

23. Ward, R. & Jones, R.. 2011. Polyurethanes and Silicone Polyurethane Copolymers.

Comprehensive Biomaterials, pp. 431. DOI: 10.1016/B978-0-08-055294-1.00272-5.

24. Dostal M, Vasku J, Sotolova O, Vasku A, Dolezel S, Hartmannova B. 1990.

Mineralization of polyurethane membranes in the total artificial-heart (THA) – a

retrospective study from long-term animal-experiments. International Journal of

Artificial Organs. vol. 13, pp. 498-502.

25. Marois Y, Paris E, Zhang Z, Doillon CJ, King MW, Guidoin RG. 1996. Vascugraft(R)

microporous polyester urethane arterial prosthesis as a thoraco-abdominal bypass in

dogs. Biomaterials, vol. 17, pp. 1289-300.

26. Marois Y, Chakfe N, Guidoin R, Duhamel RC, Roy R, Marois M, King MW. 1996.

Douville Y. An albumin-coated polyester arterial graft: in vivo assessment of

biocompatibility and healing characteristics. Biomaterials, vol. 17, pp. 3–14.

Page 38: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

38

27. Samoexpandovatelný biodegradabilní stent připravený z radiopacitního vlákna potažený

biodegradabilní fólií a léčivem a způsob jeho výroba. Patent, CZ 303231 B6.

28. Kay, I., Sabaté, M., Costa, M., Kozuma, K., Albertal, M., Ligthart, J., Coen, V.,

Levendag, P., Van der Giessen, W. & Serruys, P.. 2000. Stent and stent-edge

remodeling after conventional and radioactive stent implantation. Heart, Lung and

Circulation, vol. 9, issue 3, A163-. DOI: 10.1046/j.1443-9506.2000.08928.x

29. Bakker, D., Van Blitterswijk, C., Hesseling, S., Koerten, H., Kuijpers, W. & Grote, J..

1990. Biocompatibility of a polyether urethane, polypropylene oxide, and a polyether

polyester copolymer. A qualitative and quantitative study of three alloplastic tympanic

membrane materials in the rat middle ear. Journal of Biomedical Materials Research,

vol. 24, issue 4, pp. 489-515. DOI: 10.1002/jbm.820240407

30. Bruin P, Meeuwsen EAJ, Vanandel MV, Worst JGF, Pennings AJ. 1993. Autoclavable

highly cross-linked polyurethane networks in ophthalmology. Biomaterials, vol. 14,

issue 14, pp. 1089-1097. DOI: 10.1016/0142-9612(93)90210-S

31. Watkinson AF, Ellul J, Entwisle K, Mason RC, Adam A. 1995. Esophagealcarcinoma –

initial results of palliative treatment with covered selfexpanding endoprostheses.

Radiology, vol. 195, pp. 821–827.

32. Rossi P, Bezzi M, Salvatori FM, Panzetti C, Rossi M, Pavia G. 1997. Clinical

experience with covered wallstents for biliary malignancies: 23-month follow-up.

Cardiovascular and Interventional Radiology, vol. 20, pp. 441–447.

33. U. S. Food and Drug Administration home page. U.S. Food and Drug Administration

home page [online]. [cit. 2013-03-11]. Available from:

http://www.fda.gov/MedicalDevices/DeviceRegulationandGuidance/Overview/Classif

yYourDevice/default.htm

34. Vermette, Patrick, Griesser Hans J., Laroche Gateán and Guidoin R. Biomedical

applications of polyurethanes. Austin, Tex.: Eurekah.com, c2001. ISBN 978-1-58706-

023-6.

35. Tanzi, M., Verderio, P., Lampugnani, M., Resnati, M., Dejana, E. & Sturani, E.. 1994.

Cytotoxicity of some catalysts commonly used in the synthesis of copolymers for

biomedical use. Journal of Materials Science: Materials in Medicine, vol. 5, 6-7, pp.

393-396. DOI: 10.1007/BF00058971.

