56
Universidade de Lisboa Faculdade de Farmácia Antifouling surfaces as a strategy to prevent microorganism’s adhesion on medical devices Ana Beatriz Lopes Roque dos Santos Marques Mestrado Integrado em Ciências Farmacêuticas 2017

Ana Beatriz Lopes Roque dos Santos Marques Mestrado

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

Universidade de Lisboa

Faculdade de Farmácia

Antifouling surfaces as a strategy to prevent

microorganism’s adhesion on medical

devices

Ana Beatriz Lopes Roque dos Santos Marques

Mestrado Integrado em Ciências Farmacêuticas

2017

Page 2: Ana Beatriz Lopes Roque dos Santos Marques Mestrado
Page 3: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

Universidade de Lisboa

Faculdade de Farmácia

Antifouling surfaces as a strategy to prevent

microorganism’s adhesion on medical

devices

Ana Beatriz Lopes Roque dos Santos Marques

Monografia de Mestrado Integrado em Ciências Farmacêuticas

apresentada à Universidade de Lisboa através da Faculdade de Farmácia

Orientador: Professora Doutora Isabel Ribeiro, Professora Auxiliar

2017

Page 4: Ana Beatriz Lopes Roque dos Santos Marques Mestrado
Page 5: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

3

Resumo

As infeções associadas aos cuidados de saúde, ou infeções nosocomiais, ou ainda

infeções hospitalares são um grande problema clinico, com impacto significativo na

mortalidade e morbilidade e representam um elevado custo económicos para os

sistemas de saúde. Na europa, o número estimado de doentes com infeções nosocomiais

em 2013 foi de 4.2 milhões. Algumas das infeções hospitalares estão associadas ao

usado de dispositivos médicos, como tubos endotraqueais, cateteres urinários e

venosos. As práticas da medicina moderna levaram a um aumento da utilização destes

dispositivos médicos invasivos. O aumento da sua utilização representa um fator de

risco para o desenvolvimento destas infeções. A contaminação dos dispositivos

médicos pode ocorrer por inoculação de microrganismos provenientes das mãos de staff

médico e clinico. Contudo, é mais provável que ocorra por inoculação de

microrganismos provenientes da pele ou mucosas do paciente aquando da implantação

do dispositivo. Estas infeções resultam da interação entre fatores dos microrganismos,

dispositivo médicos e do hospedeiro. No entanto, a capacidade dos microrganismos

para aderir à superfície dos materiais e promover a formação de um biofilme é o fator

mais importante na patogénese da infeção.

Os biofilmes são comunidade estruturadas de microrganismos que conseguem aderir e

crescer em superfícies bióticas e abióticas. Desta forma, estão protegidos do ambiente

externo, incluindo componentes do sistema imunitário e antibióticos.

Quando se suspeita de uma infeção associada a dispositivos médicos, o tratamento

passa pela remoção ou substituição do dispositivo e/ou iniciar terapêutica

antimicrobiana. Contudo, a suscetibilidade dos microrganismos aos antibióticos está

diminuída. Assim, é mais lógico apostar em estratégias que previnam a formação de

biofilmes.

Apesar dos fatores associados aos microrganismos serem os mais importantes na

patogénese da infeção, os fatores associados ao dispositivo médico são os mais

modificáveis. Fatores como o tipo e forma do dispositivo podem favorecer a adesão

microbiana. Assim, a investigação tem se focado em estratégias que visam a

modificação do dispositivo de forma a prevenir a adesão de microrganismos,

colonização e consequente formação de biofilmes. As estratégias preventivas podem

ser distinguidas em estratégias anti-adesivas e antimicrobianas. As estratégias

Page 6: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

4

antimicrobianas baseiam-se na ligação superficial ou incorporação de substâncias

antimicrobianas, como antibióticos, metais ou compostos de amónio quarternário.

Dispositivos médicos funcionalizados com agentes antimicrobianos estão disponíveis

comercialmente, contudo, a sua utilização deve ser monitorizada devido ao risco de

toxicidade de desenvolvimento de resistência aos agentes utilizados. As estratégias anti-

adesivas e baseiam-se na alteração das propriedades físico-químicas dos dispositivos,

de modo a modificar as interações especificas e inespecíficas entre os microrganismos

e a superfície do dispositivo. A adesão dos microrganismos às superfícies é influenciada

pelas propriedade físico-químicas da superfície, como a carga e a hidrofobicidade. É

também influenciada pelas condições do meio, como presença de proteínas e o pH.

Assim, é necessário que estas superfícies consigam, também, inibir a adesão de outras

biomoléculas.

As propriedades anti-adesivas podem ser conferidas pela modificação química ou física

(da topografia) da superfície. Ainda assim, não é possível desenvolver superfícies com

adesão zero.

Esta monografia foca-se maioritariamente no estudo das diferentes estratégias anti-

adesivas, obtidas por modificação química, usadas em dispositivos médicos.

Como estratégia anti-adesiva têm vindo a ser utilizados os polímeros anti-adesivos têm

sido utilizados, os polímeros poli-hidrofílicos e os poli-zwiteriónicos. Ambas as classes

de polímeros são compostas por polímeros eletricamente neutros, que conseguem

diminuir as interações electroestáticos entre proteínas carregadas e os dispositivos

médicos. Além disso, formam uma camada de hidratação à superfície que funciona

como uma barreira física e energética, que previne a adsorção de proteínas. A camada

de hidratação é formada por ligações de hidrogénio entre as moléculas de água

presentes no meio e os grupos funcionais do dispositivo, no caso dos polímeros poli-

hidrofílicos. Contudo, no caso dos polímeros poli-zwiteriónicos esta é formada por

interações electroestáticas. As superfícies anti-adesivas podem ser obtidas por

diferentes métodos, tais como monocamadas auto-organizadas, revestimento com

hidrogel e polímeros em escova.

O polietilenoglicol é um polímero neutro e hidrofílico e é o polímero mais usado e

investigado para construir superfícies anti-adesivas. Este é utilizado para prevenir a

adsorção de proteínas e a adesão de bactérias, tendo demonstrado conseguir inibir

Page 7: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

5

também a adesão de leveduras. O sucesso da sua ação depende do método de produção

utilizado e da arquitetura do polímero. Apesar do PEG ser considerado uma referência

no que diz respeito aos polímeros anti-adesivos, a sua ação, quando em contacto com

fluidos fisiológicos, é diminuída ou mesmo perdida, uma vez que sofre degradação

oxidativa quando na presença destes fluidos. Assim, o PEG não é apropriado para uma

utilização a longo prazo. Por este motivo, outros polímeros e materiais têm sido

estudados como eventuais alternativas. Algumas dessas alternativas passam pelo uso

de polivinilpirrolidona, polibetaína, poli-oxazolinas, acrilatos polihidroxifuncionais,

poliacrilamida, heparina e fosforilcolina. Estes revestimentos têm demonstrado, em

testes realizados in vitro, bons resultados a inibir a adsorção de proteínas e a adesão de

microrganismo e consequentemente a formação de biofilmes em testes realizados in

vitro.

O PVP é um polímero biocompatível, altamente hidratado e consegue repelir proteínas.

Revestimentos com PVP conseguem inibir a formação de biofilme de algumas bactérias

como E. coli. Devido às suas propriedades anti-adesivas, este polímero é utilizado como

revestimento em dispositivos médicos, nomeadamente em cateteres urinários e

cateteres venosos centrais.

Por sua vez, polímeros de acrilatos polihidroxifuncionais também demonstraram a

capacidade de reduzir a formação de biofilmes de algumas estirpes bacterianas,

nomeadamente S. epidermidis e P. aeruginosa. Uma outra vantagem deste polímero é

ser capaz de reduzir a adsorção de fibrinogénio e plaquetas. Além disso, apresenta

elevada estabilidade mecânica e biológica. Devido a estas características, revestimentos

como estes polímeros são utilizados em dispositivos médicos como cateteres urinários.

As POXs possuem propriedades anti-adesivas semelhantes ao PEG, no entanto estas

apresentam uma maior estabilidade em meio fisiológicos e oxidativo. Testes in vitro

demonstraram que as POXs conseguem reduzir a formação de biofilmes de E. coli.

Devido às suas propriedades anti-adesivas entes polímeros podem ser utilizados como

revestimento de dispositivos médicos como próteses, implantes, entre outros.

Em diferentes testes in vitro, dos acrilatos polihidroxifuncionais demonstraram que

conseguem reduzir a adsorção de proteínas, como o fibrinogénio, e reduzir a adesão de

bactérias. Estes polímeros podem ser aplicados a diversas superfícies, podendo,

nomeadamente, ser aplicado como revestimentos de lentes intraoculares.

Page 8: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

6

Os polímeros de poliacrilamida demonstraram boas propriedades anti-adesivas em

testes realizados em condições in vitro. Estes revestimentos conseguem inibir a

adsorção de proteínas e células e diminuir a formação de biofilmes tanto de bactérias

como de fungos. Polímeros de poliacrilamida podem ser utilizados em dispositivos

médicos como lentes de contacto, cateteres urinários e próteses vocais.

A heparina é um polissacarídeo natural, que devido às suas propriedades

anticoagulantes e anti-adesivas têm sido amplamente utilizada para revestir dispositivos

médicos, nomeadamente em dispositivos que estão em contacto com o sangue, como

cateteres e enxertos vasculares.

A fosforilcolina pode ser usada em polímeros anti-adesivos, uma vez que consegue

reduzir a formação de biofilmes de várias estirpes bacterianas e fúngicas. Polímeros

com fosforilcolina podem ser aplicados a diversos dispositivos médicos, tais como

dispositivos de fixação óssea, stents coronários, pulmões artificiais, entre outros.

Considerando que os polímeros anti-adesivos não conseguem inibir totalmente a adesão

de microrganismos, é possível que estes que consigam aderir à superfície dos

dispositivos formem um biofilme maduro. Assim, a conjugação de polímeros com

propriedades anti-adesivas com agentes microbianos é uma abordagem promissora para

o desenvolvimento de superfícies antibacterianas. Existem vários revestimentos que

conjugam ambas as propriedades, nomeadamente polímeros de PEG funcionalizados

com antibióticos.

Os vários materiais abordados nesta monografia conseguem formar dispositivos

médicos com superfícies anti-adesivas e assim ajudar a combater as infeções associadas

a dispositivos médicos. No entanto estes têm características distintas entre si e por isso

o material escolhido para revestimento um dispositivo médico deve ser selecionado

tendo em consideração a sua aplicação especifica.

Palavras-chave: Infeções nosocomiais; Dispositivos médicos; Adesão bacteriana;

Biofilmes; Superfícies anti-adesivas.

Page 9: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

7

Abstract

Healthcare-associated infections are a major clinical problem with significant impact

on mortality and morbidity and represent an economic burden of health systems. The

number of patients with a HAIs, in 2013, in Europe, was estimated at 4.2 million. The

use of invasive medical devices, such endotracheal tubes, urinary catheters, central

venous catheters, mechanical heart valves and others represent a risk factor for these

infections. Such infections are related to biofilm formation onto medical device

surfaces. Biofilms are a structured community of microorganisms that can attach and

grow on abiotic and biotic surfaces and are protected against antimicrobials and

immune system components. Thus, the best approach against such infections is the

prevention of biofilm formation.

This monographic work aims to study the different antifouling strategies used to prevent

biofilms formation on medical devices.

The preventive approaches can be divided into antifouling and antimicrobial strategies.

Antimicrobial strategies are based on the incorporation or surface bonding of

antimicrobial substances, such as antibiotics. Antifouling strategies are based on the

modification of basic polymers to develop polymers with new surface properties. PEG

is the gold standard of the antifouling polymers however, has been reported that is not

appropriate to long-term applications. Thus, several materials such PVP, POXs,

polybetaines, poly (hydroxyfunctional acrylates), PAAm, heparin and

phosphorylcholine have been studied for anti-adhesive applications and showing to be

able to reduce protein adsorption, cell adhesion and bacterial attachment and

consequently reduce biofilm formation. Nevertheless, more studies are necessary to

assess their efficacy under real physiological conditions.

Various antifouling polymers are already being applied in medical devices such as

catheters, orthopedic implants, dental implants, vascular grafts, contact lenses, among

others. The drawback is that despite these polymers being able to reduce/delay biofilm

formation on medical device surfaces, a few microorganisms can eventually adhere and

form a biofilm. Another interesting approach is the possible association of theses

coatings with antimicrobial agents to improve the antibiofilm properties of the material.

