Montreal – Canada - International Cartilage Repair Society

Montreal – Canada - International Cartilage Repair Society Montreal – Canada - International Cartilage Repair Society

cartilage.org
from cartilage.org More from this publisher
06.01.2013 Views

2012 Montreal Canada May 12 15, 2012 10 th World Congress of the International Cartilage Repair Society Main Programme & Extended Abstracts www.cartilage.org P l a t i n u m S p o n s o r s : Please join us for the Sponsored Symposium “The Evolution of Autologous Chondrocyte Implantation and Cartilage Repair” Description: In this cartilage repair symposium, participants will learn about the genesis of autologous chondrocyte implantation, the importance of the chondrocyte and how the standards of clinical evidence, global regulation and manufacturing have evolved in this dynamic and exciting field Date: Monday, May 14th Time: 13:00 - 14:00 Location: Grand Salon For more information please visit us at booth #6.

2012<br />

<strong>Montreal</strong> <strong>–</strong> <strong>Canada</strong><br />

May 12 <strong>–</strong> 15, 2012<br />

10 th World Congress of the <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong><br />

Main Programme & Extended Abstracts<br />

www.cartilage.org<br />

P l a t i n u m S p o n s o r s :<br />

Please join us for the<br />

Sponsored Symposium<br />

“The Evolution of Autologous Chondrocyte<br />

Implantation and <strong>Cartilage</strong> <strong>Repair</strong>”<br />

Description: In this cartilage repair symposium, participants<br />

will learn about the genesis of autologous chondrocyte<br />

implantation, the importance of the chondrocyte and how<br />

the standards of clinical evidence, global regulation and<br />

manufacturing have evolved in this dynamic and exciting field<br />

Date: Monday, May 14th<br />

Time: 13:00 - 14:00<br />

Location: Grand Salon<br />

For more information please visit us at booth #6.


Chondro-Gide ® is not available in all markets. Availability is subject to<br />

the regulatory or medical practices that govern individual markets.<br />

www.geistlich-surgery.com<br />

Chondro-Gide ®<br />

enhanced marrow<br />

stimulation <strong>–</strong> also for ankle and hip?<br />

AMIC ® <strong>–</strong> A promising approach in cartilage regeneration<br />

ICRS 2012, <strong>Montreal</strong><br />

Satellite Symposium<br />

Sunday, 13 May 2012 <strong>–</strong> 13:00 until 14:00<br />

Chairman: Markus Walther, Munich, Germany<br />

Blood clot biology after marrow stimulation<br />

Caroline Hoemann, <strong>Canada</strong><br />

AMIC ® Talus <strong>–</strong> Surgical technique and first clinical results<br />

Markus Walther, Germany<br />

AMIC ® Hip <strong>–</strong> Surgical approach and 5-year clinical outcome<br />

Andrea Fontana, Italy<br />

Meet the Expert<br />

Geistlich Surgery, booth Nr. 8<br />

Sunday, 13 May, 10:45<strong>–</strong>11:15 AMIC ® Talus, Markus Walther<br />

Sunday, 13 May, 15:15<strong>–</strong>15:45 AMIC ® Talus, Martin Wiewiorski<br />

Monday, 14 May, 10:45<strong>–</strong>11:15 AMIC ® Hip, Andrea Fontana<br />

Monday, 14 May, 15:15<strong>–</strong>15:45 AMIC ® Knee Arthroscopic,<br />

Thomasz Piontek<br />

Geistlich Pharma AG<br />

Business Unit Surgery<br />

Bahnhofstrasse 40<br />

CH<strong>–</strong>6110 Wolhusen


10 th World Congress of the<br />

<strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong><br />

ICRS 2012<br />

May 12 <strong>–</strong> 15, 2012<br />

<strong>Montreal</strong> / Quebec, <strong>Canada</strong><br />

“Advancing science & education in cartilage repair worldwide!”<br />

Main Programme & Extended Abstracts<br />

Editorial Office:<br />

<strong>Cartilage</strong> Executive Office GmbH <strong>–</strong> Wetzikon <strong>–</strong> Switzerland<br />

www.cartilage.org<br />

Copyright: <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong> <strong>–</strong> ICRS 2012<br />

Cover acknowledgment:<br />

Images kindly provided by Tourism <strong>Montreal</strong> Programme Price: ! 15.00<br />

ICRS does not accept responsibility for errors or misprints


Knee <strong>Repair</strong> Solutions<br />

Save the menicus.<br />

Preserve the cartilage.<br />

Lunchtime Symposium at ICRS 2012<br />

Sunday, 13 May, 2012<br />

13:00 <strong>–</strong> 14:00<br />

Grand Salon<br />

Smith & Nephew, Inc.<br />

Andover, MA 01810<br />

USA<br />

From Simple to Complex Meniscal Injury <strong>–</strong><br />

How and When to Resect, <strong>Repair</strong> and Replace<br />

Faculty:<br />

Tim Spalding, MD, UK<br />

• Anatomy of Meniscus<br />

• When to <strong>Repair</strong>/Resect<br />

• How I <strong>Repair</strong><br />

• My Experiences<br />

• Case Studies<br />

T +1 978 749 1000<br />

www.smith-nephew.com<br />

Visit us at<br />

booth #4<br />

Peter Verdonk, MD, Belgium<br />

• <strong>Repair</strong>s using Allograft, Scaffolds<br />

• When to <strong>Repair</strong><br />

• How I <strong>Repair</strong><br />

• My Experiences<br />

• Case Studies<br />

Trademark of Smith & Nephew. Registered US Patent<br />

and Trademark Office. ©2012 Smith & Nephew. All<br />

rights reserved. Printed in USA. 03/12 2909 Rev. A


Zimmer Biologics<br />

innovative solutions for joint preservation<br />

We invite you to join us at the ICRS Zimmer Symposium:<br />

“Innovative Solutions for Joint Preservation<br />

with DeNovo ® NT Natural Tissue Graft and<br />

Chondrofix ® Osteochondral Allograft”<br />

Speakers:<br />

David Caborn, M.D.<br />

Jack Farr, M.D.<br />

Eric Giza, M.D.<br />

Norman Marcus, M.D.<br />

Date: May 14, 2012<br />

Time: 1:00 - 2:00pm EST<br />

Location: Marquette<br />

Visit us at www.zimmer.com or<br />

Call us at 1-800-348-2759<br />

©2012 Zimmer, Inc. All Rights Reserved


“ChondroCelect: When evidence<br />

makes a difference”<br />

TiGenix NV<br />

Researchpark Haasrode 1724<br />

Romeinse straat 12 bus 2, 3001 Leuven<br />

Belgium<br />

Phone +32 (0) 16 39 60 66<br />

Fax +32 (0) 16 39 79 70<br />

info@tigenix.com<br />

www.tigenix.com<br />

Prof Fredrik Almqvist, University of Ghent, Belgium<br />

Visit us<br />

at booth<br />

# 1<br />

ChondroCelect ® is a medicinal product EU/1/09/563/001<br />

For more information on ChondroCelect please read the prescribing information at booth # 1.<br />

TiGenix Satellite Symposium<br />

at ICRS 2012<br />

Monday, 14 May at 13:00<br />

Meeting Room Jolliet


Healthcare<br />

knowledge action care<br />

BST-CarGel ® :<br />

A Novel Bioscaffold for<br />

Enhanced Tissue Regeneration<br />

in <strong>Cartilage</strong> <strong>Repair</strong> Procedures<br />

Satellite Lunch Symposium<br />

Sunday, May 13 2012,<br />

from 1:00 pm <strong>–</strong> 2:00 pm<br />

Presentations<br />

• Underlying Principles of Enhancing<br />

Bone Marrow Stimulation with BST-CarGel ®<br />

• MRI-based Imaging Biomarkers for <strong>Cartilage</strong><br />

• Final Outcomes from the BST-CarGel ®<br />

Randomized Clinical Trial<br />

Faculty<br />

Co-chairs:<br />

• Pierre Ranger, MD<br />

• Matthias Steinwachs, MD<br />

Speakers:<br />

• Timothy Mosher, MD<br />

• Matthew Shive, PhD<br />

• William Stanish, MD<br />

Booth #7 : Piramal Bio-Orthopaedics<br />

Visit our booth and discover BST-CarGel ® .<br />

You will have the chance to<br />

meet the Investigators<br />

and learn how<br />

you could be involved in an international<br />

Center of Excellence training program.<br />

Piramal Bio-Orthopaedics<br />

Bringing international innovations to the world.


<strong>Cartilage</strong> <strong>Repair</strong> - “A Merging of Basic<br />

Research and Clinical Applications”<br />

ICRS is a unique forum for international collaboration in cartilaginous<br />

tissue research by bringing together clinicians, clinical researchers<br />

and basic scientists, engaged or interested in the field of cartilage biology,<br />

imaging, cartilaginous tissue engineering and translational clinical approaches to<br />

treatment of cartilage pathologies. The link between laboratory work and the daily<br />

treatments of patients in a clinical setting is extremely important to the <strong>Society</strong>.<br />

The <strong>Society</strong> envisages the scientific research and the exchange of knowledge among<br />

physicians, scientists & patients of the industry in the field of cartilage repair. To serve<br />

its purpose, the society organizes international congresses and events, publishes<br />

journals and provides a universal internet discussion-platform. Furthermore the<br />

society provides an outstanding scientific database, financially supports research<br />

projects and pays for scholarships / fellowships which are available for members only.<br />

Join ICRS Now!<br />

Benefits for ICRS Members<br />

• 2 Issues of the ICRS Newsletter<br />

• 4 Issues of the new official ICRS Journal<br />

<strong>Cartilage</strong>, published by SAGE<br />

• 4 issues of the Musculoskeletal Report (US &<br />

Europe reports)<br />

• Highly reduced registration fees for to all ICRS<br />

educational events<br />

• Eligibility for ICRS Scholarships<br />

• Eligibility for ICRS Travelling Fellowships<br />

The Annual Membership Fees are:<br />

Ordinary Members: € 135.00<br />

Junior Members: € 70.00<br />

Corporate Members: € 2300.00<br />

• Reduced price of the ICRS database software<br />

SOCRATES<br />

• Access to the “Member Only” section of the<br />

ICRS Website (incl. Membership Database and<br />

access to the <strong>Cartilage</strong> Knowledge Database,<br />

Electronic Posters, Abstracts and selected<br />

Symposia presentations)<br />

• Free advertising of your cartilage meeting<br />

/ course in the ICRS Newsletter and online<br />

event calendar<br />

Join ICRS online at<br />

www.cartilage.org<br />

office@cartilage.org<br />

HJ10010102_1108045


Programme Overview<br />

Saturday, May 12<br />

09:00 <strong>–</strong> 18:00<br />

Registration<br />

13:00 <strong>–</strong> 14:00 Plenary Session 1.0<br />

Sports Injury: ICRS <strong>–</strong> FIFA Page 56<br />

Room: Grand Salon<br />

14:15 <strong>–</strong> 15:15 14:15 <strong>–</strong> 15:15 14:15 <strong>–</strong> 15:15<br />

Session 2.1 Session 2.2 Session 2.3<br />

Growth Nerve Femoropatellar<br />

Factors Dependence Joint<br />

Page 56 Page 56 Page 57<br />

Room: Grand Salon Room: Marquette Room: Jolliet<br />

15:15 <strong>–</strong> 15:45<br />

Coffee Break/Industry Exhibition<br />

15:45 <strong>–</strong> 16:45 15:45 <strong>–</strong> 16:45 15:45 <strong>–</strong> 16:45<br />