36. Fromstein JD, Woodhouse KA. Polyurethane biomaterials. In: Wnek GE, Bowlin GL,

editors. Encyclopedia of biomaterials and biomedical engineering. New York: Marcel

Dekker; 2006. p. 1–10.

37. Zhang J-Y, Beckman EJ, Hu J, Yang G-G, Agarwal S, Hollinger JO. 2002. Synthesis

biodegradability, and biocompatibility of lysine diisocyanate–glucose polymers.

Tissue Engineering, vol. 8, pp. 771-785.

38. Gisselfält, K., Edberg, B. & Flodin, P.. 2002. Synthesis and Properties of Degradable

Poly(urethane urea)s To Be Used for Ligament Reconstructions. Biomacromolecules,

vol. 3, issue 5, pp. 951-958. DOI: 10.1021/bm025535u

39. Serrano M, Chung E & Ameer G. 2010. Advances and Applications of Biodegradable

Elastomers in Regenerative Medicine. Advanced Functional Materials, vol. 20, issue

2, pp. 192-208. DOI: 10.1002/adfm.200901040

Page 39: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

39

40. Madhavan Nampoothiri, K., Nair, N. & John, R.. 2010. An overview of the recent

developments in polylactide (PLA) research. Bioresource Technology, vol. 101, issue

22, pp. 8493-8501. DOI: 10.1016/j.biortech.2010.05.092

41. Vernengo, J., Fussell, G., Smith, N. & Lowman, A.. 2010. Synthesis and

characterization of injectable bioadhesive hydrogels for nucleus pulposus replacement

and repair of the damaged intervertebral disc. Journal of Biomedical Materials

Research Part B: Applied Biomaterials, 93B, issue 2, pp. 309-317. DOI:

10.1002/jbm.b.31547

42. Harris, J. M., Poly(ethylene glycol) chemistry and biological applications. Washington,

DC: American Chemical Society, 1997. pp. 45. ISBN: 9780841216440

43. Sabir, M. I., Xu X. and Li L. A review on biodegradable polymeric materials for bone

tissue engineering applications. Journal of Materials Science. 2009, vol. 44, issue 21,

pp. 5713-5724. DOI: 10.1007/s10853-009-3770-7.

44. Wang, S., Cui, W. & Bei, J.. 2005. Bulk and surface modifications of polylactide.

Analytical and Bioanalytical Chemistry, vol. 381, issue 3, pp. 547-556. DOI:

10.1007/s00216-004-2771-2

45. BASF - product information the chemical catalog - Pluronic. [online]. [cit. 2015-03-23].

Available on: http://worldaccount.basf.com/wa/NAFTA/Catalog/ChemicalsNAFTA/

pi/BASF/Brand/pluronic.

46. Gorna, K. & Gogolewski, S.. 2002. In vitro degradation of novel medical biodegradable

aliphatic polyurethanes based on ϵ-caprolactone and Pluronics® with various

hydrophilicities. Polymer Degradation and Stability, vol. 75, issue 1, pp. 113-122.

DOI: 10.1016/S0141-3910(01)00210-5.

47. Woodruff, M. & Hutmacher, D.. 2010. The return of a forgotten polymer—

Polycaprolactone in the 21st century. Progress in Polymer Science, vol. 35, issue 10,

pp. 1217-1256. DOI: 10.1016/j.progpolymsci.2010.04.002

48. Teoh, H. S., Hutmacher, D. W., Tan, K. C., Tam, K. F. and Zein I.: Methods for

fabricating a filament for use in tissue engineering, US Patent 6730252, 2001, 1–35.

49. Shor, L., Güçeri, S., Wen, X., Gandhi, M. & Sun, W.. 2007. Fabrication of three-

dimensional polycaprolactone/hydroxyapatite tissue scaffolds and osteoblast-scaffold

interactions in vitro. Biomaterials, vol. 28, issue 35, pp. 5291-5297. DOI:

10.1016/j.biomaterials.2007.08.018.

50. Gilding, D. & Reed, A.. 1979. Biodegradable polymers for use in surgery—

polyglycolic/poly(actic acid) homo- and copolymers. Polymer, vol. 20, issue 12, pp.