Page 10: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

8

Antifouling coatings are different from each other, thus the type of coating should be

adapted for a specific application.

Keywords: Healthcare associated infections; Medical devices; Bacterial adhesion;

Biofilm; Antifouling surfaces.

Page 11: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

9

Abbreviations

AM - Ampicillin

Amino-PPX – poly(o-amino-p-xylylene-co-p-xylylene)

ATP – Adenosine 5’- triphosphate

ATRP - Atom transfer radical polymerization

BFG – Bovine fibrinogen

BSA – Bovine serum albumin

CHT – Chitosan

CLSM – Confocal laser scanning microscope

CSF – Cerebrospinal fluid

DFS – Dynamic force spectroscopy

ECDC – European Centre for Disease Prevention and Control

EGDA – Ethylene glycol diacrylate

EPS – Extracellular polymeric substances

ePTFE – Expanded polytetrafluoroethylene

FDA – US Food and Drugs Administration

Fg – Fibrinogen

GEN – Gentamicin

HAIs – Healthcare Associated Infections

HP - Heparin

ICU – Intensive care unit

IOLs – Intraocular lenses

LbL – Layer-by-layer

Lyz – Lysozyme

MA - Maleic anhydride

Page 12: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

10

MAPC-co-BMA - Poly [(ω-methacryloyloxyalkyl phosphorylcholine-co-n-butyl

methacrylate)]

MPC – 2-methacryloyloxyethyl phosphorylcholine

OD – Optical density

OEG – Oligo (ethylene glycol)

PAAm – Polyacrylamide

PBS – Phosphate-buffered saline

PC – Phosphorylcholine

PCBMA – Poly (carboxybataine methacrylate)

PDMS – poly(dimethylsiloxane)

PEG – Poly (ethylene glycol)

PEN – Penicillin

PEO – Poly (ethylene oxide)

PEOXA – Poly (2-ethyl-2-oxazoline)

PET – Polyethylene terephthalate

PHEAA – Poly (N-hydroxyethylacrylamide)

PHEMA – Poly (2-hydroxyethyl methacrylate)

PHPMA – Poly N-(2-Hydroxypropyl methacrylamide)

PLL-g-PEG – Poly(L-lysine)-graft-poly (ethylene gylcol)

PLL-g-PMOXA – Poly(L-lysine)-graft-poly(2-methyl-2-oxazoline)

PMB – poly (MPC-co-n-butyl methacrylate)

PMMA – Poly (methyl methacrylate)

PMOXA – Poly (2-methyl-2-oxazoline)

POEGMA – Poly (oligo (ethylene glycol) methyl ether methacrylate)

POXs – Poly (2-oxazoline)s

PP – Polypropylene

Page 13: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

11

PPO – Polypropylene oxide

PPOXA – Poly (2-phenyl-2-oxazoline)

PRP – Platelet rich plasma

PS – Polystyrene

PSA – Poly (3-sulfopropyl methacrylate potassium)

PSBMA – Poly (sulfobetaine methacrylate)

PU – Polyurethane

PVDF – Poly (vinylidene fluoride)

PVP – Poly (vinylpyrrolidone)

SAM – Self-assembled monolayer

SE – Silicone elastomers

SEM – Scanning Electron Microscopy

SPR – Surface plasma resonance

TMC – N-Trimethyl chitosan

TT – Tympanostomy tubes

UV-VIS – Ultraviolet - Visible

XPS – X-ray photoelectron spectroscopy

Page 14: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

12

Index

1. Introduction .......................................................................................................... 13 1.1 Healthcare-associated infections .................................................................. 13

1.1.1 Medical device-associated infections ...................................................... 13 1.2 The role of biofilms in device-associated infections ................................... 14 1.3 Strategies to control bacterial biofilm ................................................................ 15

1.4 Strategies to prevent biofilm formation ............................................................. 15 2. Objective .................................................................................................................. 17 3. Materials and methods ............................................................................................. 17 4. Results ...................................................................................................................... 17

4.1 Antifouling surfaces ........................................................................................... 17

4.1.1 Poly (ethylene glycol) and PEG-based materials........................................ 19

4.1.2 Poly (vinylpyrrolidone) ............................................................................... 23 4.1.3 Polybetaine .................................................................................................. 25

4.1.4 Poly(2-oxazoline)s ...................................................................................... 28 4.1.5 Poly (hydroxyfunctional acrylates) ............................................................. 30 4.1.6 Polyacrylamide ........................................................................................... 32 4.1.7 Heparin ........................................................................................................ 34

4.1.8 Phosphorylcholine....................................................................................... 36 4.2 Antifouling and antimicrobial surfaces .............................................................. 38

4.2.1 PEG-Antibiotics .......................................................................................... 38 4.2.2 Heparin/Chitosan multilayer film ............................................................... 40 4.2.3 MEO2MA based copolymer ........................................................................ 42

5. Discussion ................................................................................................................ 43

6. Conclusion ............................................................................................................... 45 7. Bibliography ............................................................................................................ 46

Index of Figures:

Figure 1. Stages of biofilm formation…………………………………………………14

Figure 2. Antifouling coating…………………………………………………………16

Figure 3. Examples of surface functionalization methods……………………………18

Figure 4. Structure of Poly (ethylene glycol)………………………………………….19

Figure 5. Structure of Poly (vinylpyrrolidone)………………………………………..23

Figure 6. Structure of sulfobetaine and carboxybetaine, respectively………………..25

Figure 7. Structure of Poly(2-oxazoline)s…………………………………………….28

Figure 8. Structure of pHEAA, pHEMA and pHPMA, respectively…………………30

Figure 9. Structure of Polyacrylamide………………………………………………..32

Figure 10. Structure of Heparin……………………………………………………….34

Figure 11. Structure of Phosphorylcholine……………………………………………36

Page 15: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

13

1. Introduction

1.1 Healthcare-associated infections

Healthcare-associated infections (HAIs), also mentioned to as “nosocomial” or

“hospital infection”, are infections that patient acquire during the course of receiving

treatment for other conditions, in a hospital or another healthcare facility, and are not

present or incubating at the time of admission. (1,2) Such infections are the most

frequent adverse event in care delivery, having a significant impact on mortality,

morbidity, and quality of life, leading to an economic consequence for health systems.

(1,3) According to the European Centre for Disease Prevention and Control (ECDC)

surveillance report, the annual number of patient with a HAIs, in European acute care

hospitals, was estimated at 4.2 million, in 2013. The most frequent HAIs were

respiratory tract infections (33.6%), symptomatic urinary tract infections (22.3%), and

skin infections (21.4%). The most common microorganism associated with HCAIs

were Escherichia coli, Staphylococcus aureus, Enterococcus spp., Pseudomonas

aeruginosa, Klebsiella spp., Coagulase-negative staphylococci and Candida spp.. (4)

An important issue is that some of these infections are related with the use of medical

devices. (4)

1.1.1 Medical device-associated infections

Medical devices are widely used in medical modern practice for diagnostic and

therapeutic purposes. Nevertheless, the increasing use of this invasive devices

represents a significant risk factor for the development of HAIs. (5,6) Medical devices

such as urinary catheters, central venous catheters, endotracheal tubes, mechanical heart

valves and others are associated with high rate of infections. (2,7) According to ECDC

surveillance report, in 2013, (intensive care unit) UCI-acquire pneumonia was device-

associated in 92% of the cases, UCI-acquired urinary tract infections and UCI- acquired

bloodstream infections were reported catheter-related in 96.7% and 43.3%,

respectively. (4)

The contaminations of medical devices can occur by inoculation with microorganisms

from the hands of clinical or medical staff. However is more likely occur by inoculation

of pathogens from the patient’s skin or mucous membrane, during implantation of the

medical device. (6,8) There are many risk factors for the development of HAIs, such

Page 16: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

14

invasive surgery, prolonged use of invasive medical devices, long hospital stay,

immunocompromised patients and other underlying patient conditions. (1,2)

The medical device-associated infections result from the interaction of three factors:

microorganism, device and host factors. The ability of bacteria to adhere to materials

and promote the formation of a biofilm is probably the most important factor in the

pathogenesis of infection. (6,7)

1.2 The role of biofilms in device-associated infections

A biofilm can be defined as a structured community of microorganisms that can attach

and grow on abiotic and biotic surfaces. They produce extracellular polymeric

substances (EPS) which protects them from the external environment, including

antimicrobials and immune system components. (2,5) In many cases antimicrobial

resistance is associated with long-term persistence of biofilm infections. (6,9) Biofilms

can be composed of single species or multiple species of microorganisms. (10)

Biofilms formations include different stages (Figure 1): reversible attachment,

irreversible attachment, colonization, maturation and dispersion. Different

microorganisms have different and specific mechanisms that allow initial surface

attachment, development and detachment from the biofilm.Figure 1 (2,11)

Figure 1. Stages of biofilm formation

Page 17: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

15

First, free microorganism cells (planktonic cells) adhere to a surface by reversible

bonds, such a van der Waals force, hydrophobic and electrostatic interactions. (6,11,12)

The initial attachment is reversible, whereas is followed by an irreversible attachment.

The permanent attachment to the surface is stimulated by features of the microbial cell

surface (i.e. flagella, pili, fimbriae, glycocalyx and cell adhesion molecules). Once

adhered to the surface, microorganisms continue to multiply and accumulate in

multilayered cell clusters and begin to build an extracellular matrix. The matrix, along

with other microbial components, stabilizes biofilm structure and holds the biofilm

together. Finally, the aggregated cells may detach from the biofilm and proceed to

colonize other sites or cause systemic infection. (2,6,9,11)

1.3 Strategies to control bacterial biofilm

When a medical device-associated infection is suspected, a general decision is to

remove or replace the device and/or initiate antimicrobial therapy. (6) The clinical

experience with HAIs demonstrates that defence mechanisms of the host are incapable

of handle the infection. (8) Compared with free living cells, biofilms are more difficult

to eliminate because they are better protected against immune system components and

antibiotics. (10,13) The levels of susceptibility to antimicrobial agents are decreased

10-1000 times less. (6) The failure of antimicrobial therapy can result in chronic

infection. (14) Therefore, inhibiting biofilms formation is a more reasonable approach

than the possible dispersal of the formed biofilms. (10)

1.4 Strategies to prevent biofilm formation

As was mentioned above, these infections result from the interaction between

microorganism, device and host factors. Microorganisms factors are the most important

in the pathogenesis and some stages of biofilms formations can be possible targets of

preventions strategies. However, device factors are the most modifiable. Several

device-related factors may favor microorganisms adhesion such as the type of device

material, source of device material, surface and shape of the device. (7) Recently

research has been focused on the modification of the medical device, in order to prevent

bacterial attachment, colonization and, consequently, biofilm formation. (2) The

Page 18: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

16

preventive approaches can be distinguished in antifouling and antimicrobial strategies.

(15,16) The antimicrobial strategies are based on the incorporation or surface bonding

of antimicrobial substances, such as antibiotics, metals, quaternary ammonium and

disinfectants. Medical device with antimicrobial agents are commercially available and

are already used in clinical applications. However, the use of this devices must be

monitored due to the risk of toxicity and development of antimicrobial resistance

against the agents included. (6,10,17) Another preventive approach are the antifouling

strategies. These are based on the modification of basic polymers to develop polymers

with new surface properties, such as antiadhesive polymers (Figure 2). (6,17)

Microorganisms adhesion to surfaces is influenced by the microorganism and

physicochemical properties of the surface, such as hydrophobicity and surface charge.

It is also affected by environment properties, like the presence of proteins and pH. Thus,

it necessary that antifouling surfaces can also prevent non-specific interactions with

proteins and other biomolecules. (12,18,19)

Since, the intrinsic properties of a material influences microbial adhesion is possible to

improve the surface of the device for development infection-resistant materials. Anti-

adhesive properties can be conferred by modification of the topography or chemistry of

the surface. Nevertheless, is not possible develop surfaces with zero adhesion. (6,12)

Another effective approach can also be the combination of the two previous approaches,

i.e., conjugate both antiadhesive and antimicrobial properties. (11)

These surface modification strategies, namely antifouling polymers, are a growing area,

with applications in many fields beyond medical devices and have been target of several

research studies. (12,20,21)

Figure 2. Antifouling coating

Page 19: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

17

2. Objective

The aim of this monograph is to study the different antifouling strategies used to prevent

biofilms formation in a medical device. This work will focus mainly on antifouling

strategies by chemical modification.