Session 3.1 Session 3.2 Session 3.3<br />

Opportunities Medication Meniscus<br />

of Bioprinting and <strong>Cartilage</strong><br />

Page 57 Page 57 Page 58<br />

Room: Jolliet Room: Marquette Room: St. François<br />

17:00 <strong>–</strong> 17:45 Plenary Session 4.0<br />

Opening Ceremony & Awards Session Page 58<br />

Room: Grand Salon<br />

17:45 <strong>–</strong> 18:45 Plenary Session 5.0<br />

Honorary Lectures Page 58<br />

Room: Grand Salon<br />

19:00 <strong>–</strong> 20:30<br />

Welcome Reception<br />

Exhibition Hall<br />

Sunday, May 13<br />

07:30 <strong>–</strong> 08:15 07:30 <strong>–</strong> 08:15<br />

Workshop 6.1 Workshop 6.2<br />

Imaging of Meet the Experts<br />

<strong>Cartilage</strong> Defects (Basic Scientists)<br />

Page 59 Page 59<br />

Room: Jolliet Room: Marquette<br />

08:30 <strong>–</strong> 09:30 Plenary Session 7.0<br />

Clinical Studies using <strong>Cartilage</strong> Fragements Page 59<br />

Room: Grand Salon<br />

09:45 <strong>–</strong> 10:45 09:45 <strong>–</strong> 10:45 09:45 <strong>–</strong> 10:45<br />

Session 8.1 Session 8.2 Session 8.3<br />

<strong>Cartilage</strong> Culture Intervertebral<br />

<strong>Repair</strong>, Foot Techniques Disc<br />

and Ankle<br />

Page 59 Page 59 Page 60<br />

Room: Marquette Room: Grand Salon Room: St. François<br />

10:45 <strong>–</strong> 11:15<br />

Coffee Break/Industry Exhibition<br />

11:15 <strong>–</strong> 12:45 Free Paper Sessions<br />

9.1 ISAKOS Symposium Page 60<br />

Room: Grand Salon<br />

9.2 Chondrocytes 1 Page 60<br />

Room: Marquette<br />

9.3 Biomaterials & Scaffolds 1 Page 61<br />

Room: St. François<br />

9.4 Other Musculoskeletal Tissues Page 62<br />

Room: Jolliet<br />

13:00 <strong>–</strong> 14:00 Industry Symposia<br />

10.1 Geistlich Page 63<br />

Room: Marquette<br />

10.2 Smith & Nephew Page 63<br />

Room: Grand Salon<br />

10.3 Piramal Page 63<br />

Room: Jolliet<br />

10.4 Anika Page 64<br />

Room: St. François<br />

14:15 <strong>–</strong> 15:45 Free Paper Sessions<br />

11.1 Animal Models Page 64<br />

Room: Grand Salon<br />

11.2 Chondrocytes 2 Page 65<br />

Room: St. François<br />

11.3 Biomaterials & Scaffolds 2 Page 66<br />

Room: Marquette<br />

11.4 Imaging Page 67<br />

Room: Jolliet<br />

15:45 <strong>–</strong> 17:00<br />

General Assembly (For Members only)<br />

15:45 <strong>–</strong> 18:00<br />

Poster Viewing Cocktail<br />

Rooms: Duluth/Richelieu<br />

18:45 <strong>–</strong> 23:30 (Meeting point 06:30 Fairmont lobby)<br />

Canadian Sugar Shack & Maple Forest Party<br />

Room: Jolliet<br />

7


Programme Overview<br />

Monday, May 14<br />

07:30 <strong>–</strong> 08:15 07:30 <strong>–</strong> 08:15<br />

Workshop 13.1 Workshop 13.2<br />

Rehabilitation after Meet the Experts<br />

<strong>Cartilage</strong> <strong>Repair</strong> (Clinicians)<br />

Page 68 Page 68<br />

Room: Jolliet Room: Marquette<br />

08:30 <strong>–</strong> 09:30 Plenary Session 14.0<br />

Cell Free Approaches for <strong>Cartilage</strong> <strong>Repair</strong> Page 68<br />

Room: Grand Salon<br />

09:45 <strong>–</strong> 10:45 09:45 <strong>–</strong> 10:45 09:45 <strong>–</strong> 10:45<br />

Session 15.1 Session 15.2 Session 15.3<br />

Platelet Rich Development of Subchondral<br />

Plasma and Joint new Biomaterials Bone & <strong>Cartilage</strong><br />

Tissue <strong>Repair</strong> & Scaffolds <strong>Repair</strong><br />

Page 68 Page 69 Page 69<br />

Room: Grand Salon Room: Marquette Room: St. François<br />

10:45 <strong>–</strong> 11:15<br />

Coffee Break/Industry Exhibition<br />

11:15 <strong>–</strong> 12:45 Free Paper Sessions<br />

16.1 Osteoarthritis 1 Page 69<br />

Room: St. François<br />

16.2 <strong>Cartilage</strong>/Cell Transplantation Page 70<br />

Room: Marquette<br />

16.3 Microfracture/Bone Marrow Page 71<br />

Room: Jolliet<br />

16.4 Stem Cells 1 Page 72<br />

Room: Grand Salon<br />

13:00 <strong>–</strong> 14:00 Industry Symposia<br />

17.1 Sanofi Page 73<br />

Room: Grand Salon<br />

17.2 Zimmer Page 73<br />

Room: Marquette<br />

17.3 Tigenix Page 73<br />

Room: Jolliet<br />

17.4 BioTissue Page 74<br />

Room: St. François<br />

14:15 <strong>–</strong> 17:15 Plenary Session 18.0<br />

Stem Cells for <strong>Cartilage</strong> <strong>Repair</strong> Page 74<br />

Room: Grand Salon<br />

15:30 <strong>–</strong> 16:30 15:30 <strong>–</strong> 16:30 15:30 <strong>–</strong> 16:30<br />

Session 19.1 Session 19.2 Session 19.3<br />

Imaging YSOS Biomakers<br />

Technologies Symposium<br />

Tissue <strong>Repair</strong><br />

Page 74 Page 75 Page 75<br />

Room: Marquette Room: Jolliet Room: Grand Salon<br />

16:30 <strong>–</strong> 17:30<br />

Poster Viewing/Coffee Break Page 82<strong>–</strong>99<br />

Rooms: Duluth/Richelieu<br />

17:30 <strong>–</strong> 18:30 17:30 <strong>–</strong> 18:30 17:30 <strong>–</strong> 18:30<br />

Session 21.1 Session 21.2 Session 21.3<br />

Joint Lubrication Rehabilitation Stryker Clinical<br />

& <strong>Cartilage</strong> Health Scientist Progr.<br />

Page 75 Page 76 Page 76<br />

Room: Grand Salon Room: Marquette Room: Jolliet<br />

Tuesday, May 15<br />

08:30 <strong>–</strong> 09:30 Plenary Session 22.0<br />

Biomechanics & Stability of <strong>Cartilage</strong> <strong>Repair</strong> Page 77<br />

Room: Grand Salon<br />

09:30 <strong>–</strong> 10:00 Plenary Session 23.0<br />

Strategic Outlines ICRS Page 77<br />

Room: Grand Salon<br />

10:00 <strong>–</strong> 10:30<br />

Coffee Break/Industry Exhibition<br />

10:30 <strong>–</strong> 11:30 10:30 <strong>–</strong> 11:30 10:30 <strong>–</strong> 11:30<br />

Session 24.1 Session 24.2 Session 24.3<br />

<strong>Cartilage</strong> Animal Distraction<br />

<strong>Repair</strong> in Models Arthroplasty<br />

the Hip Joint<br />

Page 77 Page 77 Page 78<br />

Room: Marquette Room: Grand Salon Room: Jolliet<br />

11:30 <strong>–</strong> 13:00 Free Paper Sessions<br />

25.1 Osteoarthritis 2 Page 78<br />

Room: St. François<br />

25.2 <strong>Cartilage</strong> / Cell Transplantation Page 79<br />

Room: Marquette<br />

25.3 Rehabilitation Page 80<br />

Room: Jolliet<br />

25.4 Stem Cells 2 Page 81<br />

Room: Grand Salon<br />

13:00 <strong>–</strong> End of Meeting<br />

8


Programme<br />

at a glance<br />

Please Fold out this<br />

Page...<br />

The ICRS would like to acknowledge the<br />

following companies for their generous<br />

support to the 10 th ICRS World Congress<br />

in <strong>Montreal</strong> <strong>–</strong> <strong>Canada</strong>.<br />

P l a t i n u m S p o n s o r s :<br />

S i l v e r S p o n s o r :<br />

9


10<br />

I am pleased to extend my warmest greetings to everyone taking<br />

part in the 10th World Congress of the <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong>‐<br />

ICRS 2012.<br />

This convention offers a wonderful opportunity for scientists and<br />

surgeons from more than 70 countries to gather with their peers to advance their<br />

knowledge in cartilage biology and tissue engineering and to bring new hope and<br />

treatments to patients affected by cartilage damage. I am certain that delegates<br />

will make the most<br />

I am<br />

of<br />

pleased<br />

the scientific<br />

to extend<br />

presentations<br />

my warmest<br />

and<br />

greetings<br />

discussions<br />

to everyone<br />

taking<br />

taking<br />

place<br />

part<br />

here<br />

in<br />

in<br />

the<br />

Montréal,<br />

10th World<br />

as well<br />

Congress<br />

as the many<br />

of the<br />

informal<br />

<strong>International</strong><br />

networking<br />

<strong>Cartilage</strong><br />

opportunities<br />

<strong>Repair</strong> <strong>Society</strong>‐<br />

ICRS<br />

provided<br />

2012.<br />

by the conference hosts.<br />

This<br />

I would<br />

convention<br />

like to commend<br />

offers a wonderful<br />

the organizers<br />

opportunity<br />

for their<br />

for<br />

efforts<br />

scientists<br />

to promote<br />

and<br />

surgeons<br />

scientific<br />

from<br />

excellence<br />

more than<br />

in the<br />

70<br />

field<br />

countries<br />

through<br />

to<br />

research,<br />

gather with<br />

collaboration,<br />

their peers<br />

and<br />

to advance<br />

education.<br />

their<br />

knowledge in cartilage biology and tissue engineering and to bring new hope and<br />

treatments to patients<br />

On behalf<br />

affected<br />

of the<br />

by<br />

Government<br />

cartilage damage.<br />

of <strong>Canada</strong>,<br />

I am<br />

I<br />

certain<br />

wish you<br />

that<br />

all<br />

delegates<br />

an<br />

will<br />

informative<br />

make the<br />

and<br />

most<br />

productive<br />

of the scientific<br />

meeting.<br />

presentations and discussions taking place<br />

here in Montréal, as well as the many informal networking opportunities<br />

provided by the conference hosts.<br />

I would like to commend the organizers for their efforts to promote<br />

scientific excellence in the field through research, collaboration, and education.<br />

On behalf of the Government of <strong>Canada</strong>, I wish you all an<br />

informative and productive meeting. The Rt. Hon. Stephen Harper, P.C., M.P.<br />