1459-1464. DOI: 10.1016/0032-3861(79)90009-0

51. Ashammakhi, N. & Rokkanen, P.. 1997. Absorbable polyglycolide devices in trauma

and bone surgery. Biomaterials, vol. 18, issue 1, pp. 3-9. DOI: 10.1016/S0142-

9612(96)00107-X

52. Andreopoulos, AG, Hatzi, E, Doxastakis, M. 1999. Synthesis and properties of

poly(lactic acid). Journal of materials science: Materials in medicine, vol. 10, pp. 9–

33.

53. Kim TK, Yoon JJ, Lee DS, Park TG. 2006. Gas foamed open porous biodegradable

polymeric microspheres. Biomaterials, vol. 27, issue 2, pp. 152-159. DOI:

10.1016/j.biomaterials.2005.05.081

Page 40: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

40

54. Ueda, H. & Tabata, Y.. 2003. Polyhydroxyalkanonate derivatives in current clinical

applications and trials. Advanced Drug Delivery Reviews, vol. 55, issue 4. DOI:

10.1016/S0169-409X(03)00037-1

55. Flickinger MC, Drew SW. Encyclopedia of Bioprocess Technology - Fermentation,

Biocatalysis, and Bioseparation, John Wiley & Sons, 1999. Volumes 1-5. (pp. 2024 -

2048). Online version available at: http://knovel.com.

56. Defoirdt, T., Boon, N., Sorgeloos, P., Verstraete, W. & Bossier, P.. 2009. Short-chain

fatty acids and poly-β-hydroxyalkanoates. Biotechnology Advances, vol. 27, issue 6,

pp. 680-685. DOI: 10.1016/j.biotechadv.2009.04.026

57. Brzeska, J., Dacko, P., Gębarowska, K., Janik, H., Kaczmarczyk, B., Kasperczyk, J.,

Kowalczuk, M. & Rutkowska, M.. 2012-09-15. The structure of novel polyurethanes

containing synthetic poly[(R,S)-3-hydroxybutyrate]. Journal of Applied Polymer

Science, vol. 125, issue 6, pp. 4285-4291. DOI: 10.1002/app.36599.

58. Hinrichs WLJ, Kuit J, Feil H, Wildevuur CRH, Feijen J. 1992. In vivo fragmentation of

microporous polyurethane-based and copolyesterether elastomer-based vascular

prostheses. Biomaterials, vol. 13, issue 9, pp. 585-593. DOI: 10.1016/0142-

9612(92)90026-K

59. Fromstein, J. & Woodhouse, K.. 2002. Elastomeric biodegradable polyurethane blends

for soft tissue applications. Journal of Biomaterials Science, Polymer Edition, vol. 13,

issue 4, pp. 391-406. DOI: 10.1163/156856202320253929

60. Guan, J., Fujimoto, K., Sacks, M. & Wagner, W.. 2005. Preparation and

characterization of highly porous, biodegradable polyurethane scaffolds for soft tissue

applications. Biomaterials, vol. 26, issue 18, pp. 3961-3971. DOI:

10.1016/j.biomaterials.2004.10.018

61. Elliott, S., Fromstein, J., P.Santerre, J. & Woodhouse, K.. 2002. Identification of

biodegradation products formed by L-phenylalanine based segmented

polyurethaneureas. Journal of Biomaterials Science, Polymer Edition, vol. 13, issue 6,

pp. 691-711. DOI: 10.1163/156856202320269166

62. Guan, J. & Wagner, W.. 2005. Synthesis, Characterization and Cytocompatibility of

Polyurethaneurea Elastomers with Designed Elastase Sensitivity. Biomacromolecules,

vol. 6, issue 5, pp. 2833-2842. DOI: 10.1021/bm0503322

63. Hafeman, A., Zienkiewicz, K., Zachman, A., Sung, H., Nanney, L., Davidson, J. &

Guelcher, S.. 2011. Characterization of the degradation mechanisms of lysine-derived

aliphatic poly(ester urethane) scaffolds. Biomaterials, vol. 32, issue 2, pp. 419-429.