3. Materials and methods

The present review is a research at antifouling surfaces used to prevent microorganism’s

adhesion on medical devices. To write this monograph, different data bases were used,

such as Web of Knowledge, PubMed, Science Direct and B-on. First, the research was

focused in a review article in the area published over the last five years. For this search,

terms such as “antifouling strategies for medical devices”, “antifouling coatings”,

“antiadhesive coatings”, “antibiofouling polymers”, “anti-infectious surfaces”,

“antibacterial coatings” and “polymers for biomedical applications” were used. After

this, a more specific research was made to find studies for each strategy selected. Thus,

as an example the search terms such as “PEG polymers for biomedical application” and

“PEG coating preventing microbial adhesion” were selected. Articles published within

ACS, Springer or Elsevier were favoured. This monograph, intends to make a review

on different studies published in literature mentioning antifouling strategies to prevent

medical device-associated infections.

4. Results

4.1 Antifouling surfaces

Antifouling strategies are based on the modification of basic polymers to develop

polymers with new surface properties. (6,17) These modifications lead to a change in

nonspecific and specific interactions between the microorganism and the material

surface. Thus, is possible to develop medical devices resistant against microbial

adhesion. (6) Two major classes of antifouling polymers coatings have been used for

repelling protein and bacteria. These classes are based on polyhydrophilic and

polyzwitterionic materials. Since, these polymers are electrically neutral, they can

reduce electrostatic interactions between charged protein domains and device surfaces.

Page 20: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

18

The antifouling properties of these coatings are also related to the formation of a

hydration layer, near the surface. This layer prevents protein adsorption, by acting as

an energetic and physical barrier. The hydration layer, in hydrophilic surfaces, is

formed by hydrogen bonds between water molecules in the environment and functional

groups on the medical device surface. The strength of this barrier affects the

effectiveness of the hydrophilic material and depend on the physicochemical properties

of a material (e.g. molecular weight) and surface packing (e.g. polymer chain). The

hydration layer in zwitterionic surfaces is strongly bound through electrostatic

interactions. The stronger the interaction, the more resistance to adhesion is the

material. (12,15)

Manny antifouling strategies, based on these polymers, have been applied to design

medical devices, implants and biosensors with new surface properties. (22) Antifouling

surfaces can be obtained by various methods, and examples are self-assembled

monolayer (SAM), polymer brush coatings and hydrogel coatings (Figure 3). (19)

SAMs represent a highly organized molecular assembly formed spontaneously by

chemisorption followed by self-organization of long chain molecules, on the surface.

These molecules consist of three domains, i.e. head group, alkyl chain and

functionalized end region. (23,24) On the other hand, Polymer brushes are polymer

chains covalently tethered on a surface. This coating can be obtained by grafting-from

and grafting-to techniques and applied onto varied materials (e.g. silicon, titanium).

(25,26) Another technology for surface modification is hydrogel coatings. A hydrogel

is a hydrophilic polymer that can reduce microbial adhesion by formation of a soft

surface. Furthermore, hydrogel can absorb water, which also contribute to reduce

biofilm formation. (2)

Figure 3. Examples of surface functionalization methods

Page 21: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

19

4.1.1 Poly (ethylene glycol) and PEG-based materials

Poly (ethylene glycol) (PEG) (Figure 4), a neutral hydrophilic polymer and is widely

studied as an antifouling polymer. (26) The antifouling properties of PEG or PEG-based

materials are due to the hydrophilic nature of these polymers that leads to the formation

of a hydration layer which inhibits protein attachment and microorganisms adhesion.

(22)

In fact, these coatings have demonstrate to reduce adhesion of yeasts and bacteria in

vitro. (27) The success of PEG as an antifouling polymer depends, in general, of the

polymer architecture and the surface grafting method. (22) PEG can be attached to

surfaces by different grafting techniques such as “grafting from”, “grafting to” block

copolymerization, chemical functionalization, among others. (22,24)

As mentioning above, PEG is one of the most studied antifouling polymer, one of these

studies was performed by Fernandez et al. (27), that developed a multi-component

cross-linked PEG-based polymer coating to inhibit non-specific biomolecular

adsorption, cell and protein attachment. This coating is formed by three core coating

components (i.e. the active, the matrix-forming and the intermolecular cross-linking

component) and applied to surfaces from a volatile carrier solvent, in a single step. (27)

The first component is a hetero-bifunctional PEG molecule, terminated with a

succinimidyl ester (functional group) and an alkoxysilane terminus (reactive

crosslinking group). The second component is a polyoxyethylene sorbitan tetraoleate

and the third is an azidosilane. (27)

This polymer was characterizatied by X-ray photoelectron spectroscopy (XPS), water

contact angle and streaming potential and the results showed that the surface was

hydrophilic and negatively charged. The microbial adhesion was evaluated in a parallel

plate flow chamber, with a bare glass as a control. For this study five bacterial strains

Figure 4. Structure of Poly (ethylene glycol)

Page 22: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

20

were used, Streptococcus salivarius, Staphylococcus epidermidis 3399,

Sthaphylococcus epidermidis HBH 276, Escherichia coli and Pseudomonas

aeruginosa. Obtained results revealed that the coating reduces adhesion of many

clinically isolated bacterial strains in vitro. An exception was P. aeruginosa that

adhered similarly to uncoated and coated glass. (27) In another study was reported that

P. aeruginosa (adhesive strains) releases surface-active substances, such biosurfactants,

that can penetrate the coating. This may cause decreasing of poly (ethylene oxide)

(PEO) interfacial properties and increasing attractive interactions between bacteria and

PEO brush. (28) The small effect of the multi-component coating in inhibiting

attachment of P. aeruginosa could be related with this bacterial factor. (27)

Also the effectiveness and stability of the coating in physiological fluids were

evaluated. The polymer continues stable and effective against S. epidermidis 3399

adhesion after seven days’ exposure in urine. However, the antifouling effectiveness

decrease over time in protein-rich physiological fluids, like saliva. An additional

advantage of this three component coating is its compatibility with varied substrates,

such as metal oxide, glass and numerous plastics. (27)

The antifouling properties of this multi-component crosslinked PEG-based polymers

have also been investigated by other authors (29). The results showed that this multi-

component polymer reduced adhesion of non-specific proteins. Furthermore, it also

decreased adhesion of fibrinogen (Fg) and lysozyme (Lyz). The inhibition of bacterial

adhesion was assessed with two bacterial strains, S. aureus and K. pneumoniae, in a cell

flow. Results demonstrated that the adhesion of these bacteria to the uncoated surfaces

(i.e. bare glass and bare indium tin oxide) was strong and mainly irreversible (>60%

and >80% of the microorganisms remains in the surface, respectively). However,

almost all the bacteria cells were removed from the coated surfaces. (29)

The antifouling properties of another grafting technologies for PEG and PEG-based

materials also have been investigated. Roosjen et al (30) developed a (PEO)-brush

coating, covalently attached to silica and glass by reaction in a polymer melt. The

presence of a coating was evaluated by X-Ray photoelectron spectroscopy, and contact

angle measurement showed that coated surfaces were more hydrophilic that uncoated

surfaces. Its antifouling properties were assessed in a parallel plate flow chamber,

utilizing various microbial strains, S. epidermidis, Staphylococcus aureus, S. salivarius,

E. coli, P. aeruginosa, Candida albicans and Candida tropicalis, and compared to

Page 23: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

21

adhesion to bare glass. After 2 h of incubation, PEO-coated side, did not have any

bacteria or yeast adhered, however the uncoated glass was covered by microorganisms.

After 4 h, the results showed a decrease in adhesion in the PEO-coated side when

compared to uncoated glass. This reduction was more than 98% for S. epidermidis, S.

aureus and E. coli and a reduction of 88% was observed with S. salivarius.

Nevertheless, the results of P. aeruginosa adhesion were not statistically significant.

This coating also reduced the adhesion of C. albicans and C. tropicalis, in 81% and

75%, respectively. (30)

In conclusion, this study showed that bacterial adhesion to PEO-brushes was greatly

decreased with respect to adhesion to glass, except when more hydrophobic bacteria

were tested, such P. aeruginosa. Comparing to bacterial adhesion, the extent of

reduction of yeasts strains adhesion was smaller. Based on these outcomes, the authors

proposed that PEO-brushes decreased the microbial adhesion by reduction of van der

Waals interactions, between microorganisms and the surfaces. (30)

In another study, the same authors investigated the influence of the polymer molecular

weight and temperature in microbial adhesion. The adhesion of S. epidermidis,

P.aeruginosa, C. albicans and S. tropicalis in PEO chain of 526, 2000 and 9800 Da

was compared to their adhesion to bare glass. The adhesion was evaluated in a parallel

plate flow chamber at 20 ºC and 37 ºC. The results show, after 4 h of incubation, that

adhesion of P. aeruginosa and C. tropicalis was only decreased in higher molecular

weight brushes due to the hydrophobic character of these microorganisms. On the other

hand, the three PEO-brushes could reduce adhesion of S. epidermidis and C. albicans.

In this study, the retention of microorganisms was also assessed by a passage of an air

bubble. The results demonstrated that almost all microorganisms adhering to the PEO

brush were removed, independently of brush length. However, the retention of the

microorganisms on uncoated glass was greatly higher. The experiments were realized

at 20 ºC and 37 ºC however, no significant differences in adhesion or retention were

found. (31)

The polymer brush-coating with PEG-based materials was also assessed by Nejadnik

et al, (32) that studied its effect on the kinetics of bacterial growth. For this, initial

attachment and succeeding 20 h growth was evaluated with three different bacterial

strains (i.e. S. aureus, S. epidermidis and P. aeruginosa) on uncoated and coated

silicone rubber, in a parallel plate flow chamber. They used a tri-block copolymer of

Page 24: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

22

poly (ethylene oxide)-polypropylene oxide (PPO) – poly (ethylene oxide)

(PEO99PPO65PEO99), a low-density polymer brush coating, physically adsorbed to

hydrophobic silicone rubber. Initial attachment of S. aureus and S. epidermidis (after

30 min) decreased from 5.2x106 cm-2 and 4.1x106 cm-2 on uncoated silicone to 0.4x106

cm-2 and 0.1x106 cm-2 on coated silicone, respectively. On the other hand, initial

attachment of P. aeruginosa was similar on coated and uncoated silicone. The biofilms

grew on both surfaces, however, developed more slowly on brush-coatings. The strains

of Staphylococcus demonstrated a lag-time of 3 h, on uncoated silicone, after which

biofilm initiated to grow exponentially. After 8 h of growth, the entire surface was

covered by biofilm. However, on coated silicone, the lag-time was approximately 8h

and after 20 h only 71% and 60% of the surface was covered by S. aureus and S.

epidermidis biofilm, respectively. In the case of P. aeruginosa, the kinetics of bacterial

growth was completely different and the surface coverage was smaller than 24% in both

surfaces. (32)

To assess the antifouling properties after growing of a biofilm, an induced detachment

was performed by shear stress. Biofilm of Staphylococcus strains was completely

removed from the coating surface, whereas the removal from uncoated silicone was

incomplete, only 58% and 71% of the biofilm of S. aureus and S. epidermidis was

successfully removed, respectively. Biofilm of P. aeruginosa could not be stimulated

to remove, on both surfaces, by shear stress. So, the authors concluded that biofilms on

coatings developed more slowly and were more easily removed by high fluid shear.