OTTAWA<br />

2012<br />

OTTAWA<br />

2012<br />

The Rt. Hon. Stephen Harper, P.C., M.P.<br />

10


Mot du premier ministre<br />

Je suis heureux de souhaiter la bienvenue<br />

à Montréal à tous celles et ceux que réunit<br />

le World Congress of the <strong>International</strong><br />

<strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong>.<br />

Voilà pour chacun de vous une occasion<br />

choisie de rencontrer des collègues des<br />

quatre coins du monde et d’échanger avec<br />

eux sur les dernières connaissances qui<br />

marquent le développement de votre<br />

spécialité médicale. En ce sens, je salue<br />

votre participation à ce grand rendez-vous<br />

qui contribue directement à l’amélioration<br />

continue des soins et de la santé publique.<br />

Félicitations aux organisateurs de ce<br />

congrès dont on célèbre cette année la<br />

10 e édition, et bravo aux gens de<br />

l’<strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong> qui<br />

soutiennent sans relâche l’avancement de<br />

la science et de la recherche.<br />

Bon congrès à tous,<br />

A Word from the Premier<br />

Welcome to Montréal for the World<br />

Congress of the <strong>International</strong> <strong>Cartilage</strong><br />

<strong>Repair</strong> <strong>Society</strong>.<br />

What an ideal opportunity to meet<br />

colleagues from around the world and<br />

discuss the latest developments in your<br />

medical specialty. I salute you for<br />

attending this major gathering that<br />

contributes directly to the continuous<br />

improvement of healthcare and public<br />

health.<br />

Congratulations to the organizers of this<br />

10th edition of the congress and bravo to<br />

the <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong><br />

that tirelessly supports the advancement<br />

of science and research.<br />

Best wishes to all for a constructive<br />

congress.<br />

Jean Charest<br />

11


Submit your manuscript<br />

Editor-in-Chief:<br />

Roy D. Altman, M.D.<br />

http://cart.sagepub.com<br />

Pick up<br />

your free<br />

copy at<br />

the SAGE<br />

booth!<br />

<strong>Cartilage</strong> publishes articles related to the musculoskeletal system with particular attention to<br />

cartilage repair, development, function, degeneration, transplantation and rehabilitation. The<br />

journal is a forum for the exchange of ideas for the many types of researchers and clinicians<br />

involved in cartilage biology and repair. A primary objective of <strong>Cartilage</strong> is to foster the crossfertilization<br />

of the fi ndings between clinical and basic sciences throughout the various disciplines<br />

involved in cartilage repair.<br />

The journal publishes full length original manuscripts on all types of cartilage including articular,<br />

nasal, auricular, tracheal/bronchial and intervertebral disc fi brocartilage. Manuscripts on clinical<br />

and laboratory research are welcome. Review articles, editorials and letters are also encouraged.<br />

<strong>Cartilage</strong> is a forum for the exchange of knowledge among clinicians, scientists, patients and<br />

researchers.<br />

Instructions to authors and directions for submissions will be available at http://cart.sagepub.com.<br />

Manuscripts can be submitted at http://mc.manuscriptcentral.com/cart.<br />

The <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong> (ICRS) is dedicated to promotion, encouragement and<br />

distribution of fundamental and applied research of cartilage in order to permit a better knowledge<br />

of function and dysfunction of articular cartilage and its repair (www.cartilage.org).<br />

<strong>Cartilage</strong> is published by SAGE on behalf of the ICRS.


Dear ICRS Members, Colleagues and Friends<br />

The 2012 ICRS Programme Committee is committed in<br />

providing our membership, friends and industry partners,<br />

new and exciting developments in clinical and preclinical<br />

applications to the repair and regeneration of cartilage<br />

and related musculoskeletal injuries. Our overall<br />

goal for this meeting was to broaden the scope of presentations<br />

provided at our world congress and to promote<br />

better interdisciplinary education and understanding<br />

between the clinical and pre-clinical scientists.<br />

From the clinical perspective the programme will present<br />

new surgical procedures used for cartilage repair, including<br />

the use of new biomaterials and bioengineering<br />

technologies. Moreover, the programme presents the<br />

best available evidence for routinely accepted procedures,<br />

as well. The Programme Committee has also introduced<br />

some new sessions including the femoropatellar<br />

joint, medication & cartilage, follow up assessment of<br />

clinical cases, platelet rich plasma, joint tissue repair <strong>–</strong><br />

facts <strong>–</strong> the hope, hype and reality as well as another session<br />