DOI: 10.1016/j.biomaterials.2010.08.108

64. Skarja, G. & Woodhouse, K.. 1998. Synthesis and characterization of degradable

polyurethane elastomers containing an amino acid-based chain extender. Journal of

Biomaterials Science, Polymer Edition, vol. 9, issue 3, pp. 271-295. DOI:

10.1163/156856298X00659

65. Collins, M. & Birkinshaw, C.. 2013. Hyaluronic acid based scaffolds for tissue

engineering—A review. Carbohydrate Polymers, vol. 92, issue 2, pp. 1262-1279.

DOI: 10.1016/j.carbpol.2012.10.028.

66. Liu, Y., Lim, J. & Teoh, S.. 2013. Review. Biotechnology Advances, vol. 31, issue 5,

pp. 688-705. DOI: 10.1016/j.biotechadv.2012.10.003.

Page 41: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

41

67. Grenier, S., Sandig, M. & Mequanint, K.. 2007-09-15. Polyurethane biomaterials for

fabricating 3D porous scaffolds and supporting vascular cells. Journal of Biomedical

Materials Research Part A, 82A, issue 4, pp. 802-809.DOI: 10.1002/jbm.a.31194.

68. The Polymer Technology Group. [online]. [cit. 2013-03-13]. Available from:

http://www.aestheticscientist.com/PTG/BMP/BMP_index.html

69. Sharifpoor, S., Labow, R. & Santerre. 2009-10-12. Synthesis and Characterization of

Degradable Polar Hydrophobic Ionic Polyurethane Scaffolds for Vascular Tissue

Engineering Applications. Biomacromolecules, vol. 10, issue 10, pp. 2729-2739.

DOI: 10.1021/bm9004194.

70. Temenoff, J. & Mikos, A.. 2000. Injectable biodegradable materials for orthopedic

tissue engineering. Biomaterials, vol. 21, issue 23, pp. 2405-2412. DOI:

10.1016/S0142-9612(00)00108-3

71. Cohn, D., Lando, G., Sosnik, A., Garty, S. & Levi, A.. 2006. PEO–PPO–PEO-based

poly(ether ester urethane)s as degradable reverse thermo-responsive multiblock

copolymers. Biomaterials, vol. 27, issue 9, pp. 1718-1727. DOI:

10.1016/j.biomaterials.2005.10.035

72. Guelcher, S., Srinivasan, A., Dumas, J., Didier, J., McBride, S. & Hollinger, J.. 2008.

Synthesis, mechanical properties, biocompatibility, and biodegradation of

polyurethane networks from lysine polyisocyanates. Biomaterials, vol. 29, issue 12,

pp. 1762-1775. DOI: 10.1016/j.biomaterials.2007.12.046

73. Biodegradable polyurethane/urea compositions, United States Patent 8343472

74. Adhikari, R., Gunatillake, P., Griffiths, I., Tatai, L., Wickramaratna, M., Houshyar, S.,

Moore, T., Mayadunne, R., Field, J., McGee, M. & Carbone, T.. 2008. Biodegradable

injectable polyurethanes. Biomaterials, vol. 29, issue 28, pp. 3762-3770. DOI:

10.1016/j.biomaterials.2008.06.021

75. Mishra, P., Panwar, H., Bhargava, A., Gorantla, V., Jain, S., Banerjee, S. & Maudar, K..

2008. Isocyanates induces DNA damage, apoptosis, oxidative stress, and inflammation

in cultured human lymphocytes. Journal of Biochemical and Molecular Toxicology,

vol. 22, issue 6, pp. 429-440. DOI: 10.1002/jbt.20260

76. Alperin, C., Zandstra, P. & Woodhouse, K.. 2005. Polyurethane films seeded with

embryonic stem cell-derived cardiomyocytes for use in cardiac tissue engineering

applications. Biomaterials, vol. 26, issue 35, pp. 7377-7386. DOI:

10.1016/j.biomaterials.2005.05.064

77. Meyers, Marc André; Chawla, Krishan Kumar. Mechanical Behavior of Materials (2nd

Edition). Cambridge University Press, 2009. Online version available at:

http://www.knovel.com.