(32)

Sustainability of the antifouling properties of the coating was also assessed. The results

demonstrated that formation and succeeding removal of S. epidermidis biofilm did not

affect the non-adhesiveness of the coating. However, part of the antifouling properties

was lost after removal S. aureus biofilm. Nonetheless, brush coating continues to be

effective against Staphylococcus strains, compared with the adhesiveness to uncoated

silicone. The loss of non-adhesive functionality could be explained by the presence of

nitrogen containing bacterial components that remain on the brushes after biofilm

removal. A higher amount was found after removal of S. aureus biofilm when compared

to S. epidermidis. These findings can be clinically significant, once the slower biofilm

formation on polymers brushes coatings, can permit more time for treatment with

antibiotics before a more resistant biofilm developed. (32)

Page 25: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

23

Polymer coating with PEG can be used in biomaterials, and an example is titanium

surfaces. Titanium and titanium alloys are widely used as hard tissue replacements in

dental and orthopedic implants (artificial joint and bones). Are also used in prosthetic

heart valves and artificial hearts. Polyurethane (PU) is a synthetic polymer used to made

cardiovascular implants, such vascular grafts. PU surfaces can be modified using PEG

and its derivatives for additional hydrophilicity. (33–36) PEO can also be used in coated

silicone contact lenses. (37)

The drawback is that although, PEG coatings have been shown to reduce

microorganism’s adhesion in vitro, after contact with physiological fluids in vitro or in

vivo, the reduction is generally lesser or even lost. This can be due to the continuous

bulk protein assault that leads to an eventual overwhelming of the surface or coating

degradation (e.g. chain cleavage, hydrolysis, surface removal). (27,38) Furthermore, it

has been described that, under in vivo conditions, the terminal hydroxyl group of PEG

might be oxidized to an aldehyde and PEG chains, in aqueous solution, shows weak

hydrophobic interactions with proteins. Consequently, PEG may not be appropriate as

a long term antifouling surface, and other polymers are being explored as potential

alternatives. (12)

4.1.2 Poly (vinylpyrrolidone)

Poly(vinylpyrrolidone) (PVP) (Figure 5) is a highly hydrated, biocompatible, water

soluble and protein-repellent synthetic polymer. (22)

Due to its properties, PVP have been studied to originate antifouling surfaces. Antonelli

et al. (39), studied the influence of (PVP)-coated tympanostomy tubes (TT)

(commercially available) on biofilm formation of P. aeruginosa and S. aureus. Silicone

TTs with or without PVP coating were exposed to blood or phosphate-buffered saline

(PBS), for 24 h, and biofilm development was evaluated by quantitative bacterial counts

Figure 5. Structure of Poly (vinylpyrrolidone)

Page 26: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

24

and scanning electron microscopy (SEM). The results demonstrate that P. aeruginosa,

after 4 days in culture, formed a biofilm on both PVP-coated and uncoated TTs.

However, the number of bacteria was lower in PVP-coated TT. On the other hand, PVP

coating did not demonstrate a significant effect on preventing biofilm formation of S.

aureus. Thus, they conclude that PVP coating reduced biofilm formation of some

bacteria under certain conditions. (39)

The effectivity of PVP at repelling both protein and bacteria was evaluated by Sun et

al. (40). For thus, PVP crosslinked with ethylene glycol diacrylate (EGDA) was grafted

to surfaces (i.e. polyvinylidene fluoride (PVDF) membrane), via vapor-based grafting.

They have created coatings with a different crosslinking degree. The antifouling

properties of this coating was tested using bovine serum albumin (BSA) and E. coli, as

protein and bacteria models, respectively. The results of protein adsorption test show

that uncoated PVDF membrane have the major adsorption of BSA. The PVP-co-EGDA

coating reduced BSA absorption more than 90% compared to the uncoated membrane.

The adhesion of E. coli was tested on glass surfaces coated with PEGDA and PVP-co-

EGDA. The surface coated with PEGDA exhibited similar bacteria adhesion as the

uncoated glass surface. For surfaces coated with PVP-co-EGDA with different

crosslinking degrees , it was observed that the decrease in the crosslinking degree

contributed to reduce adhesion of E. coli. (40)

Other authors have also evaluated the antifouling properties of the PVP, and an example

was Liu et al. (41), that developed a PVP-grafted poly(dimethylsiloxane) (PDMS)

surfaces by surface-initiated atom transfer radical polymerization (ATRP). The

antifouling properties of the PVP-grafted PDMS surfaces was tested by the

determination of the protein adsorption and the cell and bacteria adhesion. Fg was

chosen as a model protein and its adsorption was decreased by 86% on the PVP90-

grafter PDMS when compared with the uncoated PDMS surface, even after 30 days at

ambient conditions. All other PVP-grafted PDMS surfaces tested showed good

resistance to protein adsorption. The resistance to bacterial fouling was also tested by

cell and bacterial adhesion tests, using L929 cells and E. coli, respectively. The results

demonstrated that practically no cell or bacteria adhered to PVP90-grafted PDMS

surfaces when compared to uncoated PDMS surfaces, even after 30 days in ambient

conditions. All of the other PVP-grafted PDMS surfaces tested showed similar results.

(41)

Page 27: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

25

Surfaces functionalized with PVP have also been selected improve the blood

compatibility of PU central venous catheters. (42) PVP hydrogels have been used on

indwelling urinary catheters, since this coating has showed reduce bacterial attachment.

(43) It has also been applied in biosensors and biochips, haemodialysis and blood

purification units. (24) PVP can be used to increase the water content of polymers

included into soft contact lenses, such as poly (2-hydroxyethyl methacrylate) (PHEMA)

, poly (methyl methacrylate) (PMMA) or silicone. (37)

Furthermore, PVP hydrogel also has demonstrated to reduce surface roughness and

decrease bacterial adhesion and protein absorption on central venous catheters. (43)

4.1.3 Polybetaine

Zwitterionic polybetaines have been studied as antifouling coatings due their

hydrophilic properties. (26) They are named according to the negatively charged group

and include sulfobetaine and carboxybetaine (Figure 6). (33)

One of these studies was reported by Cheng and co-workers (44) that have studied the

antifouling properties of long-chain zwitterionic poly(sulfobetaine methacrylate)

(PSBMA) grafted in surfaces (i.e. gold surfaces) using ATRP. They investigated their

resistance to short-term and long-term bacterial adherence and biofilm formation with

two bacterial strains (i.e. S. epidermidis and P. aeruginosa), using a laminar flow

chamber. For comparison, poly (oligo(ethylene glycol) methyl ether methacrylate)

(POEGMA) grafted surfaces (i.e. gold surfaces) were also investigated. Additionally,

they quantified how surface grafting methods would influence the long-term resistance

to bacterial attachment and the coating stability. In that way, were prepared SAMs of

alkanethiols with shorter-chain oligo (ethylene glycol) (OEG) and mixed

Figure 6. Structure of sulfobetaine and carboxybetaine, respectively

Page 28: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

26

SO3−/N+(CH3)3 (SA/TMA) terminated groups. Methyl (CH3) SAMs grafted on gold and

bare glass was also prepared as a reference. (44)

The short-term adhesion (i.e. after 3 h) of S. epidermidis showed a significantly

decrease on POEGMA, PSBMA, OEG SAMs and SA/TMA SAMs surfaces as

compared to CH3 SAMs and bare glass. On the other hand, short-term attachment of P.

aeruginosa to OEG SAMs was reduced by 87% when compared to the glass surface.

Adhesion of P. aeruginosa on PSBMA and POEGMA was reducing in 80% and 75%,

respectively, compared to OEG SAMs. The lower adhesion on PSBMA and POEGMA

surfaces than on SA/TMA and OEG SAMs are due to the longer molecular chains of

the antifouling groups. They found that S. epidermidis and P. aeruginosa adhered more

to SAMs that POEGMA or PSBMA. This could be a result of degradation of the SAMs

with the time and/or thickness of the coating produced using brushes. The long-term

accumulation of two bacterial strains on these surfaces was studied, utilized bare glass

and CH3 SAM on gold were selected as control. After 24 h or 48 h, qualitative images

showed that PSBMA and POEGMA reduced biofilm formation of P. aeruginosa and

S. epidermidis as compared to the controls. Both PSBMA and POEGMA reduced

bacterial adhesion, thus PSBMA (zwitterionic material) can be used as an antifouling

material. (44)

In another work, Cheng et al. (45), reported a study on the resistance to long-term

biofilm development of zwitterionic poly(carboxybetainemethacrylate) (PCBMA). The

influence of various conditions, such a temperature, bacterial strain and zwitterionic

group (PCBMA and PSBMA) on the antifouling properties were investigated. The

PCBMA and PSBMA brushes were grafted from glass and gold surfaces by ATRP. The

antifouling performance of PCBMA coating was further investigated by parallel flow

cell system using two bacterial strains (i.e. P. aeruginosa and P. putida). The results

showed that PCBMA coating decreased long-term biofilm formation of P. aeruginosa

up to 10 days in 95% at 25 ºC and up to 2.5 days in 93% at 37 ºC, compared to

unmodified glass. The coating also reduced the biofilm formation of P. putida up to 8

days in 95% at 30 ºC. PSBMA surface was also evaluated by parallel flow cell, using a

strain of P. aeruginosa at 25 ºC, and compared with PCBMA surface. Results showed

that PSBMA coated surfaces could decrease biofilm formation over 9 days, in other

words, the capability of PSBMA coating to resist biofilm development was similar to

Page 29: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

27

PCBMA. They concluded that PCBMA surfaces could significantly delay biofilm

formation. (45)

In order to investigate the blood compatibility of these polymers, Jiang et al. (46),

grafted sulfobetaine onto PU and assessed the protein absorption with bovine fibrinogen

(BFG) and platelet adhesion with platelet rich plasma (PRP). The quantification of

absorbed BFG on the both coated and uncoated surfaces was made using 125I-labeled

protein, with a radioimmunoassay. Water contact angle results demonstrated that

uncoated PU surfaces was more hydrophobic that sulfobetaine coated surfaces (78 º ±

3º, 60º ± 3º, respectively). The results showed that the adsorption of BFG onto coated-

PU was reduced by 8.8%. The results of platelet adhesion were observed by SEM and

showed that the number of platelets adhered to the surface decreased in coated-PU when

compared to uncoated-PU, after 1 h and 3 h. These tests demonstrated that films grafted

with sulfobetaine could reduce protein adsorptions and resist to platelet adhesion. (46)

The biological and mechanical stability of carboxybetaine zwitterionic hydrogel was

investigated by Hsiang-Chieh Hung et al.(47). In this study, three main terminal

sterilization techniques (i.e. ethylene oxide gas, steam autoclave and gamma

irradiation) were used. The results showed that carboxybetaine hydrogels were stable

at an oxidative gas environment and high pressure and temperature without altering

their antifouling properties. (47)

PSBMA coating was studied as a grafted layer to modify cellulose and PU surface due

to their anti-thrombogenicity properties. Nevertheless, medical devices with PSBMA,

commercially available, are rarely seen. US Food and Drug Administration (FDA) have

approved two PSBMA modified peripherally inserted central catheters. (43) Vascular

catheters of PU surfaces can be modified using polybetaine polymers, such as

carboxybetaine, phosphobetaine and carboxybetaine. (33) Furthermore, PSBMA can

be also applied to endotracheal tubes, orthopedic devices and urinary catheters. (43,48)

Page 30: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

28

4.1.4 Poly(2-oxazoline)s

Poly(2-oxazoline)s (POXs) (Figure 7), such as poly(2-ethyl-2-oxazoline) (PEOXA),

poly(2-methy-2-oxazoline) (PMOXA) and poly(2-phenyl-2-oxazoline) (PPOXA) have

been investigated as PEG alternatives. (22)

POXs and PEG demonstrate similar antifouling properties if the optimum density of

the brush is well chosen. However, POXs have numerous advantages such as lower

viscosity, higher stability, and a less demanding synthesis. Manny surface attachment

techniques are available, such as “grafting from” and “grafting-to”. The surface

immobilization process plays a significant role in the stability and efficiency of the

coating. Two types of POXs coatings can be distinguished, depending on the structure

of the used polymer and the immobilization method (e.g. brush-like POXs or linear

POXs). Various parameters influence the properties of the coatings, such as side chain

grafting density, the film thickness and the surface density. (49,50)

The non-adhesive properties of PMOXA was compared to PEG in a study performed

by Moeller and co-workers. (51) They used niobium surfaces coated with poly(L-

lysine)-graft-poly (ethylene gylcol) (PLL-g-PEG) (2kDa) and poly(L-lysine)-graft-

poly(2-methyl-2-oxazoline) (PLL-g-PMOXA) (5kDa) of optimal grafting densities (α

= 0.29 and 0.22, respectively) and assessed the bacterial attachment of E. coli at 37 ºC

in a physiological buffer. The result demonstrated that bacterial attachment was reduced

by 99% in PLL-g-PEG or PLL-g-PMOXA coated surfaces compared to PLL coated

surfaces and uncoated surfaces. Thus, they concluded that both PMOXA and PEG-

based surface coatings could prevent bacterial attachment to a similar extent. (51)

Other authors have been investigated the antifouling properties of PMOXA. The

microbial adhesion of two genetically engineered E. coli strains to poly(L-lysine)-graft-

poly(2-methyl-2-oxazoline) (PLL-g-PMOXA) in niobium surfaces, was assessed by

Pidhatika et al (52). The results showed that dense short brushes (grafting density =

Figure 7. Structure of Poly(2-oxazoline)s

Page 31: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

29

0.33) decrease bacterial adhesion, in greater extent, compared to PPL-g-PMOXA with

different grafting density and uncoated surfaces. This copolymer reduced adhesion of

fimbriated and non-fimbriated E. coli bacteria in both low and high (physiological

level) ionic strength however, less dense brushes did not prevent bacterial adhesion to

the same level. (52)

In another study, Pidhatika et al (50)., compared the stability of the PMOXA coating

with PEG coating in niobium oxide surfaces. In a way to perform a direct comparison,

they selected a PLL-PMOXA and PLL-PEG graft copolymers. The antifouling

properties were assessed from protein resistance test, before and after stability tests.