on cell free approaches for cartilage repair.<br />

From the basic science (pre-clinical) perspective important<br />

research areas of biomarkers, proteomics and<br />

genomics as well as new imaging technologies will be<br />

approached. New sessions of interest include nerve de-<br />

In Strategic collaboration with:<br />

pendence on cartilage development, the use of bioprinting<br />

in cartilage regeneration, evaluation of cellular therapies<br />

from chondrocytes to stem cells as well as genetic<br />

influences on cartilage repair. In addition, there will be<br />

a dedicated special session on the intervertebral disc<br />

(brought back by popular demand!) and also a special<br />

session on subchondral bone and its relevance with repair<br />

and osteoarthritis.<br />

We hope you enjoy both, the quality of the presented<br />

science and the great opportunity for networking and social<br />

interactions with participants from all over the world.<br />

Sign-up for the “Fun Run for Healthy Joints” or for the<br />

exciting “Canadian Sugar Shack & Maple Forest Party”,<br />

both events hold on Sunday, May 13.<br />

Sincerely,<br />

Jos Malda and Wayne McIlwraith<br />

Scientific Programme Chairmen 2012<br />

Michael Buschmann and Patrick Lavigne<br />

Congress Co-Chairmen 2012<br />

13<br />

Michael Buschmann<br />

Patrick Lavigne<br />

Wayne McIlwraith<br />

Jos Malda


14<br />

Past World Congresses, Presidents and Award Winners<br />

Past World Congresses<br />

1997 <strong>–</strong> 1st World Congress<br />

Freiburg, Switzerland; Roland Jakob<br />

1998 <strong>–</strong> 2nd World Congress<br />

Boston, USA; Alan Grodzinsky<br />

2000 <strong>–</strong> 3th World Congress<br />

Gothenburg, Sweden; Lars Peterson<br />

2002 <strong>–</strong> 4th World Congress<br />

Toronto, <strong>Canada</strong>; Shawn O’Driscoll<br />

2004 <strong>–</strong> 5th World Congress<br />

Gent, Belgium; Rene Verdonk<br />

2006 <strong>–</strong> 6th World Congress<br />

San Diego, USA; Bert Mandelbaum, Bill Bugbee<br />

2007 <strong>–</strong> 7th World Congress<br />

Warsaw, Poland; Jaroslaw Deszczynski,<br />

Jacek Kruczynski, Konrad Slynarski<br />

2009 <strong>–</strong> 8th World Congress<br />

Miami, USA; Jack Farr, Tom Minas<br />

2010 <strong>–</strong> 9th World Congress<br />

Sitges / Barcelona, Spain; Ramon Cugat, Pedro Guillen<br />

Past Presidents<br />

1997 Roland Jacob, Switzerland<br />

1998 Roland Jacob, Switzerland<br />

1999 Alan Grodzinsky, USA<br />

2000 Lars Peterson, Sweden<br />

2001 Lars Peterson, Sweden<br />

2002 Shawn O’Driscoll, USA<br />

2003 Shawn O’Driscoll, USA<br />

2004 Ernst Hunziker, Switzerland<br />

2005 Ernst Hunziker, Switzerland<br />

2006 Mats Brittberg, Sweden<br />

2007 Mats Brittberg, Sweden<br />

2008 Bert Mandelbaum, USA<br />

2009 Lisa Fortier, USA<br />

2010 Lisa Fortier, USA<br />

2011 Daniël Saris, NL<br />

2012 Daniël Saris, NL (current)<br />

Honorary Fellows<br />

2007 Alan Grodzinski, USA<br />

2007 Roland Jakob, Switzerland<br />

2007 Lars Peterson, Sweden<br />

2009 Mats Brittberg, Sweden<br />

2012 Tom Minas, USA<br />

2012 Stefan Nehrer, Austria<br />

Award Winners<br />

Genzyme <strong>–</strong> ICRS Award for Excellence in<br />

<strong>Cartilage</strong> Research<br />

2004 Ronald Dorotka et al, Austria<br />

2006 Mark Randolph et al, United Kingdom<br />

2007 Gerjo Van Osch et al, The Netherlands<br />

2009 Avner Yayon et al, Israel<br />

2010 Attila Aszody et al, Germany<br />

Genzyme <strong>–</strong> ICRS Lifetime Award<br />

2004 Lars Peterson, Sweden<br />

2006 Allan Gross, <strong>Canada</strong><br />

2007 Arnold Caplan, USA<br />

2009 Richard Steadman, USA<br />

2010 Mats Brittberg, Sweden<br />

Best Rated Abstracts<br />

2007 K. Nakagawa et al, Japan<br />

2007 C. Moser et al, Germany<br />

2009 J.F. Harrington et al, USA<br />

2010 S. D’Arcy et al, Ireland<br />

2012 G. Van Den Akker, NL


Table of Contents<br />

Programme at a Glance (Fold-out) 8<strong>–</strong>9<br />

Welcome messages 10<strong>–</strong>13<br />

Past Meetings, Presidents & Award Winners 14<br />

ICRS <strong>Society</strong>- and Meeting Committees 16<br />

General Information 17<strong>–</strong>22<br />

Hotel, Travel & Social Event Information 23<strong>–</strong>26<br />

Invited Faculty Members 27<strong>–</strong>45<br />

Exhibitor's Guide, Exhibition Plan & Sponsor List 46<strong>–</strong>54<br />

Scientific Programme / Agenda 56<strong>–</strong>81<br />

Poster Sessions 82<strong>–</strong>99<br />

Extended Abstracts 102<strong>–</strong>181<br />

“Advancing science & education in cartilage repair worldwide!”<br />

15


16<br />

ICRS <strong>Society</strong>- and Meeting Committees<br />

ICRS Executive Board<br />

President:<br />

Daniël Saris, Utrecht, The Netherlands<br />

Past-President:<br />

Lisa Fortier, Ithaca, USA<br />

Vice-President:<br />

Anthony Hollander, Bristol, UK<br />

Secretary General:<br />

Norimasa Nakamura, Osaka, JP<br />

Treasurer:<br />

Susan Chubinskaya, Chicago, USA<br />

ICRS General Board<br />

Altman Roy, Los Angeles, USA<br />

Buschmann Michael, <strong>Montreal</strong>, CA<br />

Chubinskaya Susan, Harrison, USA<br />

Erggelet Christopher, Zurich, CH<br />

Fortier Lisa, Ithaca, USA<br />

Hollander Anthony, Bristol, UK<br />

Kon Elizaveta, Bolgna, Italy<br />

Malda Jos, Utrecht, NL<br />

Marlovits Stefan, Vienna, AT<br />

McIlwraith Wayne, Fort Collins, USA<br />

Minas Tom, Chestnut Hill, USA<br />

Nakamura Norimasa, Suita, Japan<br />

Nehrer Stefan, Krems, AT<br />

Peterson Lars, Västra Frölunda, SE<br />

Daniël Saris, Utrecht, NL<br />

Van Osch Gerjo, Rotterdam, NL<br />

Kennneth Zaslav, Richmond, USA<br />

Invited Faculty Members<br />

Please find the “who is who” of our distinguished invited<br />

faculty members on pages 27 to 45.<br />

(Only invited faculty members, who have sent us their<br />

biosketch and portrait pictures by March 20, could be<br />

included in this book and therefore it does not represent<br />

an exhaustive listing.)<br />

Scientific Programme Committee<br />

Co-Chairmen:<br />

Malda Jos, Utrecht, NL<br />

McIlwraith Wayne, Fort Collins, USA<br />

Members:<br />

Caterson Bruce, Cardiff, Wales UK<br />

Erggelet Chris, Zurich, CH<br />

Kafienah Wael, Bristol, UK<br />

Jurvelin Jukka, Kuopio, FI<br />

Richardson James, Oswestry, UK<br />

Ferretti Mario, São Paulo, BR<br />

Educational Committee<br />

Co-Chairmen:<br />

Stefan Nehrer, Krems, Austria<br />

Tom Minas, Chstnut Hill, USA<br />

Members:<br />

Alberto Gobbi, Milano, Italy<br />

Görtz Simon, La Jolla, USA<br />

Nesic Dobrila, Bern, CH<br />

Polacek Martin, Tromso, NO<br />

Slynarsky Konrad, Warsaw, PL<br />

Tratttnig Siegfried, Vienna, AT<br />

Local Organising Committee<br />

Congress Co-Chairmen:<br />

Buschmann Michael, <strong>Montreal</strong>, CA<br />

Lavigne Patrick, <strong>Montreal</strong>, CA<br />

Members:<br />

Gross Alan, Toronto, CA<br />

Hoemann Caroline, <strong>Montreal</strong>, CA<br />

Hurtig Mark, Guelph, CA<br />

Kandel Rita, Toronto, CA<br />

Mc Cormack Robert, Vancouver, CA<br />

Morelli Moreno, <strong>Montreal</strong>, CA<br />

Poole Robin, <strong>Montreal</strong>, CA<br />

Stanish William, CA


General Information<br />

Venue<br />

Fairmont <strong>–</strong> The Queen Elizabeth Hotel<br />

900 Rene-Levesque West<br />

Montréal (Québec) H3B 4A5<br />

Phone: +1 514 861-3511<br />

Fax: +1 514 954-2256<br />

www.fairmont.com/queenelizabeth<br />

queenelizabeth.hotel@fairmont.com<br />

Organizing Office<br />

<strong>Cartilage</strong> Executive Office GmbH<br />

Mr. Stephan Seiler<br />

Spitalstrasse 190, House 3<br />

8623 Wetzikon, Switzerland<br />

Phone +41 44 503 73 70<br />

Fax +41 44 503 73 72<br />

office@cartilage.org<br />

Congress Dates<br />

Start: Saturday, May 12 at 13.00<br />

End: Tuesday, May 15 at 13.00<br />

Language<br />

The official language of the congress is English.<br />

No simultaneous translation will be provided.<br />

Congress Registration<br />

Pre-Registration<br />

Until April 30, only internet online registrations are accepted<br />

at www.cartilage.org. After that date, only onsite<br />

registration at the Hotel Fairmont congress registration<br />

desk will be possible.<br />

On-Site Registration<br />

The registration desks are located in the mezzanine of<br />

the Fairmont conference centre.<br />

Congress Registration Fees<br />

Early, until February 29<br />

ICRS Members $ 390.00<br />

Non-Members $ 550.00<br />

Industry Representatives $ 650.00<br />

Junior-Members/Residents/Nurses* $ 290.00*<br />

Acc. Persons** $ 90.00**<br />

Late, March 1 to April 10<br />

ICRS Members $ 490.00<br />

Non-Members $ 650.00<br />

Industry Representatives $ 750.00<br />

Junior-Members/Residents/Nurses* $ 390.00*<br />

Acc. Persons** $ 100.00**<br />

On-Site, as from April 11<br />

ICRS Members $ 550.00<br />

Non-Members $ 690.00<br />

Industry Representatives $ 850.00<br />

Junior-Members/Residents/Nurses* $ 450.00*<br />

Acc. Persons** $ 110.00**<br />

* to be accompanied by a certificate signed by the head<br />

of department, if not a Junior Member of ICRS<br />

** accompanying persons do not have access to attend<br />

scientific sessions but can visit the industry exhibition<br />

and are cordially invited to attend the Welcome<br />

Reception, Coffee Breaks and the Opening & Closing<br />

ceremonies<br />

Attention: ICRS-Members who are not in good standing<br />

with their membership dues 2011/2012 will automatically<br />

be charged with the non-member fee by the registration<br />

system.<br />

Payment <strong>–</strong> Registration Fees<br />

Registration fees must be paid to ICRS within 10 days after<br />

the date of registration or latest until the end of the<br />

registration period of the respective fee type. Payments<br />

can be made as follows:<br />

By Bank remittance to<br />

Berner Kantonalbank (BEKB)<br />

CH-3001 Bern, Switzerland<br />

Clearing-No: 790<br />

BIC/SWIFT Code: KBBECH22<br />

Konto-Nr.: 16 246244671<br />

IBAN: CH97 0079 0016 2462 4467 1<br />

By Credit Card<br />

VISA, Euro/Mastercard and American Express<br />

Cash (onsite)<br />

CAD$, US$ & Euros are accepted<br />

17


18<br />

General Information<br />

Regular Registration Fee includes<br />

- Access to all Scientific Sessions<br />

(except Workshops)<br />

- Access to the Technical Industry Exhibition<br />

- Access to the Scientific Poster Exhibition<br />

(Wine & Cheese)<br />

- Welcome Reception<br />

- Coffee Breaks<br />

- Congress Bag with Main Programme<br />

- Personal Badge and Certificate of Attendance<br />

On-site Registration Desk Opening Hours<br />

The secretarial office and the registration desk will be<br />

open as follows:<br />

Friday May 12 16.00 <strong>–</strong> 18.30<br />

Saturday May 12 10.00 <strong>–</strong> 18.30<br />

Sunday May 13 07.30 <strong>–</strong> 18.30<br />

Monday May 14 07.30 <strong>–</strong> 18.30<br />

Tuesday May 15 07.30 <strong>–</strong> 18.30<br />

On-site Check-in Procedures<br />

Delegates must personally check-in at the appropriate<br />

registration desk at the main entrance with a valid ID. An<br />

express lane for check-in of pre-registered and prepaid<br />

delegates will be available.<br />

Participants, who are not yet in possession of their badges<br />

when leaving their home country, are requested to<br />

bring the confirmation letter or a copy of the invoice with<br />

them. This will facilitate an efficient congress check-in.<br />

Cancellations of Congress Registrations &<br />

Name Changes<br />

Written notification is required for all registration cancellations<br />

and name changes. Cancellation of registration<br />

should be sent to the ICRS Congress Office in Zurich.<br />

Name changes will be charged with € 100.00 each.<br />

Refunds for registration cancellations<br />

are as follows<br />

80% for cancellations before February 20<br />

50% for cancellations from February 21 to April 20<br />

No refund can be made thereafter<br />

In case of cancellation due to the rejection of abstracts<br />

or refuse of entry visa to <strong>Canada</strong>, the full amount of the<br />

registration fee will be refunded.<br />

Cancellation of the Congress<br />

by the Organizer<br />

In case of cancellation of the congress by the organizer,<br />

congress fees will be refunded in case of cancellation<br />

due to other reasons than war, war-like events, acts of<br />

terrorism or epidemics, in which case only a proportional<br />

part would be refundable.<br />

Badges<br />

After successful registration and payment of your registration,<br />

your personal congress badge and further information<br />

will be sent to the address (in Europe only),<br />

indicated on the online registration form (registrations<br />

received/paid by March 30). All participants, registered<br />

after march 30, should pick-up their badges at the registration<br />

desk. The personalized badge is not transferrable<br />

and it is your admission card to the congress.<br />

Badge Replacement: Please do not forget or lose your<br />

badge. In case of loss, a replacement badge will only be<br />

provided against an administrative charge of $ 50.00.<br />

Security Checks<br />

For organizational and security reasons, badges have to<br />

be worn all the time at the congress venue. A lanyard will<br />

be given to you with the congress bag. ID-checks may occur<br />

at any time.<br />

Final Programme<br />

Participants will find a copy of the “Final Programme &<br />

Abstract book” in the congress bags. The bags will be<br />

handed out upon congress check-in. The congress abstracts<br />

will be available well in advance of the congress<br />

in a searchable database on our website as well as in pdf<br />

format. New this year will be that congress abstracts and<br />

the programme of interest can be downloaded to your<br />

iPhones and/or to other mobile devices.<br />

Additional copies may be purchased at the rate of<br />

€ 15.00 per book, at the registration desk.


General Information<br />

Speaker’s Ready Room / AV Centre<br />

The Speaker Ready Room is located on Congress Level 1<br />

at Room Saint Charles. All presentations must be in English<br />

and must be provided on CD-ROM or USB-Memory<br />

Stick to be placed on the central server on-site. It is mandatory<br />

that the data carriers are delivered to the Speaker<br />

Ready Room at least 3 hours prior to the respective session.<br />

The computers in the server room are equipped with<br />

Microsoft Windows 7 and Microsoft Office 2010. If you<br />

use Macintosh, please convert your Keypoint presentation<br />

or your PowerPoint Presentation for MAC into Power-<br />

Point for PC Windows format. In case of using QuickTime<br />

movies into you presentation, please take care to convert<br />

the movies into a standard video codec like MPEG<br />

2. If you have any doubt, please contact the AV Centre 4<br />

hours before your presentation. Our technicians will have<br />

enough time to verify and adapt your presentation if needed.<br />

If you use Macintosh, please convert your Keypoint<br />

presentation or your PowerPoint Presentation for MAC<br />

into PowerPoint for PC Windows format. In case of using<br />

QuickTime movies into you presentation please take care<br />

to convert the movies into a standard video codec like<br />

MPEG 2. If you have any doubt, please contact the AV<br />

Centre 4 hours before your presentation. Our technicians<br />

will have enough time to verify and adapt your presentation<br />

if needed.<br />

It will not be possible to use your own laptop or your memory<br />

stick for your presentation in the session rooms.<br />

If a presenter has included videos into the PPT presentations,<br />

she/he should make sure that the movies run<br />

on the most commonly used video software with Windows<br />

compatible codec. Example: MPEG 2.<br />

The material remains the property of the speakers and<br />

will only be re-used by ICRS with the speaker’s permission.<br />

Without this formal permission, your data will be<br />

definitely deleted after the congress.<br />

Opening hours Speaker’s Ready Room / AV Centre:<br />

Friday May 11 16.00 <strong>–</strong> 18.30<br />

Saturday May 12 08.00 <strong>–</strong> 18.30<br />

Sunday May 13 07.00 <strong>–</strong> 18.30<br />

Monday May 14 07.00 <strong>–</strong> 18.30<br />

Tuesday May 15 07.00 <strong>–</strong> 14.00<br />

Podium Presentations / Free Papers<br />

Speaking Time: 7 Minutes<br />

Discussion Time: 3 Minutes<br />

It is absolutely necessary that all podium presenters respect<br />

the given speaking time in order not to delay the<br />

entire congress schedule. Session Moderators are instructed<br />

to interrupt a presentation in case of exceeding<br />

the speaking time of 7 Minutes.<br />

Traditional Wall Poster Sessions<br />

Sunday May 13 from 16.00 <strong>–</strong> 18.00 (Wine & Cheese)<br />

Monday May 14 from 16.30 <strong>–</strong> 17.30 (Coffee Break)<br />

In addition to the electronic poster exhibit, the authors where<br />

asked to produce traditional paper posters by reserving<br />

in advance a poster wall with ICRS. All congress participants<br />

are strongly encouraged to join both poster sessions.<br />

To facilitate discussions and networking, all wall poster<br />

presenters are required to stay near their poster boards<br />

during both poster sessions. Authors should encourage<br />

a lively discussion with interested participants. The<br />

presenters should introduce themselves as poster presenters<br />

and be well prepared to answer questions and<br />

initiate discussions.<br />

The wall posters are located in rooms Duluth/ Mackenzie<br />

& Richelieu/Peribonca/Bersimis at congress level 1 and<br />

the electronic posters stations are located in Room Saint<br />

Maurice. The abstracts of the posters can be found on<br />

our website www.cartilage.org and in the electronic poster<br />

viewing system onsite.<br />

Electronic Poster Exhibition (EPOS)<br />

(partly sponsored by Geistlich Surgery)<br />

16 Workstations will be available for viewing about 250<br />

electronic scientific exhibits.<br />

The system offers great flexibility and provides enhanced<br />

opportunities for communication. The ability to use moving<br />

images, to link to related websites, to search quickly<br />

the whole of the scientific exhibition for specific topics<br />

in minute detail, to e-mail entire exhibits to one’s-self or<br />

to a colleague and to access the exhibit from any internet-linked<br />

computer in the world are amongst its many<br />

advantages besides the post congress availability of the<br />

presentations during many years.<br />

Users have also the possibility to informally score/rate<br />

the electronic posters by clicking on the “stars” in the system,<br />

besides to bookmark important ones.<br />

19


20<br />

General Information<br />

Our onsite staff will be happy to introduce you to the<br />

system and assist you during these hours.<br />

E-Poster Certificates:<br />

2 Awards “Magna cum Laude”<br />

2 Awards “Cum Laude”<br />

2 Awards “Certificate of Merit”<br />

Publication <strong>–</strong> Supplement Journal “<strong>Cartilage</strong>”<br />