78. Sin, D., Miao, X., Liu, G., Wei, F., Chadwick, G., Yan, C. & Friis, T.. 2010.

Polyurethane (PU) scaffolds prepared by solvent casting/particulate leaching (SCPL)

combined with centrifugation. Materials Science and Engineering: C, vol. 30, issue 1,

pp. 78-85. DOI: 10.1016/j.msec.2009.09.002.

79. Mithieux SM, Rasko JEJ, Weiss ASAS. 2004. Synthetic elastin hydrogels derived from

massive elastic assemblies of self-organized human protein monomers. Biomaterials,

vol. 25, pp. 4921–7.

Page 42: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

42

80. Gosline, J., Lillie, M., Carrington, E., Guerette, P., Ortlepp, C. & Savage, K.. 2002-02-

28. Elastic proteins. Philosophical Transactions of the Royal Society B: Biological

Sciences, vol. 357, issue 1418, pp. 121-132. DOI: 10.1098/rstb.2001.1022

81. W.F. Daamen, J.H. Veerkamp, J.C.M. van Hest, T.H. van Kuppevelt. 2007. Elastin as a

biomaterial for tissue engineering. Biomaterials, vol. 28, issue 30, pp. 4378-4398.

DOI: 10.1016/j.biomaterials.2007.06.025.

82. Blit, P., Battiston, K., Yang, M., Paul Santerre, J. & Woodhouse, K.. 2012. Electrospun

elastin-like polypeptide enriched polyurethanes and their interactions with vascular

smooth muscle cells. Acta Biomaterialia, vol. 8, issue 7, pp. 2493-2503. DOI:

10.1016/j.actbio.2012.03.032.

83. Wang, T.-W., Wu, H.-C., Huang, Y.-C., Sun, J.-S., Lin, F.-H. 2006. Biomimetic

bilayered gelatin-chondroitin 6 sulfate-hyaluronic acid biopolymer as a scaffold for

skin equivalent tissue engineering. Artificial Organs, vol. 30 (3), pp. 141-149.

84. F. Duranti, G. Salti, B. Bovani, M. Calandra, M.L. Rosati. 1998. Injectable hyaluronic

acid gel for soft-tissue augmentation. A clinical and histological study. Dermatologic

Surgery, vol. 24, pp. 1317–1325.

85. Price, R., Myers, S., Leigh, I. & Navsaria, H.. 2005. The Role of Hyaluronic Acid in

Wound Healing. American Journal of Clinical Dermatology, vol. 6, issue 6, pp. 393-

402. DOI: 10.2165/00128071-200506060-00006

86. Xu, F., Nacker, J., Crone, W. & Masters, K.. 2008. The haemocompatibility of

polyurethane–hyaluronic acid copolymers. Biomaterials, vol. 29, issue 2, pp. 150-160.

DOI: 10.1016/j.biomaterials.2007.09.028.

87. Kundu, B., Rajkhowa, R., Kundu, S. & Wang, X.. 2013. Silk fibroin biomaterials for

tissue regenerations. Advanced Drug Delivery Reviews, vol. 65, issue 4, pp. 457-470.

DOI: 10.1016/j.addr.2012.09.043.

88. Hu, J., Chen, B., Guo, F., Du, J., Gu, P., Lin, X., Yang, W., Zhang, H., Lu, M., Huang,

Y. & Xu, G.. 2012. Injectable silk fibroin/polyurethane composite hydrogel for

nucleus pulposus replacement. Journal of Materials Science: Materials in Medicine,

vol. 23, issue 3, pp. 711-722. DOI: 10.1007/s10856-011-4533-y.

89. Dash, M., Chiellini, F., Ottenbrite, R. & Chiellini, E.. 2011. Chitosan—A versatile

semi-synthetic polymer in biomedical applications. Progress in Polymer Science, vol.

36, issue 8, pp. 981-1014. DOI: 10.1016/j.progpolymsci.2011.02.001.