They concluded that PMOXA copolymer was more stable than PEG copolymer and

kept their antifouling properties under different environmental conditions (oxidative

solution, solution that mimics the ionic strength of body fluids and a solution that

mimics the physiological solution with oxidative substance). (50)

POXs polymers have been studied for biomedical applications and could, ultimately,

be applied to medical devices such as catheters, prosthesis, implants and wound

dressings. (53,54)

Page 32: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

30

4.1.5 Poly (hydroxyfunctional acrylates)

Hydroxyfunctional acrylate, such as Poly (N-hydroxyethylacrylamide) (PHEAA),

PHEMA and poly N-(2-Hydroxypropyl methacrylamide) (PHPMA) (Figure 8) are

neutral, hydroxyl-rich monomers and have been studied as antifouling coatings. (26,55)

The antifouling properties of these polymers have been determined in different studies.

Zhao et al. (55), developed a PHEMA and PHPMA polymers brushes grafted on gold

surfaces via surface-initiated ATRP. The stability of these polymers was determinated

in function of incubation time in PBS. PHEMA polymer with ~32 nm of film thickness,

remained practically unchanged in 20 days, demonstrating a very high structural

stability. PHPMA polymer also showed a very high structural stability in 40 days in

PBS. The protein adsorption from single-protein solution (i.e. Fg, BSA, and Lyz in PBS

buffer) was also investigated and the results showed that PHEMA and PHPMA brushes

have low protein adsorption. Additionally, they also investigated the protein adsorption

from human blood plasma and serum. The results of surface plasma resonance (SPR)

showed that these polymers brushes have very low fouling to 10% human blood plasma

and serum. However, PHPMA only exhibit low fouling protein with appropriated film

thickness. Antiadhesive properties were evaluated by short-term static bacteria

adhesion. Thus, bacterial adhesion of Cytophaga lytica to PHEMA and PHPMA, after

2 h at 25 ºC, was measured. The results demonstrated that both PHEMA and PHPMA

had very low bacteria adhesion as compared to the bacterial attachment on controls (i.e.

PSA (poly(3-sulfopropyl methacrylate potassium), PSBMA and bare gold). Antifouling

properties of PHEMA were better than PHPMA and this could be explain by the fact

that PHEMA surface was more hydrophilic that PHPMA surface. (55)

Figure 8. Structure of pHEAA, pHEMA and pHPMA, respectively

Page 33: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

31

In another work, Zhao et al. (56), developed a PHEAA grafted on gold surfaces by

ATRP and studied the antifouling properties in different complex biological media,

such as undiluted human blood plasma and serum, different single protein solutions,

and bacteria. They studied the resistance of nonspecific protein adsorption from

undiluted serum and plasma on PHEAA brushes with different film thickness by SPR

spectroscopy. The results showed that adsorption was practically undetectable on the

PHEAA brush at a film thickness of 12 nm (selected to the bacterial adhesion test). To

assessed bacterial attachment resistance of PHEAA brushes two bacterial strains were

used, E. coli and S. epidermidis, in a static bacterial adhesion assay. They also studied

the adhesion of these two strains in PHEMA and PHPMA, for positive control and bare

gold for negative control. The results demonstrate that attachment of S. epidermidis on

PHEAA, after 66 h, was reduced in approximately 59% compared to PHEMA and

PHPMA. The adhesion of E. coli on PHEAA was reduced 79%, compared to PHEMA

and PHPMA brushes. The author concluded that PHEAA brushes had better and

extended resistance to both protein adsorption and bacterial adhesion (approximately 3

days) when compared to PHEMA and PHPMA brushes. The better antifouling

properties of PHEAA could be due to the fact that this polymer contains more

hydrogen-bond donors which enhance surface hydration. This was confirmed by water

contact angle test, which demonstrate that PHEAA was more hydrophilic that PHEMA

and PHPMA (water contact angle was 15º, 31, 33º, respectively.) (56)

Poly (hydroxyfunctional acrylate) can be grafted onto various inorganic substrates, such

as silica, quartz, graphite and silver. These materials have been widely used in implants,

tissue engineering scaffolds and contact lenses. PHEMA can also be used in artificial

organs, blood-contacting implants, contact lenses and intraocular lenses (IOLs).

(22,37,56)

PHEMA hydrogel are used to coat ventricular catheters, since they reduce cell

attachment, in vitro, when compared to uncoated catheters. (57)

PHEMA hydrogel can also be used to entrap nebulized antimicrobial solutions or as a

drug delivery system in PVP endotracheal tubes and urinary catheters. (58,59)

Page 34: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

32

4.1.6 Polyacrylamide

Polyacrylamide (PAAm) (Figure 9) is a stable, hydrophilic, biocompatible, electrically

neutral and polar polymer. (12,60) PAAm can form low-fouling surfaces due to the

surface hydration layer formed by hydrogen bond between water and amide groups.

(61)

The antifouling properties of PAAm polymers has been investigated in different

studies. One of these studies was performed by Fundeanu et al (62)., that assessed the

antifouling properties of amino-poly(o-amino-p-xylylene-co-p-xylylene) (PPX)-

PAAm brush grafting to silicone rubber by ATRP. The bacterial adhesion was studied

using two bacterial strains, S. aureus and E. coli, in a parallel plate flow chamber. The

results demonstrated that initial deposition rates and adhesion were greatly reduced for

both strains in amino-PPX-PAAm brush-coated silicone rubber compared to uncoated

silicone rubber. After 4 h, the bacterial adhesion of E. coli and S. aureus on coated

silicone rubber was reduced in 99% and 93%, respectively, compared to adhesion on

uncoated silicone rubber. (62)

In another study, a PAAm brush coating grafting on silicone rubber surfaces (water

contact angle of 109º) by ATRP and the efficacy of the coating was evaluated against

three microbial strains (i.e. S. aureus, S. salivarius and C. albicans), by parallel plate

flow adhesion. The functionalization of the silicone rubber surfaces with PAAm

allowed to create a more hydrophilic surface (water contact angle of 28º). The results

showed that microbial adhesion on coated surfaces was reduced when compared to

uncoated silicone rubber. Adhesion of S. aureus, S. salivarius and C. albicans was

reduced in 58%, 52% and 77%, respectively. The detachment of all adhering bacteria

and yeast from the brush-coating confirmed the weak adhesive forces between adhering

microorganisms and the coating. This coating had good hydrolytic stability and did not

degrade upon incubation in saliva for one month. (63)

Figure 9. Structure of Polyacrylamide

Page 35: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

33

The influence of PAAm polymers at inhibiting protein adsorption and cell adhesion was

also assessed. For this, Liu et al. (61), grafted PAAm on gold surfaces by surface-

initiated ATRP and tested protein adsorption, cell adhesion and bacterial attachment.

The water contact angle of PAAm grafted surfaces was 14.8º. The protein adsorption

resistance was assessed by SPR, with three model proteins, Fg, BSA and Lyz. The

results showed that PAAm-grafted surfaces resisted to protein adsorption, compared to

uncoated gold surfaces. The results also demonstrated that PAAm surfaces suppressed

protein adsorption in a similar way to PEG or pSBMA. The PAAm surface also resisted

to protein adsorption from serum and plasma and cell attachment. The bacterial

attachment was assessed with two bacterial strains, P. aeruginosa and S. epidermidis.

Results demonstrated that PAAm-grafted surfaces resisted to the initial bacterial

attachment, leading to more than 97% reduction for both strains compared to uncoated

gold surfaces. (61)

PAAm brushes have good hydrolytic stability, can resist to protein adsorption, cell

adhesion, bacterial attachment and are stable on physiological fluids. (61,63,64)

Some authors have reported studies of PAAm grafted to silicone rubber surface with

promising results for in vivo applications. Silicone rubber is widely used as a biomedical

polymer for intravenous and urinary catheters, voice prostheses, oxygenators and

contact lenses. (60,62,63)

Page 36: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

34

4.1.7 Heparin

Heparin (HP) (Figure 10) is a naturally occurring polysaccharide and consists of two

sulfated sugar monomers, which occur as repeating disaccharides units. (65)

Heparin has been used as an antiadhesive coating, since it can increase hydrophilicity

by formation of a hydrated layer between the surface and the bacteria. It also used to

increase the blood compatibility of medical devices due it’s anticoagulants properties.

The exposure of biomaterials to physiologic fluids cause the activation of blood defence

mechanisms leading to activation of the coagulation cascade, cellular inflammatory

mechanisms, complement system and platelets. These activations may compromise the

performance of medical devices. (65,66)

The influence of HP on the biding of S. epidermidis to fibronectin and consequently

their influence in biofilm formation was investigated by Arciola et al. (66) by dynamic

force spectroscopy (DFS). They investigated the attachment of S. epidermidis to a

fibronectin-functionalised gold-plated disk, incubated with HP solution or with

antihuman fibronectin monoclonal antibody solution. Results showed a decrease of

bacteria adhered to disk surfaces, in both cases. So, HP specifically inhibiting the

interaction of S. epidermidis adhesins and fibronectin. (66)

This influence was also tested by Lundberg and co-authors in IOLs under in vitro flow

conditions. The IOLs, fabricated with PMMA, with or without HP coating, were

incubated with human cerebrospinal fluid (CSF) for 1h or with CSF plus 0.50% plasma

for 12h. In this study two S. epidermidis strains were used, and bacterial adhesion was

assessed by bioluminescence. The results demonstrated that bacterial adhesion was

decreased on IOLs coated with HP compared to uncoated IOLs, with both strains, after

incubation with CSF plus 0.50% plasma for 12h. So, results suggested that IOLs with

HP coating were less predisposed to bacterial adhesion than uncoated IOLs. (67)

García-Sáenz et al. (68), also studied the in vitro adhesion of S. epidermidis to IOLs.

Figure 10. Structure of Heparin

Page 37: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

35

They used two strain of S. epidermidis, slime-positive and slime-negative and compared

its adhesion to IOLs of PMMA, silicone, acrylic and HP-modified PMMA. The results

showed that slime-positive strains adhered at a higher level than slime-negative strains.

The results also demonstrated that bacterial adhesion to HP coated PMMA was lower

than adhesion to uncoated PMMA, because this coating formed a hydrated surface that

reduced bacterial adhesion. (68)

Heparin coating can be applied on diverse surfaces, used in medical devices, such

polyvinylchloride, polymethylmethacrylate, polyurethane, polyethylene and silicone.

This coating has been studied to reduce bacterial adhesion to catheters, artificial lenses,

and vascular grafts of PU. Heparin can also be used in orthopaedic devices, such

fracture fixation, knee replacements, tendon and ligament reconstruction and others.