Congress abstracts are citable and will be published<br />

electronically in the congress supplement of our official<br />

Journal “<strong>Cartilage</strong>”, published by Sage ISSN 1947-6043.<br />

We furthermore encourage all authors to submit their<br />

full manuscripts to our peer reviewed journal “<strong>Cartilage</strong>”<br />

which publishes full length original manuscripts on<br />

all types of cartilage including articular, nasal, auricular,<br />

tracheal/bronchial, and intervertebral disc fibrocartilage.<br />

Manuscripts on clinical and laboratory research are<br />

welcome. Instructions to authors for submissions are<br />

available at http://cart.sagepub.com<br />

General Assembly / Business Meeting<br />

(for members only)<br />

Sunday, May 13, 2012, 15.45 <strong>–</strong> 17.00 - Room Jolliet<br />

All ICRS members are kindly invited to attend our General<br />

Assembly. Retired Members and Corporate Members<br />

have no right to vote, but are most welcome to attend the<br />

Business Meeting.<br />

CME <strong>–</strong> Credits (22.25 Hours)<br />

The ICRS 2012 <strong>–</strong> 10th World Congress of the <strong>International</strong><br />

<strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong> is an Accredited Group Learning<br />

Activity (Section 1) as defined by the Maintenance of Certification<br />

programme of The Royal College of Physicians<br />

and Surgeons of <strong>Canada</strong>, and approved by The Canadian<br />

Orthopaedic Association. The ICRS 2012 is designated<br />

for a maximum of 22.25 hours of Section 1 Credits. Live<br />

educational activities, occurring in <strong>Canada</strong>, recognized<br />

by the Royal College of Physicians and Surgeons of <strong>Canada</strong><br />

as Accredited Group Learning Activities (Section 1)<br />

are deemed by the European Union of Medical Specialists<br />

(EUMS) eligible for ECMEC credits but are they not<br />

recognized by the American Medical Association AMA towards<br />

the Physician’s Recognition Award (PRA).<br />

Certificate of Attendance / CME Credits<br />

To obtain your confirmation of attendance and your CME<br />

Credits, you may choose one of two options<br />

Print your certificate onsite at the Congress<br />

Please use one of the dedicated workstations next to the<br />

ICRS Registration Desk to print out your certificate. After<br />

having identified yourself by means of your badge number<br />

and your last name, the personalized certificate of<br />

attendance will be printed automatically.<br />

Print your certificate at your home<br />

Upon your return from <strong>Canada</strong>, please visit www.cartilage.org.<br />

After having identified yourself online by means<br />

of your badge number and last name you can print-out at<br />

home your personalized certificate of attendance.<br />

Time Zone<br />

Standard time zone daylight saving: UTC/GMT -4 hours.<br />

Time zone abbreviation: EDT <strong>–</strong> Eastern Daylight Time<br />

Language<br />

The official congress language is English. No simultaneous<br />

translation will be provided. French is Québec’s official<br />

language but English is widely spoken in Montréal.<br />

The city has more than 80 cultural groups and over 20%<br />

of the population speaks three languages.<br />

Climate<br />

Spring climate in <strong>Montreal</strong> can be particularly pleasant,<br />

with temperatures beginning to rise. Late May is a<br />

popular time to visit the city, when temperatures averaging<br />

around 18°C / 64°F but occasional snow flurries in<br />

spring are not unknown.<br />

Clothing<br />

Warm & casual wear is recommended. Some <strong>Montreal</strong><br />

establishments (nightclubs and bars) have a 'no jeans,<br />

no running shoes policy,' and certain dining rooms<br />

require men to wear jackets to dinner.<br />

Credit Cards / Cash Machines<br />

All major credit cards are widely accepted. Bank cash machines<br />

can be found everywhere and debit cards are also<br />

widely used. If you are using a foreign card at a bank machine,<br />

your money will be disbursed in Canadian funds.


General Information<br />

Currency<br />

The currency in <strong>Canada</strong> is the Canadian Dollar $ CAD.<br />

1 € Euro ~ 1.30 $ CAD (Date of printing, March 2012)<br />

Tipping<br />

Tips or service charges are not usually added to a bill in<br />

<strong>Canada</strong>. In general, a tip of up to 15% of the sub-total (before<br />

taxes) is given. This applies to waiters, waitresses,<br />

barbers and hairdressers, taxi drivers, etc. At hotels, airports<br />

and railway stations, bellhops, doormen, porters,<br />

etc. are generally paid $1 <strong>–</strong> $2 per item of luggage<br />

Health Care<br />

Vaccinations are not required for entry into <strong>Canada</strong> but<br />

an individual travel- and health- insurance is highly recommended<br />

because health insurance plans often do<br />

not extend full coverage for medical services received<br />

outside the country of residence.<br />

Entry Formality / Visa<br />

We strongly recommend that all participants needing a<br />

visa to attend the Congress in <strong>Canada</strong> to start the application<br />

process as soon as possible. We also advise contacting<br />

the local Canadian Consulate before beginning<br />

the process to obtain and follow all instructions. Most<br />

consulates and embassies have web sites that can be<br />

accessed through www.cic.gc.ca.<br />

Currently, exempt from VISA requirement for tourist or<br />

business travel to <strong>Canada</strong> include: citizens of Andorra,<br />

Antigua and Barbuda, Australia, Austria, Bahamas,<br />

Barbados, Belgium, Botswana, Brunei, Croatia, Cyprus,<br />

Denmark, Estonia, Finland, France, Germany, Greece,<br />

Hungary, Iceland, Ireland, Israel (National Passport holders<br />

only), Italy, Japan, Korea (Republic of), Latvia (Republic<br />

of), Lithuania, Liechtenstein, Luxembourg, Malta,<br />

Monaco, Namibia, Netherlands, New Zealand, Norway,<br />

Papua New Guinea, Poland, Portugal, St. Kitts and Nevis,<br />

St. Lucia, St. Vincent, San Marino, Singapore, Slovakia,<br />

Solomon Islands, Spain, Swaziland, Sweden, Slovenia,<br />

Switzerland, United States, and Western Samoa. This information<br />

may change without further notice.<br />

Disclaimer<br />

ICRS and the congress organizer cannot accept any liability<br />

for the acts of any suppliers to this meeting nor the safety<br />

of any attendee while in transit to or from this event.<br />

All participants are strongly advised to carry proper travel<br />

and health insurance as the ICRS cannot accept liability<br />

for any accidents or injuries that may occur. The information<br />

in this programme is subject to change without prior<br />

notice. For updated information please visit frequently<br />

our congress online programme on our website.<br />

Cancellation of the Congress by the Organizer<br />

Congress fees will be reimbursed if the congress is cancelled<br />

for other reasons than war, warlike events, and acts<br />

of terrorism or sickness epidemics. In the latter circumstances<br />

only a proportion of the congress fee would be<br />

refundable.<br />

Technical Industry Exhibition<br />

A technical industry exhibition with will take place at<br />

the Fairmont convention centre. It will be open every<br />

day throughout the meeting and exhibitors will present<br />

a wide range of orthopaedic- and cartilage repair related<br />

products. Participants are encouraged to take advantage<br />

of this unique opportunity to be updated with the most<br />

recent advances and latest news from our industry partners.<br />

Interested companies may contact the ICRS Executive<br />

Office for further exhibit- and sponsoring information.<br />

Intermissions<br />

During intermissions, coffee, tea and refreshments will<br />

be served in the exhibition area as a courtesy from the<br />

ICRS.<br />

Services for Persons with Disabilities<br />

Please inform the ICRS office or indicate on the accommodation<br />

online booking form any special requirements<br />

you may have to allow us to best serve you. The Fairmont<br />

has international-standard facilities for the physically<br />

challenged, including access ramps, elevators and toilets<br />

for people with special needs.<br />

Security / Badges<br />

The safety of all congress attendees is of utmost importance<br />

to our society. ICRS and the Fairmont have taken<br />

security precautions to ensure the maximum possible<br />

safety for all participants. Identity check controls may occur<br />

at any time by the security staff. Congress badges are<br />

personalized, not transferable and guarantee individual<br />

access to different section of the event. For organizational<br />

and security reasons, badges have to be worn all the<br />

time at the congress venue.<br />

21


22<br />

General Information<br />

Electricity, Weights and Measures<br />

• Electricity: 110 volts 60 hz (US Sockets)<br />

• Weights: Kilo/Gramm system<br />

• Measures: Metric system<br />

Water<br />

Canadian tap water is among the cleanest in the world<br />

and is safe to drink.<br />

Internet/WLAN<br />

As a special courtesy, ICRS will provide free WLAN Hot<br />

Spots and a public Internet corner to all attendees and<br />

exhibitors.<br />

Meals, Snacks & Refreshments<br />

A Restaurant & Bar near the Hotel Lobby will be at<br />

your disposal during the opening hours. Delegates<br />

may purchase light meals, snacks, sandwiches, salads,<br />

sweets and soft drinks against payment. No lunch or<br />

lunch boxes will be provided by the ICRS. However, most<br />

Industry Satellite Symposia organizers will offer exclusive<br />

lunch boxes during their symposia at lunch time.<br />

During morning intermissions, coffee, tea and refreshments<br />

are served in the exhibition area. The coffee<br />

breaks are offered by ICRS to all delegates and company<br />

representatives.<br />

Mobile Phones<br />

Please turn off or put your mobile phone to the “silentmodus”<br />

during all scientific sessions.<br />

Photos/Recording<br />

Taking pictures, video- or audio- recording during presentations<br />

is not allowed.<br />

Security<br />

The safety of all congress attendees is of the utmost<br />

importance to the ICRS meeting. The conference centre<br />

and ICRS have taken security precautions to ensure the<br />

maximum possible safety for all the participants. Identity<br />

check controls may occur at any time.<br />

Smoking<br />

The ICRS World Congress is a non-smoking congress.<br />

Smoking is not permitted at the conference centre except<br />

in designated smoking areas outside of the building.<br />

Opening Ceremony, ICRS Awards<br />

Saturday May 12, 2012, 17.00 <strong>–</strong> 17.45<br />

Room: Grand Salon<br />

Awards<br />

The following offical ICRS honors will be awarded:<br />

• ICRS <strong>–</strong> Genzyme Lifetime Award<br />

• ICRS <strong>–</strong> Genzyme Award for Excellence in <strong>Cartilage</strong><br />

Research (US$ 5000.00)<br />

• 6 Electronic Poster Awards<br />

• Best Rated Abstract


Hotel and Travel Information/Official Flight Carrier<br />

Fairmont <strong>–</strong> The Queen Elizabeth Hotel<br />

(Official Hotel & Congress Venue)<br />

At the centre of Montréal's vibrant cultural and commercial<br />

district sits the city's grandest and most gracious hotel<br />

Fairmont - The Queen Elizabeth with 1037 comfortable<br />

rooms and suites.<br />

Located above the train station (Via Rail & AMTRAK) and<br />

connected to the extensive underground city of thousands<br />

of boutiques, restaurants and cafés, and within<br />

walking distance of sports and cultural attractions, this<br />

<strong>Montreal</strong> hotel reflects the city’s distinct elegance and<br />

charm. The Fairmont comprises a skillfully integrated<br />

Health Club featuring state-of-the-art equipment and an<br />

indoor pool, as well as three distinctive restaurants to experience<br />

Montréal's gastronomy at its best.<br />

Special Courtesy<br />

As a special courtesy, the hotel offers complimentary<br />

health club access and free high speed internet access<br />

in the hotel rooms. Children under 18 years, sharing the<br />

room with their parents are free of charge. Each additional<br />

adult beyond double room occupancy is charged<br />

with only CAD$ 40.00.<br />

Room Rates<br />

(Breakfast & approx. 18% Taxes not included)<br />

Fairmont Room: CAD$ 189.00 (Sgl/Dbl)<br />

Moderate Room*: CAD$ 165.00 (Sgl/Dbl)*<br />

Junior Suite: CAD$ 289.00 (Sgl/Dbl)<br />

Fairmont Gold**: CAD$ 309.00**<br />

* Moderate rooms available to students/residents<br />

& physiotherapists only<br />

** Business Club Floor, rate includes breakfast<br />

Reservation Department<br />

Fairmont <strong>–</strong> The Queen Elizabeth Hotel<br />

c/o Ms. Mattea Savino<br />

Phone: +1 514 861 3511, Extension: 2463<br />

Email: mattea.savino@fairmont.com<br />

Toll Free<br />

U.S. & <strong>Canada</strong>: 1 (800) 257-7544<br />

<strong>International</strong>: 800 441 1414<br />

Travel and Transportation<br />

The Montréal-Pierre Elliott Trudeau <strong>International</strong> Airport<br />

(YUL) is only a 20 Minutes Taxi-drive away from the Hotel<br />

Fairmont (14 Miles or 22 KM).<br />

• Taxi: There is a flat rate of around CAD$ 40.00 to downtown<br />

hotels. Arrangements for a private limousine transfer<br />

can be made with through your hotel concierge upon<br />

request.<br />

• 747 Express Bus: Featuring nine stops in each direction,<br />

the 747 service is provided 24 hours a day, 365 days a year<br />

transportation between downtown Montréal (this bus stops<br />

right in front of the Fairmont) and Montréal-Pierre Elliott<br />

Trudeau <strong>International</strong> Airport. For more information visit<br />

the STM website: www.stm.info/english/info/a-747.htm<br />

Official Airline Network <strong>–</strong> Code AC04S12<br />

SAVE UP TO 20% ON TRAVEL WITH THE STAR ALLIANCE<br />

NETWORK<br />

The Star Alliance member airlines are pleased to be<br />

appointed as the Official Airline Network for ICRS 2012<br />

<strong>Montreal</strong>, <strong>Canada</strong>.<br />

To obtain the Star Alliance Conventions Plus discounts<br />

and for booking office information please visit www.staralliance.com/conventionsplus<br />