90. Wang, Y., Hong, Q., Chen, Y., Lian, X. & Xiong, Y.. 2012. Surface properties of

polyurethanes modified by bioactive polysaccharide-based polyelectrolyte multilayers.

Colloids and Surfaces B: Biointerfaces, vol. 100, pp. 77-83. DOI:

10.1016/j.colsurfb.2012.05.030.

91. Aoyagi, S., Onishi, H. & Machida, Y.. 2007. Novel chitosan wound dressing loaded

with minocycline for the treatment of severe burn wounds. International Journal of

Pharmaceutics, vol. 330, 1-2, pp. 138-145. DOI: 10.1016/j.ijpharm.2006.09.016.

92. Ryszkowska, J., Auguścik, M., Sheikh, A. & Boccaccini, A.. 2010. Biodegradable

polyurethane composite scaffolds containing Bioglass® for bone tissue engineering.

Composites Science and Technology, vol. 70, issue 13, pp. 1894-1908. DOI:

10.1016/j.compscitech.2010.05.011.

Page 43: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

43

93. Khan, A.S., Z. Ahmed, M.J. Edirisinghe, F.S.L. Wong, & I.U. Rehman. 2008.

Preparation and characterization of a novel bioactive restorative composite based on

covalently coupled polyurethane�nanohydroxyapatite fibres. Acta Biomaterialia, vol.

4, (5), pp. 1275-1287. DOI: 10.1016/j.actbio.2008.04.016.

94. Dumas, J., Davis, T., Holt, G., Yoshii, T., Perrien, D., Nyman, J., Boyce, T. &

Guelcher, S.. 2010. Synthesis, characterization, and remodeling of weight-bearing

allograft bone/polyurethane composites in the rabbit. Acta Biomaterialia, vol. 6, issue

7, pp. 2394-2406. DOI: 10.1016/j.actbio.2010.01.030.

95. Klemm, D., Kramer, F., Moritz, S., Lindström, T., Ankerfors, M., Gray, D. & Dorris,

A.. 2011. Nanocelluloses: A New Family of Nature Based Materials. Angewandte

Chemie International Edition, vol. 50, issue 24, pp. 5438-5466. DOI:

10.1002/anie.201001273

96. Lin, N., Huang, J. & Dufresne, A.. 2012. Preparation, properties and applications of

polysaccharide nanocrystals in advanced functional nanomaterials. Nanoscale, vol. 4,

issue 11, pp. 3274-3294. DOI: 10.1039/c2nr30260h

97. Moon, R., Martini, A., Nairn, J., Simonsen, J. & Youngblood, J.. 2011. Cellulose

nanomaterials review. Chemical Society Reviews, vol. 40, issue 7, pp. 3941-3994.

DOI: 10.1039/c0cs00108b

98. Lin, S., Loira Calvar, I., Catchmark, J., Liu, J., Demirci, A. & Cheng, K.. 2013.

Biosynthesis, production and applications of bacterial cellulose. Cellulose, vol. 20,

issue 5, pp. 2191-2219. DOI: 10.1007/s10570-013-9994-3

99. Eichhorn, S., Dufresne, A., Aranguren, M., Marcovich, N., Capadona, J., Rowan, S.,

Weder, C., Thielemans, W., Roman, M., Renneckar, S., Gindl, W., Veigel, S., Keckes,

J., Yano, H., Abe, K., Nogi, M., Nakagaito, A., Mangalam, A., Simonsen, J., Benight,

A., Bismarck, A., Berglund, L. & Peijs, T.. 2010. Review. Journal of Materials

Science, vol. 45, issue 1, pp. 1-33. DOI: 10.1007/s10853-009-3874-0

100. Dong, X.M., Revol, J.-F., Gray, D.G. 1998. Effect of microcrystallite preparation

conditions on the formation of colloid crystals of cellulose. Cellulose, vol. 5, pp. 19-

32.