(19,33,69)

Heparin-coated blood contacting medical devices are widely used in a clinical

application and can be divided into eluting and non-eluting technologies. These

technologies can be applied to medical devices such as cardiopulmonary bypass,

catheters, haemodialysis catheters, vascular grafts, extracorporeal circulation devices,

coronary stents, stents-grafts and others. (65)

Page 38: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

36

4.1.8 Phosphorylcholine

Phospholipid polymers with phosphorylcholine (PC) (Figure 11) head groups are

appropriate for biomedical use, once they are biocompatible and can inhibit bacteria

and proteins adhesion.Figure 11 Phospholipids coating can also reduce platelets

adherence once theirs structure mimics biological cell membranes. These polymers

have antifouling properties and decrease thrombus formation by the formation of a

hydration layer. (70)

Polymers with phosphorylcholine have demonstrated to increase the hydrophilicity of

silicone elastomers (SE), (e.g. the water contact angle of unmodified SE was 100.8º

whereas, water contact angle of 2-methacryloyloxyethyl phosphorylcholine (MPC) -

modified SE was 20º). Additionally, this coating also showed to increase the

hemocompatibility of SE once it could reduce protein adsorption and platelets

adherence. (70)

The antifouling properties of phospholipids polymers surface was studied by Patel et

al. (71), under dynamic flow conditions, in PBS and 20% human serum. Thus,

polyethylene terephthalate (PET) surfaces were coated with Poly [(ω-

methacryloyloxyalkyl phosphorylcholine-co-n-butyl methacrylate)] (MAPC-co-

BMA), with two different methylene chain length. The results showed that

phospholipid polymer surfaces in PBS significantly reduced bacterial adhesion of S.

epidermidis when compared to uncoated PET surfaces. They also demonstrated that the

length of the methylene chain on MAPC unit also influenced bacterial adhesion since a

shorter chain length was more effective at reducing bacterial adhesion. Bacterial

adhesion in the presence of 20% human serum was markedly reduced to <10% when

compared to PBS. (71)

Other studies have been realized to assess the antifouling properties of phospholipids

polymers. One example was the work performed by Fujii et al. (72), that have

investigated the effect of poly (MPC-co-n-butyl methacrylate) (PMB) coating grafted

Figure 11. Structure of Phosphorylcholine

Page 39: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

37

on stainless steel plates by radical polymerization, on bacterial adhesion. The formation

of a biofilm of S. aureus, S. epidermidis and P. aeruginosa was assessed by the

observation with a culture experiments for confocal laser scanning microscope (CLSM)

and the number of bacteria was measured by ATP (Adenosine 5’- triphosphate) assay.

The results demonstrated that PMB-coated surfaces reduced bacterial adhesion of the

three strains under study when compared to uncoated surfaces. Additionally, to the

resistance of the bacteria enveloped in the biofilm antibiotics were added. The

application of antibiotic decreased the number of S. epidermidis and S. aureus on both

uncoated and coated surfaces. In the case of P. aeruginosa, the addition of gentamicin

decreased the number of adhered bacteria whereas, the addition of cefazolin increased

that number on both coated and uncoated surfaces. This variance is explained by the

fact that P. aeruginosa was not susceptible to cefazoline. Thus, they conclude that PMB

coating prevent biofilm formation in in vitro conditions. (72)

The influence on biofilm formation was also tested by Hirota and co-authors (73), that

have studied the influence of MPC polymers on adherence of 4 microbial strains, S.

aureus, S. mutans, P. aeruginosa and C. albicans, to PET surfaces. The results

demonstrated that bacterial adhesion of all four strains, after 1h of incubation, were

significantly lower in MPC-coated surfaces (water contact angle = 10º) compared to

uncoated surfaces (water contact angle = 72º). Thus, application of an MPC coating on

medical devices surfaces could prevent microbial retention and biofilm formation. (73)

The PC coating can be used for medical devices such bone fixation devices, implantable

artificial hearts, artificial blood vessels, coronary stents, vascular grafts of PU, artificial

lungs, intravascular stents, intravascular guide wires, oxygenators and soft contact

lenses. (33,72–74) These coating can also be used in urological devices, such urinary

catheters and ureteral stents. (48,75)

Page 40: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

38

4.2 Antifouling and antimicrobial surfaces

In some situations, the conjugation of antimicrobial and antifouling properties may be

advantageous. Since an antifouling coating cannot completely prevent initial

microorganism attachment, a few adhering microorganisms cells can ultimately form a

mature biofilm. (12) An ideal antibacterial surface should be capable of preventing

initial attachment, kill all microorganism that can attach and lastly remove dead

microorganisms. (38)

4.2.1 PEG-Antibiotics

Hydrophilic polymers like PEG are commonly used as spacers for the immobilization

of bioactive molecules such antibiotics to create surfaces with both antifouling and

antimicrobial properties. PEG is one of the most selected antifouling polymers and is

also used to immobilize several antibiotics, such penicillin (PEN), gentamicin (GEN)

and ampicillin (AM), on polymeric substrates. (38)

One example is the modification of ePTFE (expanded polytetrafluoroethylene) surfaces

with penicillin using PEG as a spacer. To evaluated antibacterial properties of the

coating, ePTFE surfaces were immersed into bacterial cultures of P. aeruginosa and S.

aureus, for 3-4 h at 37 ºC. The antibacterial activity was evaluated by measuring the

absorbance by ultraviolet-visible (UV-VIS) spectrometer. The results showed that %

absorbance for S. aureus was lower in the presence of the PEN-PEG-MA-ePTFE

compared to uncoated ePTFE, PEG-MA-ePTFE or PEN-ePTFE. Nevertheless, for P.

aeruginosa % absorbance in presence of PEN-PEG-MA-ePTFE was higher than in

presence of uncoated ePTFE. They concluded that PEN-PEG coating is highly effective

against S. aureus. (76)

In another study, an antibacterial effectiveness of this coating was determined. The

same authors incubated ePTFE, maleic anhydride (MA)-ePTFE, PEG-MA-ePTFE,

PEN-PEG-MA-ePTFE, and PEN-ePTFE surfaces into S. aureus culture, at 37 ºC, for

3, 6, 9, 12 and 24 h. At those times, an aliquot was collect and spread on agar plates to

grow the colonies. After incubation CFUs were determined. This study demonstrated

that antimicrobial efficiency of PEN-PEG-MA-ePTFE surface remained substantial

after 24 h of exposure to S. aureus. They also monitored the ester linkages between

PEG and PEN by ATR-FTIR spectrometry to assessed the hydrolytic stability of the

Page 41: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

39

coating. The results revealed that this coating was still effective even with a 32% loss

of PEN. (77)

The attachment of penicillin to ePTFE is only effective against gram-positive bacteria.

To create an antibacterial coating that inhibits proliferation of an extensive range of

bacteria, Aumsuwan et al., developed a PEG-ampicillin coating. Ampicillin is a broad-

spectrum antibiotic with activity against gram-positive and gram-negative bacteria. To

determine an antibacterial effectiveness of this coating, AM-PEG-MA-ePTFE surfaces

are immersed into bacterial cultures of gram-positive bacteria (i.e. S. aureus, E. faecalis,

B. thuringiensis,) and gram-negative bacteria (i.e. P. putida, S. enterica and E. coli) at

37 ºC for 5 h. Antibacterial properties were assessed by measuring the optical density

(OD). The results demonstrates that AM-PEG-MA-ePTFE surface reduces bacterial

grown when compared to ePTFE, MA-ePTFE and PEG-MA- ePTFE in all bacterial

strains. Thus, attachment of ampicillin prevented the development of microbial films.

(78)

Another different approach can be the attachment of two different antibiotics to surfaces

(e.g. polypropylene (PP)). Thus, is possible eliminate the growth of gram-positive and

gram-negative bacteria, simultaneously. PEN-PEG-MA-PP and GEN-PEG-MA-PP are

effective against S. aureus and S. putida, respectively. An GEN/PEN-PEG-MA-PP

surface was developed and was possible to create an antibacterial PP surfaces with

different antimicrobial strengths, by altering the antibiotics concentration. (79)

Expanded polytetrafluoroethylene is a synthetic polymer widely used in medical

devices such as mitral valve tendon replacements, vascular grafts and soft tissue in

plastic and reconstructive surgeries. (76)

Page 42: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

40

4.2.2 Heparin/Chitosan multilayer film

Chitosan (CHT) is a natural biocompatible cationic antimicrobial molecule. The

mechanism of antibacterial action of CHT is not completely understood. However, it is

probably related to the interactions between negatively charged microbial cells

membranes and positively charged CHT. These interactions result in alterations of the

cell surface, causing leak of intracellular components of microbial cells that leads to

cell death. (38,80–82) As describe above, HP is an anionic antifouling molecule with

antiadhesive and anticoagulants properties. The construction of multilayer film with

these two polysaccharides can form a film that combine antifouling and antimicrobial

properties. (82)

A heparin/chitosan multilayer can be prepared via layer-by-layer (LbL) technique,

resulting in a coating with antifouling and antimicrobial properties. (38,82,83)

Several studies about the antimicrobial properties of this multilayer films have been

reported. Fu et al. (82), developed a multilayer HP/CHT grafter into PET surfaces. The

initial adhesion of E. coli on PET substrates was determinate by SEM. PET substrates

were immersed in an E. coli suspension for 4 h at 37 ºC. The results showed that the

number of bacteria adhered onto HP/CHT multilayer films was lower than onto the

uncoated PET. The in vitro antibacterial activity of the multilayer film was assessed

and the results revealed that the coating could kill the bacteria successfully. The authors

observed that 46%-68% of the microbial cells were no longer viable after 7 h of

exposure to the multilayer-modified PET films and that this reduction was enhanced

with time. On the other hand, the number of viable cells only decreased 7% after 7 h

of contact with PET films. The surface properties of this multilayer film was pH

dependent, once pH influences the amount of chitosan on the surface. The number of

viable bacteria at pH 3.8, 2.9 and 6.0 was 68%, 58% and 46%, respectively. (82)

Another studied was performed by Follmann and co-workers (84), that developed a

multilayer film by LbL technique of N-Trimethyl chitosan (TMC)/HP grafting on

polystyrene (PS) chemically modified surfaces. TMC is a quaternary polycation

obtained from the N-methylation of CHT and have similar properties to the CHT but, a

higher antimicrobial effect. TMC/HP multilayer with different quaternization degree

(TMC20 and TMC80) and PS-modified surfaces (control) were incubated in a bacterial

Page 43: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

41

suspension of E. coli for 4 h at 37 ºC. The initial adhesion was tested and the results

showed that bacterial cells was only detected in the control, once this surface has low

hydrophilicity compared to TMC/HP multilayer surfaces. The in vitro antibacterial

activity of the both TMC/HP multilayer prepared in this study was compared.

TMC80/HP assembled at pH 3.0 exhibited no significant influence on the viability of

bacterial cells as compares to control. However, TMC20/HP assembled at pH 3.0 and

TMC80/HP prepared at pH 7.4 it reduced the number of viable-cells in 32.7% and

64.6%, respectively. TMC80/HP assembled at pH 7.4 showed good antibacterial

properties, so it is possible to use it as an antimicrobial coating. (84)

Wang et al., development a (heparin/chitosan)10− (poly(vinylpyrrolidone)/poly (acrylic

acid))10 [(HP/CHT)10−(PVP/PAA)10] multilayer film by LbL self-assembly. The

multilayer film was formed by a top-down degradable PVP/PAA film that can resist

bacterial attachment for 24 h. The multilayer film is also constituted by a subjacent

HP/CHT film, with antibacterial properties. The antimicrobial properties of the

multilayer film were assessed by water-borne assay with S. aureus. Some samples were

chosen for the assay: silicon wafer, (HP/CHT)10 multilayer film, (HP/CHT)10-

(PVP/PAA)10 multilayer film that thermal cross-linked at 170 °C for 4 h and

(HP/CHT)10-(PVP/PAA)10 multilayer film that thermal cross-linked at 110 °C for 16 h.

These substrates were exposed to an S. aureus suspension. In the case of the

(HP/CHT)10 multilayer film, a few bacteria were observed on the surface after 24 h.

The (HP/CHT)10−(PVP/PAA)10 multilayer film with thermal cross-linked at 110 °C for

16 h, exhibited few bacteria adhered at 24 h. On the other hand,

(HP/CHT)10−(PVP/PAA)10 multilayer films that were cross-linked at 170 °C for 4 h,

exhibited several bacteria adhered. After 24 h, the top-down was completely degraded

and the (HP/CHT)10 was exposed, providing a contact-killing surface. To assess it, the

(HP/CHT)10−(PVP/PAA)10 multilayer film cross-linked at 110 °C for 16 h and then

degraded at 37 °C. After 24 h, the multilayer film and bare glass (control) was exposed

to S. aureus (previously stained). Fluorescent microscopy images demonstrate that the

bacteria on uncoated glass were alive. However, almost all of the adhered bacteria on

the multilayer film had been killed (85)

The HP/CHT multilayer film could be potentially applied to medical devices such as

cardiovascular implants. (82) On the other hand, [(HP/CHT)10−(PVP/PAA)10]

multilayer film has potential to be applied in implants. However, this films can only be

Page 44: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

42

applied into medical devices that can be thermally treated at a high temperature such as

bone nails and steel stents. (85)

4.2.3 MEO2MA based copolymer

Copolymer brushes based on 2-(2-methoxyethoxy)ethyl methacrylate (MEO2MA) have

also antifouling properties. Furthermore, these copolymers have reactive hydroxyl

groups which allowed controlled loading of antimicrobial peptides. Glinel et al.,

prepared copolymer brushes based on MEO2MA and oligo(ethylene glycol)

methacrylate (OEGMA). This copolymer was prepared by surface-initiated ATRP, on

silicon surfaces, and was subsequently functionalized by magainin I, a natural

antibacterial peptide. The antibacterial activity of this coating was tested against two

bacterial strains (i.e. L. ivanovii and B. cereus). Coated silicon surfaces were incubated

in bacterial suspension for 3 h and observed by optical microscopy. The results revealed

the absence of bacteria for both strains on the nonfunctionalized copolymer brushes.