and:Choose “For delegates”<br />

<strong>–</strong> Under “Delegates login” enter the conventions code<br />

AC04S12. Choose one of the participating airlines listed<br />

or call the respective reservation contact listed and quote<br />

the conventions code AC04S12 when booking the ticket.<br />

Registered participants plus one accompanying person<br />

travelling to the event can qualify for a discount of up to<br />

20%, depending on fare and class of travel booked.<br />

The participating airlines for this event are: Adria Airways,<br />

Aegean Airlines, Air <strong>Canada</strong>, Air China, ANA, Asiana<br />

Airlines, Austrian Airlines, Blue1, bmi, Brussels Airlines,<br />

Continental Airlines, Croatia Airlines, EgyptAir, LOT<br />

Polish Airlines, Lufthansa, Scandinavian Airlines, Singapore<br />

Airlines, South African Airways, Spanair, SWISS<br />

<strong>International</strong> Air Lines, TAM Airlines, TAP Portugal, THAI,<br />

Turkish Airlines, United, US Airways<br />

Discounts are offered on most published business and<br />

economy class fares, excluding website/internet fares,<br />

senior and youth fares, group fares and Round the World<br />

fares. Please note: For travel from Japan and New Zealand<br />

special fares or discounts are offered by the participating<br />

airlines on their own network.<br />

23


As the official airline network for ICRS 2012, we’d like to<br />

thank you for choosing the Star Alliance network and hope<br />

that all goes really well for you here today.<br />

Whilst you concentrate on the day’s events, we hope you’ll<br />

consider us the next time you need to attend a conference.<br />

With over 21,000 flights a day to 1,185 airports across 189 countries,<br />

our 26 member airlines will extend a wide choice of flights to any<br />

future conference you’re planning to attend. And no matter which<br />

of those airlines’ frequent flyer programmes you belong to, you<br />

can earn and redeem miles across all of them.<br />

So the next time you want to concentrate all your energies<br />

on your conference, we hope you’ll decide to leave the travel<br />

arrangements to us.<br />

www.staralliance.com<br />

Information correct as at 02/2012


Social Events<br />

Saturday, May 12<br />

Welcome Reception<br />

from 19.00 <strong>–</strong> 20.30<br />

All participants, industry representatives and accompanying<br />

persons are invited to join the opening ceremony<br />

and welcome cocktail at the Fairmont exhibition area.<br />

This reception is offered to you by the ICRS.<br />

After the cocktail, participants have free time for their<br />

own leisure to discover <strong>Montreal</strong> and enjoy one of the<br />

many nice restaurants / bars that <strong>Montreal</strong> offers to you.<br />

Sunday, May 13<br />

2 nd ICRS Fun Run for Healthy Joints<br />

from 06.15 <strong>–</strong> 07.00 AM<br />

ICRS and the Fairmont understand that maintaining a<br />

fitness routine while traveling is difficult. Congress participants<br />

wanting to get off the treadmill and take their<br />

workout outside can join the complimentary ICRS Fun<br />

Run led by members of the ICRS & Fairmont management<br />

teams. Runners meet at 6:00 A.M. in the Lobby for<br />

a four-mile jog through Old <strong>Montreal</strong>, the Latin Quarter,<br />

on Mount-Royal or along the Lachine Canal returning by<br />

7:00 A.M.<br />

Sunday, May 13<br />

Canadian Sugar Shack & Maple Forest Party<br />

from 19.30 <strong>–</strong> 23.00<br />

We transport you back in time and provide you with a<br />

momentary glimpse of life as it was hundreds of years<br />

ago for Québec and Canadian pioneers. You will enjoy<br />

Québec folkloric traditions in an authentic “Sugar<br />

Shack” setting in a maple wood farm, known as a place<br />

where Québec heritage is respected and relived through<br />

fun and traditional activities. Discover Quebec folklore<br />

and enjoy its traditional sugaring-off party in true family<br />

style. Dine and let you be immersed in a magical and<br />

unforgettable atmosphere together with your colleagues<br />

from all around the world. Here, you can look forward to<br />

delicious specialties, regional beverages and enjoy typical<br />

music and cultural show acts.<br />

Price per person:<br />

Individual Participants: € 85.00<br />

Industry Representatives: € 110.00<br />

(incl. transport, dinner, drinks & entertainment)<br />

Monday, May 14<br />

Free evening for participants’ individual programmes<br />

and for industry related events. The Hotel Concierge will<br />

be pleased to recommend and book a table for you at one<br />

of your favourite restaurants in town.<br />

25


26<br />

Points of interest<br />

Local Attractions, Sightseeing and things to do in <strong>Montreal</strong>….<br />

The following attractions are only a small selection of<br />

points of interest in <strong>Montreal</strong> and do not constitute a<br />

recommendation by the ICRS. More information and<br />

reservation for tours are available through the hotel concierge.<br />

Botanical Garden and Insectarium<br />

<strong>Montreal</strong>‘s Botanical Garden is the second largest botanical<br />

garden in the world, next to the one in London,<br />

England. It features species from all over the world:<br />

plants, fountains, thematic greenhouses, butterflies. The<br />

Insectarium is a section of the Botanical Gardens featuring<br />

the butterfly pavilion, as well as a collection of 25,000<br />

insects on display. The attraction is located 10 minutes<br />

away from the Fairmont.<br />

Bell Centre<br />

The Bell Centre opened on March 16, 1996, and is the<br />

home to the National Hockey League‘s <strong>Montreal</strong> Canadiens.<br />

It is a five minute walk from the hotel Fairmont<br />

The Queen Elizabeth. Hockey tickets can be purchased<br />

directly through our concierge. Le Centre Bell also offers<br />

guided tours.<br />

Six Flag La Ronde Amusement Park<br />

Six Flags La Ronde Amusement Park features 35 different<br />

rides, something for every member of the family. The<br />

park is located on Ile-Ste Hélène, about a 15 minute drive<br />

from the Fairmont. It is also accessible via subway. It is<br />

open from mid-May to September. For more information,<br />

please call 1 800 797-4537 or 514 872-4537.<br />

<strong>Montreal</strong> Casino<br />

The <strong>Montreal</strong> Casino is one of the largest casinos in the<br />

world! It captures all the excitement, all the drama, all<br />

the international allure of the world class city that is its<br />

home: Montréal. Likewise, the Casino spares no effort<br />

to ensure you the thrill of a lifetime and unforgettable<br />

memories.<br />

Mount Royal<br />

Mount Royal is a small mountain and park overlooking<br />

the city, which offers great views from several scenic<br />

points. Prominent in St. Joseph‘s Oratory, with its copper<br />

dome surpassed in size only by St. Peter‘s in Rome.<br />

Those who enjoy strolling in nature will appreciate Mount<br />

Royal‘s many paths. Bird watching is possible at the bird<br />

sanctuary. It is also the home of the University of <strong>Montreal</strong>,<br />

the second largest French-language University in<br />

the world.<br />

Old <strong>Montreal</strong><br />

Discover the city‘s oldest district with its wealth of history<br />

and architecture unique in North America, a variety of<br />

museums, boutiques, art galleries and restaurants. Other<br />

<strong>Montreal</strong> sightseeing opportunities and attractions in the<br />

old port will please the whole family: IMAX (3-D movies<br />

on a giant screen), bateau mouche cruises, and the site of<br />

several summer and winter festivals. There‘s also the SOS<br />

Labyrinth (Mayaventura) which is great for kids, cycling and<br />

rollerblading. As well, don‘t miss the Lachine Canal with<br />

its 8.7 miles (14 km) of recreational paths. Old <strong>Montreal</strong> is<br />

located about five minutes from the Fairmont.<br />

Olympic Stadium<br />

Across the street from the Botanical Garden, the Olympic<br />

Stadium is located about 15 minutes away from the Fairmont<br />

and is easily accessible by subway. Visit the Montréal<br />

Tower Observatory, located in the highest inclined<br />

tower in the world, with its 175 metre elevation and<br />

45-degree angle. Also on-site is the Biodôme, an oasis<br />

in the heart of the city. The Montréal Biodôme recreates<br />

some of the most beautiful ecosystems of the Americas.<br />

A must-see!


Invited Faculty 2012 in alphabetical order (not complete)<br />

Amendola Annunziato (Ned), Prof., MD<br />

Orthopaedics and Rehabilitation,<br />

University Iowa, USA<br />

Annunziato (Ned) Amendola, MD, Professor<br />

of orthopaedics and rehabilitation at the University of Iowa is<br />

the director of the University of Iowa Sports Medicine program<br />

and Head team physician. He currently holds the Kim and John<br />

Callaghan Endowed Chair in sports medicine. He is internationally<br />

recognized in sport related and reconstructive surgery<br />

of the knee and ankle. His clinical research interests focus on<br />

improving the understanding, prevention, treatment, and rehabilitation<br />

of sports and activity-related injuries. His basic research<br />

interests include articular cartilage biology and healing,<br />

effects of unloading and joint resurfacing. Dr Amendola earned<br />

his medical degree and completed his orthopaedic residency<br />

at the University of Western Ontario in London, Ontario. Prior<br />

to joining the University of Iowa in 2001, he was an associate<br />

Professor and chief of orthopaedic surgery at the University of<br />

Western Ontario University Hospital. Dr Amendola is a Diplomate<br />

of the American Board of Orthopaedic Surgery, Fellow of<br />

the Royal College of Surgeons of <strong>Canada</strong>, and Diplomate of the<br />