101. Habibi, Y., Lucia, L. & Rojas, O.. 2010. Cellulose Nanocrystals. Chemical Reviews,

vol. 110, issue 6, pp. 3479-3500. DOI: 10.1021/cr900339w

102. Marcovich, N., Auad, M., Bellesi, N., Nutt, S. & Aranguren, M.. 2006. Cellulose

micro/nanocrystals reinforced polyurethane. Journal of Materials Research, vol. 21,

issue 04, pp. 870-881. DOI: 10.1557/jmr.2006.0105

103. Rueda, L., A. Saralegi, B. Fernández-d’Arlas, Q. Zhou, A. Alonso-Varona, L. A.

Berglund, I. Mondragon, M. A. Corcuera, and A. Eceiza. 2013. In situ polymerization

and characterization of elastomeric polyurethane-cellulose nanocrystal

nanocomposites. Cell response evaluation. Cellulose, vol. 20, pp. 1819-1828.

104. Saralegi, A., L. Rueda, L. Martin, A. Arbelaiz, A. Eceiza, and M.A. Corcuera. 2013.

From elastomeric to rigid polyurethane/cellulose nanocrystal bionanocomposites.

Composites Science and Technology, vol. 88, pp. 39-47.

105. Klemm, D., Schumann, D., Udhardt, U., Marsch, S. & Aranguren, M.. 2001. Bacterial

synthesized cellulose — artificial blood vessels for microsurgery. Progress in Polymer

Science, vol. 26, issue 9, pp. 1561-1603. DOI: 10.1016/S0079-6700(01)00021-1.

Page 44: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

44

106. Wippermann, J., Schumann, D., Klemm, D., Kosmehl, H., Salehi-Gelani, S. & Wahlers,

T.. 2009. Preliminary Results of Small Arterial Substitute Performed with a New

Cylindrical Biomaterial Composed of Bacterial Cellulose. European Journal of

Vascular and Endovascular Surgery, vol. 37, issue 5, pp. 592-596. DOI:

10.1016/j.ejvs.2009.01.007.

107. Cherian, B., Leão, A., De Souza, S., Costa, L., De Olyveira, G., Kottaisamy, M.,

Nagarajan, E. & Thomas, S.. 2011. Cellulose nanocomposites with nanofibres isolated

from pineapple leaf fibers for medical applications. Carbohydrate Polymers, vol. 86,

issue 4, pp. 1790-1798. DOI: 10.1016/j.carbpol.2011.07.009

108. Kweon, H. 2003. A novel degradable polycaprolactone networks for tissue engineering.

Biomaterials, vol. 24, issue 5, pp. 801-808. DOI: 10.1016/S0142-9612(02)00370-8

Page 45: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

45

8. LIST OF SHORTCUTS AND SYMBOLS

c crystallinity

AAL alanine-alanine-lysine

AFM atomic force microscopy

ATR-IR attenuated total reflectance infrared spectroscopy

BASYC® bacterial synthesized cellulose trademark

BD 1,4-butanediol

BC bacterial cellulose

BDI tetramethylene diisocyanate or 1,4-diisocyanatobutane

CT computed tomography

CNC cellulose nanocrystals

CNC-PEG cellulose nanocrystals grafted with poly(ethylene glycol)

CNF cellulose nanofibrils

DBTL dibutyltin dilaurate

DMA dynamical mechanical analysis

DMF N,N-Dimethylformamide

D-PHI degradable polar hydrophobic ionic polyurethane

DSC differential scanning calorimetry

DVO divinyl oligomer

B tensile strain at break or extensibility (%)

ELP4 elastin-like polypeptide-4

Ey Young's modulus (MPa)

FTIR Fourier transform infrared spectroscopy

GA glycolic acid

H12MDI dicyclohexylmethane diisocyanate

HA hydroxyapatite

HA hyaluronic acid

HDI hexamethylene diisocyanate or 1,6-diisocyanatehexane

LA lactic acid

LDI lysine diisocyanate

Page 46: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

46

MDI Methylene diphenyl diisocyanate or 4,4'-methylene diphenyl diisocyanate

NCO/OH isocyanate ratio or isocyanate index

PCL polycaprolactone

PCN poly(hexamethylene carbonate)