However, a few bacteria were detected on functionalized brushes (coverage did not

exceed 1% of the surface). These results demonstrated that bacterial cells interacted

with magainin I peptide. CLSM images showed that the adhered cells (previously

stained) corresponding to dead bacteria. Thus, the antifouling properties of the brushes

combined with the antimicrobial peptide lead to a delay of bacterial biofilm formation.

(20)

The antibacterial effect of these coatings was also assessed by Blin et al., that developed

hydrophilic copolymer brush (poly-(MEO2MA-co-HOEGMA)) grated to silica-based

microparticles by surface-initiated ARTP, subsequently functionalized with magainin

I. The antibacterial effect was assessed against L. ivanovii by incubating the modified

surface in a bacterial suspension for 3 h at 25 ºC. The viability of the cells, previously

stained, was assessed by CLSM and fluorescence microscopy. The observations reveal

that most of the cells adhered to coated silica were dead. (86)

These copolymers can be applied to the biomedical field, in particular to implants. (20)

Page 45: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

43

5. Discussion

Medical device-associated infections represent a significant risk for patient’s health.

Thus, different strategies have been used to prevent these infections. One of these

strategies is the surface modification of biomaterials with antifouling polymers.

PEG is the most studied and used antifouling polymer, due to its noncytotoxic,

nonimmunogenic and biocompatibility properties. One advantage of PEG is that this

polymer can be applied to varied biomaterials and can be used as antifouling polymer

in orthopedic and dental implants, catheters of PU, among others. However, has been

reported that PEG has reduced stability due to its oxidative degradation in vivo. Despite

PEG representing the gold standard in this respect, it not appropriate for a long-term

application. Thus, it is necessary investigate new polymers and materials with

antifouling properties, biocompatible and stables at physiological conditions. Examples

of other polymers, that have been studied for their antifouling applications, can be PVP,

POXs and PAAm. (22,50,52,87)

Medical devices with heparin coatings are already used, namely in blood-contacting

devices. POXs polymers have demonstrated antifouling properties similar to PEG but,

a higher stability and the possibility of keeping their antiadhesive properties even in

physiological environments. This polymer can be used in some medical devices such

catheters, prosthesis and others. PC coating it widely used in medical devices, for

example was used for developed glaucoma drainage device, intraocular lenses, contact

lenses (omafilcon A) based on the copolymers of PHEMA and MCP. PC polymers can

also be applied in coronary stents, ventricular assist device and vascular grafts. This

polymer also has been studied for orthopedic devices usage. PSBMA polymers have

been applied to endotracheal tubes, orthopedic devices and peripherally inserted central

catheters (FDA approval). (43)

Theses polymers have demonstrated to be able to reduce biofilm formation of some of

the most common microorganisms associated with HCAIs, such E. coli, S. aureus,

among others. Additionally, polymers like PEG and PAAm are also able to reduce

biofilm formation of yeast. (30,51,63,72)

There are limitations to compared these different strategies and assessed the best one

since the tests are performed under different conditions. Furthermore, most of the

Page 46: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

44

studies only focus the initial stage of the biofilm formation. Thus, a standardized test is

essential, in order to directly compare the studies. (18,19)

Despite the good results obtained under in vitro conditions, there are difficulties in

demonstrating the effectiveness of these surfaces under real clinical conditions. The in

vitro test does not reflect the environment that medical device will be exposed.

Furthermore, there are not enough studies for testing the stability, toxicity, and safety

of these polymers under in vivo conditions. Thus, it necessary an in vivo test for an

appropriate study of the medical device. These difficulties create limitations in

correlating the in vitro, in vivo preclinical and clinical data. (12,18,19)

A conjugation of antifouling and antimicrobial surfaces can be a promising approach,

once an antifouling coating cannot completely prevent biofilm formation. Ideally, these

surfaces are able to prevent initial microorganism attachment, kill microorganism that

might attach and remove the dead microorganisms. (12)

Page 47: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

45

6. Conclusion

HAIs are a major clinical problem with significant impact on mortality and morbidity.

The best approach against such infection is the prevention of biofilm formation.

Consequently, several surface modification techniques have been developed. Polymers

such PEG, PVP, polybetaines and others have demonstrated good anti-adhesive

properties under in vitro condition. However, in some cases it is necessary studies to

assess their efficacy under physiological conditions. These antifouling coatings are

different from each other and have advantages and disadvantages. Thus, the type of

coating should be adapted for a specific application. Some of these polymers are already

applied in medical devices such catheters, orthopaedic implants, dental implants,

vascular grafts, contact lenses and other.

Despite these polymers being able to reduce microorganism attachment on medical

device surfaces, a few adhered bacteria can eventually form a biofilm. Therefore, the

association of theses coatings with antimicrobial agents is an interesting approach.

Page 48: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

46

7. Bibliography

1. World Health Organization. Health care-associated infections Fact sheet. World

health Organization. 2015.

2. Percival SL, Suleman L, Donelli G. Healthcare-Associated infections, medical

devices and biofilms: Risk, tolerance and control. J Med Microbiol.

2015;64(4):323–34.

3. WHO. Guidelines on Core Components of Infection Prevention and Control

Programmes at the National and Acute Health Care Facility Level. 2016. 1-90 p.

4. European Centre for Disease Prevention and Control. Annual epidemiological

report 2014. Antimicrobial resistance and healthcare-associated infections. 2015.

5. Desrousseaux C, Sautou V, Descamps S, Traoré O. Modification of the surfaces

of medical devices to prevent microbial adhesion and biofilm formation. J Hosp

Infect. 2013;85(2):87–93.

6. von Eiff C, Jansen B, Kohnen W, Becker K. Infections Associated with Medical

Devices. Drugs. 2005;65(2):179–214.

7. Darouiche RO. Device‐Associated Infections: A Macroproblem that Starts with

Microadherence. Clin Infect Dis. 2001;33(9):1567–72.

8. von Eiff C, Kohnen W, Becker K, Jansen B. Modern strategies in the prevention

of implant-associated infections. Int J Artif Organs. 2005;28(11):1146–56.

9. Kostakioti M, Hadjifrangiskou M, Hultgren SJ. Bacterial biofilms: development,

dispersal, and therapeutic strategies in the dawn of the postantibiotic era. Cold

Spring Harb Perspect Med. 2013;3(4):1–23.

10. Srivastava S, Bhargava A. Biofilms and human health. Biotechnol Lett.

2016;38(1):1–22.

11. Thebault P, Jouenne T. Antibacterial coatings. 2015.

12. Neoh KG, Li M, Kang E-T, Chiong E, Tambyah PA. Surface modification

strategies for combating catheter-related complications: recent advances and

challenges. J Mater Chem B Chem B. 2017;5(11):2045–67.

13. Simões NG, Bettencourt AF, Monge N, Ribeiro IAC. Novel Antibacterial

Page 49: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

47

Agents: An Emergent Need to Win the Battle Against Infections. Mini-Reviews

Med Chem [Internet]. 2017;17(14):1–13. Available from:

http://www.eurekaselect.com/145452/article

14. Davies D. Understanding biofilm resistance to antibacterial agents. Nat Rev

Drug Discov. 2003;2(2):114–22.

15. Harding JL, Reynolds MM. Combating medical device fouling. Trends

Biotechnol. 2014;32(3):140–6.

16. Hasan J, Crawford RJ, Ivanova EP. Antibacterial surfaces: The quest for a new

generation of biomaterials. Trends Biotechnol. 2013;31(5):295–304.

17. Pontes C, Alves M, Santos C, Ribeiro MH, Gonçalves L, Bettencourt AF, et al.

Can Sophorolipids prevent biofilm formation on silicone catheter tubes? Int J

Pharm. 2016;513(1):697–708.

18. Salwiczek M, Qu Y, Gardiner J, Strugnell RA, Lithgow T, Mclean KM, et al.

Emerging rules for effective antimicrobial coatings. Trends Biotechnol.

2014;32(2):82–90.

19. Campoccia D, Montanaro L, Arciola CR. A review of the biomaterials

technologies for infection-resistant surfaces. Biomaterials. 2013;34(34):8533–

54.

20. Glinel K, Jonas AM, Jouenne T, Leprince J, Galas L, Huck WTS. Antibacterial

and antifouling polymer brushes incorporating antimicrobial peptide. Bioconjug

Chem. 2009;20(1):71–7.

21. Ferreira P, Alves P, Coimbra P, Gil MH. Improving polymeric surfaces for

biomedical applications: a review. J Coatings Technol Res. 2015;12(3):463–75.

22. Lowe S, O’Brien-Simpson N, Connal L. Antibiofouling polymer interfaces:

poly(ethylene glycol) and other promising candidates. Polym Chem. 2014;

23. Casalini S, Bortolotti CA, Leonardi F, Biscarini F. Self-assembled monolayers

in organic electronics. Chem Soc Rev. 2017;46(1):40–71.

24. Hasan A, Pandey M. L. Review: Polymers, Surface-Modified Polymers, and Self

Assembled Monolayers as Surface-Modifying Agents for Biomaterials. Polym

Plast Technol Eng. 2015;54(13):1358–78.

Page 50: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

48

25. Kim W, Jung J. Polymer brush: A promising grafting approach to scaffolds for

tissue engineering. BMB Rep. 2016;49(12):655–61.

26. Krishnamoorthy M, Hakobyan S, Ramstedt M, Gautrot JE. Surface-initiated

polymer brushes in the biomedical field: Applications in membrane science,

biosensing, cell culture, regenerative medicine and antibacterial coatings. Chem

Rev. 2014;114(21):10976–1026.

27. Mei HC Van Der, Lochhead MJ, Ferna ICS, Grainger DW, Busscher HJ. The

inhibition of the adhesion of clinically isolated bacterial strains on multi-

component cross-linked poly (ethylene glycol)-based polymer coatings.

Biomaterials. 2007;28:4105–12.

28. Roosjen A, Busscher HJ, Norde W, Mei HC Van Der. Bacterial factors

influencing adhesion of Pseudomonas aeruginosa strains to a poly ( ethylene

oxide ) brush. Microbiology. 2006;152:2673–82.

29. Harbers GM, Emoto K, Greef C, Metzger SW, Woodward HN, Mascali JJ, et al.

Functionalized poly(ethylene glycol)-based bioassay surface chemistry that

facilitates bio-immobilization and inhibits nonspecific protein, bacterial, and

mammalian cell adhesion. Chem Mater. 2007;19(18):4405–14.

30. Roosjen A, Kaper HJ, Mei HC Van Der, Norde W, Busscher HJ. Inhibition of

adhesion of yeasts and bacteria by poly ( ethylene oxide ) -brushes on glass in a

parallel plate flow chamber. Microbiology. 2003;149:3239–46.

31. Roosjen A, Mei HC Van Der, Busscher HJ, Norde W. Microbial Adhesion to

Poly ( ethylene oxide ) Brushes : Influence of Polymer Chain Length and

Temperature. Langmuir. 2004;(17):10949–55.

32. Nejadnik MR, Mei HC Van Der, Norde W, Busscher HJ. Bacterial adhesion and

growth on a polymer brush-coating. Biomaterials. 2008;29:4117–21.

33. Adipurnama I, Yang M, Butruk-raszeja B. Surface modification and

endothelialization of polyurethane for vascular tissue engineering applications:

a review. Biomater Sci. 2017;5:22–37.

34. Liu X, Chu PK, Ding C. Surface modification of titanium, titanium alloys, and

related materials for biomedical applications. Mater Sci Eng. 2005;47:49–121.

35. Buxadera-palomero J, Calvo C, Torrent-camarero S, Gil FJ, Mas-moruno C,

Page 51: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

49

Canal C, et al. Biofunctional polyethylene glycol coatings on titanium: An in

vitro -based comparison of functionalization methods. Colloids Surfaces B

Biointerfaces. 2017;152:367–75.