Canadian Academy of Sports Medicine. He is an active member<br />

of many orthopaedic and sports medicine organizations,<br />

including Director on the American Academy of Orthopaedic<br />

Surgeons, American Board of Orthopaedic Surgery, and Board<br />

of Directors of American Orthopaedic <strong>Society</strong> for Sports Medicine<br />

and ISAKOS. He is a past president of Canadian Academy of<br />

Sports Medicine (1997).<br />

Apprich Sebastian, MD<br />

Medical University of Vienna, High Field MR -<br />

Centre, Vienna, Austria<br />

Sebastian Apprich is a member of the High Field<br />

MR-Centre of Excellence (under the direction of Univ. Prof. Dr.<br />

Siegfried Trattnig) at the Department of Radiology at the Medical<br />

University of Vienna since 2008 and has a special interest in<br />

the development and implementation of new MR techniques in<br />

clinical orthopaedic research. He received his MD at the Medical<br />

University of Innsbruck in 2010. As an undergraduate he already<br />

demonstrated his growing interest in orthopaedic imaging research<br />

and investigated the potential of new isotropic MR - sequence<br />

for the detection of cartilage defects and meniscal tears<br />

for his doctoral thesis. He spent one year as a postdoctoral fellow<br />

at the Department of Orthopaedic Surgery at the Inselspital<br />

Bern (Switzerland; under the direction of Univ. Prof. Dr. Klaus<br />

Siebenrock), concentrating on new MR imaging modalities for<br />

early detection of cartilage defects in femoroacetabular impingement<br />

patients. In 2011, Sebastian Apprich returned to Vienna<br />

to the High Field MR-Centre of Excellence where he is in charge<br />

of the planning and execution of a number of musculoskeletal<br />

imaging studies. After several years of research, he has begun<br />

specialized clinical training in orthopaedics. Sebastian Apprich’s<br />

research has been focused on imaging cartilage and its repair,<br />

with main focus on biochemical quantitative MRI techniques.<br />

Although he is still young, a number of author- and co-authorships<br />

in top journals attest to his specialist knowledge in MRI.<br />

Barry Frank, Prof., MD, PhD<br />

National Centre for Biomedical Engineering<br />

Science (NCBES),National University of Ireland,<br />

Galway, Ireland<br />

Frank Barry is Professor of Cellular Therapy at the National University<br />

of Ireland Galway, Director of the University’s National<br />

Centre for Biomedical Engineering Science (NCBES) and a principle<br />

investigator at the Regenerative Medicine Institute (REME-<br />

DI). Here he directs a large group of researchers who focus on<br />

the development of new repair strategies in stem cell therapy<br />

and gene therapy in orthopaedics. Previously he was Director of<br />

Arthritis Research at Osiris Therapeutics in Baltimore, MD and a<br />

Research Fellow at Shriners hospital for Children, Tampa, FL. He<br />

has contributed to the fields of tissue engineering and regenerative<br />

medicine by developing innovative and successful cellular<br />

therapies for the treatment of acute joint injury and arthritic disease.<br />

This has included the generation of a large body of new<br />

data in ground-breaking preclinical studies, and has lead to the<br />

first phase of clinical testing of mesenchymal stem cells in clinical<br />

trials for joint injury. In a career that has spanned both industry<br />

and academic research; he has been a driver in the development<br />

of cellular therapy as a biological repair strategy. It is his<br />

belief that the application of new technologies in regenerative<br />

medicine, including cellular therapy, gene therapy, growth factor<br />

augmentation, implantable scaffolds and nonmaterial, will have<br />

a profound impact in Orthopaedics.<br />

Beier Frank, Prof., PhD<br />

Department of Physiology & Pharmacology,<br />

University of Western Ontario, USA<br />

Frank Beier received his PhD in Biology at the<br />

University of Erlangen-Nürnberg, Germany, in 1995. He is now<br />

a Professor in the Department of Physiology & Pharmacology<br />

at The University of Western Ontario. Dr. Beier is the current director<br />

of the Collaborative Graduate Program in Developmental<br />

Biology at The University of Western Ontario. He holds a Tier II<br />

<strong>Canada</strong> Research Chair in Musculoskeletal Health, received a<br />

Dean's Award of Excellence for Graduate Student Teaching in<br />

2008 and a Faculty Scholar Award in 2009. He was the 2010<br />

Scientist of the Year at the Children's Health Research Institute.<br />

Dr. Beier’s lab works on the signalling pathways and molecular<br />

mechanisms that regulate the biology of cartilage cells (chondrocytes)<br />

and other skeletal cells, both during development and<br />

in osteoarthritis. In this research, the Beier lab uses a combination<br />

of gene expression profiling, in-depth studies of identified<br />

genes in genetically altered mice and surgical models in mice<br />

and rats. Beier’s studies are currently funded by CIHR and NIH.<br />

Currently he is a member of CIHR and NIH peer review committees,<br />

of the Faculty of 1000 and the Editorial Board of PLoS One.<br />

27


28<br />

Invited Faculty 2012 in alphabetical order (not complete)<br />

Bonassar Lawrence, Ass. Prof., PhD<br />

Cornell University. Biomedical Engineering,<br />

Ithaca, New York, USA<br />

Brittberg Mats, Ass. Prof., MD, PhD<br />

Orthopaedic Department, <strong>Cartilage</strong> Research<br />

Unit, Göteborg University, Sweden<br />

Mats Brittberg is a member of the <strong>Cartilage</strong> Research<br />

Unit, Department of Orthopaedics, at<br />

University of Gothenburg and orthopaedic surgeon at Kungsbacka<br />

Hospital, Sweden. He received his MD in 1978 and completed<br />

a specialization in orthopaedics in 1985. In 1992 he passed the<br />

Swedish Orthopaedic Board Exam (S.O.B.E.) and earned a PhD<br />

in 1996. In 2002, he became Associate Professor of orthopaedics<br />

at University of Gothenburg and now also Gothenburg university<br />

lecturer. Mats Brittberg’s research has been focused on<br />

cartilage regeneration with autologous chondrocytes. Today the<br />

main interest is the European Connective Tissue Engineering<br />

centre (ECTEC) which is a collaboration between the Sahlgrenska<br />

Academy at University of Gothenburg and Chalmers Technical<br />

University. The research focus on musculoskeletal tissue engineering<br />

and pain mechanisms in joint injuries and osteoarthritis.<br />

Mats Brittberg has also had research collaboration with Virginia<br />

Tech in USA on biotribology in cartilage as well as research<br />

collaborations with other centres in Europe and North America.<br />

Mats Brittberg has been on the board of TESi (Tissue engineering<br />

<strong>Society</strong> <strong>International</strong>) and has been chairing the <strong>Cartilage</strong><br />

Committee of ESSKA 2006-08. Since the start 1997, he has been<br />

working with ICRS, as a secretary, Vice-president and President<br />

(2006-2008) and finally Past-President (2008-2009). He is now<br />

associate editor for the Sage journal ¨CARTILAGE¨ and associate<br />

editor with ESSKA journal and in the editorial board of Osteoarthritis<br />

and <strong>Cartilage</strong>. In September, 2010, Mats Brittberg<br />

received the ICRS Genzyme Lifetime Achievement Award in cartilage<br />

research. Mats Brittberg is also clinical representative in<br />

the program committee of ORS for the meeting in 2013.<br />

Buckwalter Joseph, Prof., MD<br />

Orthopaedics and Rehabilitation,<br />

University of Iowa, USA<br />

Dr. Joseph A. Buckwalter is Professor and chairman<br />

of Orthopaedics and Rehabilitation and Arthur Steindler<br />

Chair of Orthopaedics at the University of Iowa. His clinical practice<br />

and research interests include osteoarthritis, joint injuries<br />

and primary tumours of the skeleton and musculoskeletal soft<br />

tissues. He has served as chairman of the American Academy<br />

of Orthopaedic Surgeons Council on Research and president of<br />

the Orthopaedic Research <strong>Society</strong>, the American Board of Orthopaedic<br />

Surgery, and the American Orthopaedic Association.<br />

His research awards include the Kappa Delta Award, the Cabaud<br />

Award for Research in Sports Medicine, the American Orthopaedic<br />

Association Award for Distinguished Achievement in Ortho-<br />

paedic Research, the Orthopaedic Research <strong>Society</strong> and American<br />

Orthopaedic Association Alfred Shands Award for Research,<br />

and the Orthopaedic Research <strong>Society</strong>-Orthopaedic Research<br />

and Education Foundation Distinguished Investigator Award. He<br />

is the Senior Editor of the Journal of Orthopaedic Research. He is<br />

a member of the Royal College of Surgeons of Edinburgh and the<br />

Institute of Medicine, National Academies of Sciences, and is<br />

the author of more than 500 scholarly publications. His current<br />

research involves study of the pathogenesis of post-traumatic<br />

osteoarthritis and methods of preventing osteoarthritis following<br />

joint injuries.<br />

Bugbee William, Prof., MD<br />

University of California, Department of<br />

Orthopaedics La Jolla, San Diego, USA<br />

Bill Bugbee is attending physician at Scripps<br />

Clinic, La Jolla and Professor, department of Orthopaedics,<br />

university of California, San Diego. His clinical interests are in<br />

arthritis surgery of the hip, knee and ankle, joint replacement,<br />

osteochondral allograft transplantation and cartilage restoration.<br />

Research interests include biologic response to implants,<br />

innovation in knee replacement technique and design, Osteochondral<br />

transplantation, cartilage tissue engineering and biologic<br />

joint repair.<br />

Buschmann Michael, Prof., PhD<br />

Director Biomedical Science and Technology<br />

Research Group (FRSQ), École Polytechnique<br />

de <strong>Montreal</strong>, <strong>Canada</strong><br />

Michael Buschmann received a B. Engineering Physics from the<br />

University of Saskatchewan in 1984, and a Ph.D. in Medical Engineering<br />

and Medical Physics from the Division of Health Sciences<br />

and Technology at the Massachusetts Institute of Technology<br />

and Harvard University in 1992. His postdoctoral training in<br />

cartilage microscopy and histology was then completed at the<br />

University of Bern in Switzerland in 1994. Since 1994, Dr. Buschmann<br />

has established a multidisciplinary research program at<br />

École Polytechnique (http://www.polymtl.ca/tissue/) that focuses<br />

on the use of biomaterials to repair joint tissues, gene<br />

therapy and diagnostic technologies to assess the function of<br />

articular cartilage. Work that has been translated to industry as<br />

medical devices include a biomaterial that stimulates cartilage<br />

repair (BST-CarGel) and an arthroscopic instrument that maps<br />

articular cartilage function (Arthro-BST). Dr. Buschmann is Director<br />

of the FRSQ Group in Biomedical Science and Technology<br />

and has received the Innovator Prize from the Quebec Association<br />

for Industrial Research (ADRIQ), the Melville Medal from<br />

the American <strong>Society</strong> of Mechanical Engineers (ASME), and an<br />

Award of Merit of the Canadian Arthritis Network of Centres of<br />

Excellence.


Invited Faculty 2012 in alphabetical order (not complete)<br />

Caborn David, MD<br />

Jewish Hospital, Louisville, Kentucky, USA<br />

Caterson Bruce, Prof., PhD<br />

Cardiff School of Biosciences, connective tissue<br />

biology lab, Wales, UK<br />

Bruce Caterson, Prof., of Biochemistry<br />

in the Connective Tissue Biology Group within<br />

the School of Biosciences, Cardiff University,<br />

Wales, U.K.<br />

Over the past 27 years Professor Bruce Caterson’s research has<br />

focussed on the production, development and use of monoclonal antibody<br />

(mAb) technologies for studies of connective tissue proteoglycan<br />

metabolism in health and disease. These studies have focussed<br />

on matrix proteoglycan metabolism in musculoskeletal tissues with<br />

a particular emphasis on studies involving molecular mechanism underlying<br />

the pathogenesis of degenerative joint diseases; i.e. osteoarthritis<br />

and rheumatoid arthritis. Our lab has now developed and<br />

characterised numerous mAbs that recognise both carbohydrate and<br />

protein epitopes and neoepitopes that are present on proteoglycans<br />

in all connective tissues throughout the body. Many of these mAbs<br />

are now commercially available to researchers worldwide.<br />

He has degrees from Monash University, Clayton, Victoria, Australia,<br />

(B.Sc. and Ph.D. in Biochemistry, 1971 & 1976, respectively).<br />

From 1975<strong>–</strong>1995 he spent 20 years in academia in the USA;<br />

1975<strong>–</strong>82, Postdoc <strong>–</strong> Assistant Professor, UAB, AL; 1982<strong>–</strong>89, Associate<br />

Professor <strong>–</strong> Professor, West Virginia University; 1989<strong>–</strong>95,<br />