PCU polycarbonate-urethane

PEG polyethylene glycol

PEO-PPO-PEO polyethylene oxide-co-polypropylene oxide-co-polyethylene oxide

PEU polyether-urethane

PGA polyglycolic acid

PHA polyhydroxyalkanoates

PHB polyhydroxybutyrate

PHV polyhydroxyvalerate

PLA polylactic acid

PLGA polylactic-co-glycolic acid

PP polypropylene

PU polyurethane

Q water uptake (%)

B tensile stress at break or toughness (MPa)

SBF simulated body fluids

SEM scanning electron microscopy

SMC smooth muscle cell

TE tissue engineering

TEDA triethylenediamine

TGA thermogravimetric analysis

U.S. FDA United States Food and Drug Administration

WAXS Wide-angle X-ray scattering

w/w weight/weight

wt.% weight percent

Page 47: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

47

9. APPENDIX

Text is not available in the public version.

10. LIST OF FIGURES

Figure 1: General formation of polyurethane.

Figure 2: Modelled primary structure of segmented polyurethane [20].

Figure 3: Hydrogen bonding between the chains of a polyurethane based on 1,6 – hexane

diisocyanate and 1,4 – butane diol depicted according to [20].

Figure 4: Chemical structure of a) dibutyltin dilaurate and b) stannous octoate.

Figure 5: Reactions of isocyanate with other functional groups.

Figure 6: Schematic showing hypothesized mechanisms of PU scaffold degradation and

soluble breakdown products. (A) Hydrolysis of ester bonds to yield -hydroxy

acids (dotted line). (B) Abstraction of the -hydrogen atom in the R group

adjacent to the urethane bonds by reaction with hydroxyl radicals to yield lysine

or ethanolamine and carbon dioxide. (C) Abstraction of the -hydrogen atom in

lysine by reaction with hydroxyl radicals to yield multiple degradation products,

including NH4, -ketoacids, oximes, CO2, and carboxylic acids (degradation

products not shown to simplify the diagram) [63].

Figure 7: Schematics of (a) single cellulose chain repeat unit, showing the directionality of

the 1 -> 4 linkage and intrachain hydrogen bonding (dotted line), (b) idealized

cellulose microfibril showing one of the suggested configurations of the

crystalline and amorphous regions, and (c) cellulose nanocrystals after acid

hydrolysis dissolved the disordered regions. Figure depicted according to [97].

11. LIST OF TABLES

Table 1: Applications of polyurethanes in tissue engineering divided by U.S. FDA

standards [11].

Table 2: Isocyanates used in polyurethane chemistry for tissue engineering.

Table 3: Physical properties of the biodegradable polymers used as polyols in formation of

polyurethane.

Page 48: MODIFICATION OF BIODEGRADABLE POLYURETHANES BY

48

Research activities

Articles

Vojtová, L., Kupka, V., Žídek, J., Wasserbauer, J., Sedláček, P., & Jančář, J. (2012).

Biodegradable polyhydroxybutyrate as a polyol for elastomeric polyurethanes. Chemical

Papers, 66(9), pp. 869-874.

Kupka, V., Vojtová L., Jančář, J., Solvent Free Synthesis and Structural Evaluation of

Biodegradable Polyurethane Films Based on Poly(ethylene glycol) and Poly(caprolactone),

Manuscript.

Competitions

2012/12 competition/conference: Student Competition and

Conference, Faculty of Chemistry, BUT, Brno. Awarded

presentation: Polyurethane Elastomers with Potential Use

in Medicine. The best presentation in doctoral section voted

by participants.

2013/6 competition: Ceitec Ph.D. competition, Brno, final round.

Presentation: Solvent Free Synthesized Polyurethane

Elastomers. Participation in the final round.

2014/5 competition: Jean-Marie Lehn’s Prize in Chemistry, Prague,

final round. Presentation: Polyurethane Elastomers with

Potential Use in Medicine. Participation in the final round.

Teaching

2012/autumn semester Practical class in structural analysis of polymers (master degree

students)

2014/15 Supervision of High school scientific activity. Student: Alžběta

Kryštofová from Řečkovice Grammar School, Brno. Topic:

Hydrolytical Stability of Biodegradable Polyurethanes