36. Zhao L, Chu PK, Zhang Y, Wu Z. Antibacterial Coatings on Titanium Implants.

J Biomed Mater Res Part B. 2009;470–80.

37. Luensmann D, Jones L. Protein deposition on contact lenses: The past, the

present, and the future. Contact Lens Anterior Eye. 2012;35:53–64.

38. Yu Q, Wu Z, Chen H. Dual-function antibacterial surfaces for biomedical

applications. Acta Biomater. 2015;16:1–13.

39. Antonelli PJ, Sampson EM, Ojano-dirain C. Biofilm Formation on Silicone

Tympanostomy Tubes With Polyvinylpyrrolidone Coating. Otolaryngol head

neck Surg. 2011;137(1):19–23.

40. Sun M, Wu Q-Y, Xu J, He F, Brown AP, Ye Y. Vapor-based grafting of

crosslinked poly(N-vinyl pyrrolidone) coatings with tuned hydrophilicity and

anti-biofouling properties. J Mater Chem B. 2016;(March).

41. Liu X, Tong W, Wu Z, Jiang W. Poly(N-vinylpyrrolidone)-grafted

poly(dimethylsiloxane) surfaces with tunable microtopography and anti-

biofouling properties. RSC Adv. 2013;3:4716–22.

42. Liu X, Xu Y, Wu Z, Chen H. Poly(N-vinylpyrrolidone)-Modified Surfaces for

Biomedical Applications. Macromol Journals. 2013;12:147–54.

43. Li J, Taylor M, Zhang Z. Anti-fouling Mefical Coatings. In: Antimicrobial

Coatings and Modifications on Medical Devices. 2017. p. 189–214.

44. Gang Cheng, Zheng Zhang, Shenfu Chen, James D. Bryers and SJ. Inhibition of

Bacterial Adhesion and Biofilm Formation on Zwitterionic Surfaces.

Biomaterials. 2007;28(29):4192–9.

45. Cheng G, Li G, Xue H, Chen S, Bryers JD, Jiang S. Zwitterionic carboxybetaine

polymer surfaces and their resistance to long-term biofilm formation.

Biomaterials. 2009;30(28):5234–40.

46. Jiang Y, Rongbing B, Ling T, Jian S, Sicong L. Blood compatibility of

polyurethane surface grafted copolymerization with sulfobetaine monomer.

Page 52: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

50

colloids Surf B - Elsevier. 2004;36:27–33.

47. Xia HanHsiang-Chieh Hung PJ, Sun F, Xu X, Yang W, Bai T, Jiang S.

Sterilization, hydration-dehydration and tube fabrication of zwitterionic

hydrogels. Am Vac Soc. 2017;411:1–8.

48. Singha P, Locklin J, Handa H. A review of the recent advances in antimicrobial

coatings for urinary catheters. Acta Biomater. 2017;50:20–40.

49. Tauhardt L, Kempe K, Gottschaldt M, Schubert US. Poly(2-oxazoline)

functionalized surfaces: from modification to application. Chem Soc Rev.

2013;42:7998–8011.

50. Pidhatika B, Rodenstein M, Chen Y, Rupert T. Comparative Stability Studies of

Poly (2-methyl-2-oxazoline) and Poly (ethylene glycol) Brush Coatings.

Biointerphases. 2012;7.

51. Moeller J, Pidhatika B, Möller J, Vogel V, Konradi R. Nonfouling Surface

Coatings Based on Poly (2-methyl-2-oxazoline). Chimia (Aarau). 2008;62.

52. Pidhatika B, Möller J, Benetti EM, Konradi R, Rakhmatullina E, Mühlebach A,

et al. The role of the interplay between polymer architecture and bacterial surface

properties on the microbial adhesion to polyoxazoline-based ultrathin films.

Biomaterials. 2010;31(36):9462–72.

53. Vasilev K, Macgregor M. Advanced Biomedical Devices Facilitated by Plasma

Deposited Polyoxazoline Coatings. Biostat Biometrics Open Acc. 2017;2(3):1–

5.

54. Ramiasa MN, Cavallaro AA, Mierczynska A, Christo SN, Gleadle JM, Hayball

JD, et al. Plasma polymerised polyoxazoline thin films for biomedical

applications. Chem Commun. 2015;51:4279–82.

55. Zhao C, Li L, Wang Q, Yu Q, Zheng J. Effect of Film Thickness on the

Antifouling Performance of Poly ( hydroxy-functional methacrylates ) Grafted

Surfaces. Langmuir. 2011;27:4906–13.

56. Zhao C, Zheng J. Synthesis and Characterization of Poly( N -

hydroxyethylacrylamide) for Long-Term Antifouling Ability.

Biomacromolecules. 2011;12:4071–9.

Page 53: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

51

57. Hanak BW, Hsieh CY, Donaldson W, Browd SR, Lau KKS, Shain W. Reduced

cell attachment to poly(2-hydroxyethyl methacrylate)-coated ventricular

catheters in vitro. J Biomed Mater Res - Part B. 2017;1–12.

58. McCoy CP, Irwin NJ, Brady C, Jones DS, Carson L, Andrews GP, et al. An

Infection-Responsive Approach to Reduce Bacterial Adhesion in Urinary

Biomaterials. Mol Pharm. 2016;13(8):2817–22.

59. Jones DS, McCoy CP, Andrews GP, McCrory RM, Gorman SP. Hydrogel

Antimicrobial Capture Coatings for Endotracheal Tubes: A Pharmaceutical

Strategy Designed to Prevent Ventilator-Associated Pneumonia. Mol Pharm.

2015;12(8):2928–36.

60. Cringus-fundeanu I, Luijten J, Mei HC Van Der, Busscher HJ, Schouten AJ.

Synthesis and Characterization of Surface-Grafted Polyacrylamide Brushes and

Their Inhibition of Microbial Adhesion. Langmuir. 2007;23(13):5120–6.

61. Liu Q, Singh A, Lalani R, Liu L. Ultralow Fouling Polyacrylamide on Gold

Surfaces via Surface- Initiated Atom Transfer Radical Polymerization.

Biomacromolecules. 2012;13:1086–92.

62. Fundeanu I, Klee D, Schouten AJ, Busscher HJ, Mei HC Van Der. Solvent-free

functionalization of silicone rubber and efficacy of PAAm brushes grafted from

an amino-PPX layer against bacterial adhesion. Acta Biomater.

2010;6(11):4271–6.

63. Fundeanu I, Mei HC Van Der, Schouten AJ, Busscher HJ. Polyacrylamide brush

coatings preventing microbial adhesion to silicone rubber. Colloids Surfaces B

Biointerfaces. 2008;64:297–301.

64. Fundeanu I, Mei HC Van Der, Schouten AJ, Busscher HJ. Microbial adhesion to

surface-grafted polyacrylamide brushes after long-term exposure to PBS and

reconstituted freeze-dried saliva. Wiley Period. 2010;997–1000.

65. Biran R, Pond D. Heparin coatings for improving blood compatibility of medical

devices. Adv Drug Deliv Rev. 2017;112:12–23.

66. Renata C, Bustanji Y, Conti M, Campoccia D, Baldassarri L, Samor B, et al.

Staphylococcus epidermidis – fibronectin binding and its inhibition by heparin.

Biomaterials. 2003;24:3013–9.

Page 54: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

52

67. Lundberg F, Gouda I, Larm O, Galin MA. A new model to assess staphylococcal

adhesion to intraocular lenses under in vitro flow conditions. Biomaterials.

1998;19:1727–33.

68. García-Sáenz M, Arias-puente A, Fresnadillo-Martinez M, Matilla-rodriguez A.

In vitro adhesion of Staphylococcus epidermidis to intraocular lenses. Cataract

Refract Surg. 2000;3350(0):12–4.

69. Campoccia D, Montanaro L, Arciola CR. The significance of infection related to

orthopedic devices and issues of antibiotic resistance. Biomaterials.

2006;27:2331–9.

70. Lin S, Rohini V, Lakew S, Peng S, Tsai H, Hsiue G. Plasma initiated graft

polymerization of 2-methacryloyloxyethyl phosphorylcholine on silicone

elastomer surfaces to enhance bio (hemo) compatibility. Surf Coat Technol.

2017;315:342–9.

71. Patel JD, Iwasaki Y, Ishihara K, Anderson JM. Phospholipid polymer surfaces

reduce bacteria and leukocyte adhesion under dynamic flow conditions. Wiley

Period. 2005;

72. Fujii K, Matsumoto HN, Koyama Y, Iwasaki Y, Ishihara K. Prevention of

Biofilm Formation with a Coating of 2-Methacryloyloxyethyl

Phosphorylcholine Polymer. J Vet Med. 2007;

73. Hirota K, Murakami K, Nemoto K, Miyake Y. Coating of a surface with 2-

methacryloyloxyethyl phosphorylcholine (MPC) co-polymer significantly

reduces retention of human pathogenic microorganisms. FEMS Microbiol Lett.

2005;248:37–45.

74. Lewis AL, Tolhurst LA, Stratford PW. Analysis of a phosphorylcholine-based

polymer coating on a coronary stent pre- and post-implantation. Biomaterials.

2002;23:1697–706.

75. Lewis AL, Cumming ZL, Goreish HH, Kirkwood LC, Tolhurst LA, Stratford

PW. Crosslinkable coatings from phosphorylcholine-based polymers.

Biomaterials. 2001;22:99–111.

76. Aumsuwan N, Heinhorst S, Urban MW. Antibacterial Surfaces on Expanded

Polytetrafluoroethylene; Penicillin Attachment. Biomacromolecules.

Page 55: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

53

2007;8:713–8.

77. Aumsuwan N, Heinhorst S, Urban MW. The Effectiveness of Antibiotic Activity

of Penicillin Attached to Expanded Poly(tetrafluoroethylene) (ePTFE) Surfaces:

A Quantitative Assessment. Bioconjug Chem. 2007;8:3525–30.

78. Aumsuwan N, Danyus RC, Heinhorst S, Urban MW. Attachment of Ampicillin

to Expanded Poly(tetrafluoroethylene): Surface Reactions Leading to Inhibition

of Microbial Growth. Biomacromolecules. 2008;9:1712–8.

79. Aumsuwan N, Mcconnell MS, Urban MW. Tunable Antimicrobial

Polypropylene Surfaces : Simultaneous Attachment of Penicillin (Gram +) and

Gentamicin (Gram -). Biomacromolecules. 2009;10:623–9.

80. Ma Z, Garrido-Maestu A, Jeong KC. Application, mode of action, and in vivo

activity of chitosan and its micro- and nanoparticles as antimicrobial agents: A

review. Carbohydr Polym [Internet]. 2017;176:257–65. Available from:

http://dx.doi.org/10.1016/j.carbpol.2017.08.082

81. Sahariah P, Másson M. Antimicrobial Chitosan and Chitosan Derivatives: A

Review of the Structure-Activity Relationship. Biomacromolecules [Internet].

2017;1–65. Available from:

http://pubs.acs.org/doi/abs/10.1021/acs.biomac.7b01058

82. Fu J, Ã JJ, Yuan W, Shen J. Construction of anti-adhesive and antibacterial

multilayer films via layer-by-layer assembly of heparin and chitosan.

Biomaterials. 2005;26:6684–92.

83. Guo S, Zhu X, Jun X. Controlling cell adhesion using layer-by-layer approaches

for biomedical applications. Mater Sci Eng. 2017;70:1163–75.

84. Follmann HDM, Martins AF, Gerola AP, Burgo TAL, Nakamura C V, Rubira

AF, et al. Antiadhesive and Antibacterial Multilayer Films via Layer-by-Layer

Assembly of TMC/Heparin Complexes. Biomacromolecules. 2012;13:3711–22.

85. Wang B, Ren K, Chang H, Wang J, Ji J. Construction of Degradable Multilayer

Films for Enhanced Antibacterial Properties. Appl Mater Interfaces.

2013;5(I):4136–43.

86. Blin T, Purohit V, Jouenne T, Glinel K. Bactericidal Microparticles Decorated

by an Antimicrobial Peptide for the Easy Disinfection of Sensitive Aqueous

Page 56: Ana Beatriz Lopes Roque dos Santos Marques Mestrado

54

Solutions. Biomacromolecules. 2011;12:1259–64.

87. Hadjesfandiari N, Yu K, Mei Y, Kizhakkedathu J. Polymer brush-based

approaches for the development of infection-resistant surfaces. J Mater Chem B.

2014;2:4968–78.