Professor & Endowed Chair in Orthopaedic Research, University<br />

of North Carolina at Chapel Hill, NC. In 1995 he moved to Cardiff<br />

University into an Established Chair and from 1998 - 2003 was<br />

Head of Connective Tissue Biology in the School of Biosciences.<br />

He is currently Associate Director of Musculoskeletal Research in<br />

the School of Medicine.<br />

In the past, he has served on several USA national research committees<br />

(N.I.H. Pathobiochemistry; Arthritis Foundation & Orthopaedic<br />

Research and Education Fund), been a member of Editorial<br />

Boards (J. Biol. Chem., Archives of Biochemistry and Biophysics,<br />

and Osteoarthritis & <strong>Cartilage</strong>). He was also the past-President<br />

(1993) and Board of Directors member of the USA-based Orthopaedic<br />

Research <strong>Society</strong> (1988<strong>–</strong>1996). In the UK, he has served<br />

on the Wellcome Trust Cell & Molecular Grant Review Panel, been<br />

President of the <strong>Society</strong> for Back Research and was past-Chairman<br />

of the British <strong>Society</strong> for Matrix Biology and past-President of the<br />

British Orthopaedic Research <strong>Society</strong>.<br />

His primary research interests have centred around using monoclonal<br />

antibody technologies to study matrix proteoglycan<br />

structure, function and metabolism in health and disease with<br />

particular emphasis on musculoskeletal tissues. Recent research<br />

has focussed on the glycobiology of chondroitin/dermatan sulphate<br />

glycosaminoglycans in the stem/progenitor cell niche. In<br />

the past 37 years he has published a total of 170 full papers and<br />

27 chapters and reviews. In 1986 he was awarded the Benedum<br />

Distinguished Scholar Award in Biosciences and Medicine from<br />

West Virginia University, in 1998 the Kappa Delta Elizabeth Win-<br />

ston Lanier Award for Outstanding Orthopaedic Research from<br />

the American Academy of Orthopaedic Surgeons and Orthopaedic<br />

Research <strong>Society</strong> and recently, in 2011 the Fell-Muir Award<br />

from the British <strong>Society</strong> for Matrix Biology.<br />

Chubinskaya Susan, Prof., PhD<br />

Rush University Medical Center, Chicago, USA<br />

Susan Chubinskaya, PhD, The Ciba-Geigy, Professor<br />

of Biochemistry; has a primary appointment as Professor of Biochemistry<br />

with secondary appointments as Professor of Internal<br />

Medicine (Section of Rheumatology) and Orthopaedic Surgery at<br />

Rush University Medical Center. She is also a member of <strong>Cartilage</strong><br />

Restoration Research Core at Rush. Susan was born in Kiev, Ukraine<br />

and received her Ph.D. in 1990 from the Department of Metastasis,<br />

Institute of Oncology, Ukrainian Academy of Sciences, Kiev, Ukraine.<br />

In 1992 she immigrated with her family to the United States.<br />

From 1993 to 1996 she was a postdoctoral fellow at the Department<br />

of Biochemistry at Rush. In 1996 she joined the faculty at Rush Medical<br />

College. She is an internationally recognized expert in the<br />

field of growth factors/bone morphogenetic proteins in cartilage<br />

repair and regeneration. She is a Recipient of William J. Stickel Gold<br />

award, Am. Pod. Med. Assn., 1997, Eugene T. Nordby MD Research<br />

Award, Internat. Intradiscal Therapy Soc., 2005, Ciba-Geigy Endowed<br />

Chair award, Rush University Medical Center, 2007<strong>–</strong>; grantee<br />

Research grant, NIH/NIA, 1998<strong>–</strong>99, NIH/NIAMS, 2002-2006, 2009.<br />

Her research is continuously sponsored by Pharmaceutical and Biotech<br />

Companies (Proctor & Gamble, Stryker Biotech, Glaxo Welcome,<br />

Hoechst Pharmaceutical, Regentis, Arthrex, Zimmer, etc).<br />

She is a member of the OARSI Communication Committee, ORS<br />

program committee, and ICRS Executive Board/Treasurer. Susan<br />

published 5 book chapters, 56 peer-reviewed articles, and more<br />

than 130 peer-reviewed abstracts. Susan’s research focuses on the<br />

biology of cartilage and intervertebral disc homeostasis in physiological<br />

and pathophysiological processes. More specifically, her<br />

laboratory studies the mechanisms of interaction between growth<br />

factors, pro-inflammatory cytokines and neuromediators in articular<br />

cartilage and intervertebral disc; the mechanisms responsible<br />

for cartilage degeneration in aging, trauma, and osteoarthritis, molecular<br />

mechanisms of tissue engineering, and the development of<br />

diagnostic and prognostic biomarkers in arthritis.<br />

Cole Brian, Prof., MD, MBA<br />

Chairman Department of Surgery, Rush Oak<br />

Park Hospital, Rush University Medical Center, USA<br />

Brian Cole is a Professor in the Department of<br />

Orthopaedics with a conjoint appointment in the Department<br />

of Anatomy and Cell Biology at Rush University Medical Center<br />

in Chicago, Illinois. He is the Section Head of the <strong>Cartilage</strong> Research<br />

Program at Rush University Medical Center and the <strong>Cartilage</strong><br />

Restoration Center at Rush, a multidisciplinary program<br />

specializing in the restoration of articular cartilage and meniscal<br />

deficiency. He also serves as the head of the Orthopaedic<br />

Master's Program and trains residents and fellows in sports medicine.<br />

Most recently, he received appointment as the Chair of<br />

Surgery at Rush Oak Park Hospital. Dr. Cole received his MD and<br />

29


30<br />

Invited Faculty 2012 in alphabetical order (not complete)<br />

MBA from the University of Chicago in 1990, completed his residency<br />

at The Hospital for Special Surgery in New York in 1996,<br />

and his Sports Medicine Fellowship at the University of Pittsburgh<br />

in 1997. He specializes in arthroscopic shoulder, elbow<br />

and knee surgery. He is the principal investigator for numerous<br />

FDA clinical trials and regularly performs basic science research.<br />

He has authored and edited several hundred peer-reviewed publications,<br />

including highly recognized orthopaedic textbooks<br />

on arthroscopy, sports medicine and cartilage transplantation.<br />

Dr. Cole is the team physician for the Chicago Bulls NBA Basketball<br />

team, co-team physician for the Chicago White Sox Major<br />

League Baseball team and DePaul University in Chicago.<br />

Dahlberg Leif, Prof., MD, PhD<br />

Head of Joint and Soft Tissue Unit, Department<br />

of Orthopaedics, Skåne University Hospital,<br />

Lund University, Sweden<br />

Leif Dahlberg is Professor and Head of Joint and Soft Tissue Unit,<br />

Department of Orthopaedics, Skåne University Hospital, Lund<br />

University, Sweden. He received his MD at Karolinska Institutet,<br />

Stockholm in 1983. In 1994 he earned a PhD in Lund. In 1998<br />

he became Associate Professor at Lund University and in 1999<br />

Staff Surgeon. University Lecturer in 1999 and Professor of Orthopaedics<br />

in 2008 at Lund University. The research group as<br />

follows: Leif Dahlberg is main supervisor to seven PhD-students<br />

and co-supervisor to three PhD-students. The group includes<br />

three post-docs, research coordinator, secretary, university administrator,<br />

statistician and clinical research nurse/study coordinator.<br />

Dahlberg has research collaborators at Lund University,<br />

the Medical Radiation Physics and the Medical Radiology Unit at<br />

Skåne University Hospital in Malmö. He has also collaboration<br />

with the Sahlgrenska Academy at University of Gothenburg and<br />

the Swedish University of Agricultural Sciences in Uppsala. His<br />

international collaborators are in <strong>Canada</strong>, USA, Japan, South Africa<br />

and Finland. Dahlberg’s research focuses on joint health and<br />

disease in order to optimize non-operative treatment of joint disease.<br />

Research includes cartilage molecular studies, MRI and<br />

clinical interventions, specifically using human models of OA development<br />

in post-injured patients. Dahlberg is Director of the<br />

National Quality Registry, BOA (Better management of patients<br />

with Osteoarthritis), Expert consultant of the National Board of<br />

Health and Welfare (SoS) and Expert consultant of the Swedish<br />

Association of Local Authorities and Regions (SKL). Dahlberg<br />

has published more than 100 scientific articles, is cited more<br />

than 2700 times and has an h-index of 27.<br />

Daisuke Sakai, Ass. Prof.<br />

Department of Orthopaedic Surgery,<br />

Tokai University School of Medicine, Isehara,<br />

Kanagawa, Japan<br />

His clinical interest is in management of various spinal diseases,<br />

including complex spinal deformity and scoliosis. He also has<br />

particular interest in application of functional rehabilitation for<br />

spinal problems. Dr. Sakai's current researches focus on maintenance<br />

of homeostasis and intrinsic stem cell system of the<br />

intervertebral disc cells and stem cell therapy, investigation of<br />

master transcription gene network of the intervertebral disc cell<br />

lineage, specifically the role of TGF-beta/Wnt/Notch signalling.<br />

Dr. Sakai has given over 110 presentations, invited lectures and<br />

seminars, in universities, professional societies, international<br />

conferences (invited lecturer of 2004 and 2010 Gordon Research<br />

Conference in musculoskeletal biology and bioengineering and<br />

cartilage biology and pathology), and government organizations<br />

around the world. He actively participated in grant review for<br />

NIH, NSERC, SNF, RGC (Hong Kong) and other funding agencies,<br />

and serves as an editorial board member for Journal of Orthopaedic<br />

Science, reviewer for Arthritis &Rheumatism, Arthritis Research<br />

& Therapy, Journal of Orthopaedic Research and Spine.<br />

He also serves as committee member of the international affairs<br />

committee of the Japanese Orthopaedic Association.<br />

De Bari Cosimo, Prof., MD, PhD<br />

Translational Medicine at the University<br />

of Aberdeen, UK<br />

Cosimo graduated in Medicine (maxima cum<br />

laude) from the University of Bari (Italy), where he also underwent<br />

specialist training in Rheumatology. He then moved to Belgium,<br />

where he obtained his PhD from the Catholic University<br />

of Leuven and was recipient of the Rotary Young Investigator<br />

Award 2003 from the Royal Belgian <strong>Society</strong> for Rheumatology.<br />

In 2003 Cosimo moved to the UK in the Department of Rheumatology<br />

at King's College London. In May 2005 he was awarded a<br />

Clinician Scientist Fellowship from the Medical Research Council<br />

and in December 2005 he was appointed Clinical Senior Lecturer<br />

& Consultant Rheumatologist. Since September 2007 Cosimo is<br />

Professor of Translational Medicine at the University of Aberdeen,<br />

where he heads the Regenerative Medicine Group in the<br />

Musculoskeletal Research Programme. Cosimo has expertise in<br />

translational stem cell research for musculoskeletal repair, regenerative<br />

medicine and tissue engineering. His current research<br />

interests focus on the development of novel stem cell-based<br />

therapies for the musculoskeletal system, mainly articular cartilage<br />

and bone; they also include the study of the resident joint<br />

stem cells and their niches in health and diseases such as osteoarthritis<br />

and rheumatoid arthritis. Cosimo is a member of the<br />

Research & Training Committee of the Osteoarthritis Research<br />

<strong>Society</strong> <strong>International</strong> (OARSI) and of the editorial board of several<br />

journals including Regenerative Medicine and Arthritis &<br />

Rheumatism. He serves on the Scientific Advisory Panel of Action<br />

Medical Research and on the Fellowships Implementation<br />

Committee of Arthritis Research UK.<br />

Dhollander Aad, MD PT<br />

Ghent University Hospital<br />

Aad Dhollander studied physiotherapy (PT,<br />

2002) and afterwards medicine (MD, 2009) at<br />

the Ghent University. In 2009, he began his residency in Orthopaedic<br />

Surgery and Traumatology at the Ghent University Hospital<br />

(Head of the Department: Prof. Dr. Jan Victor). His clinical and<br />

research interests focus on traumatology, knee- and hip-surgery.<br />

Currently, he is doing his PhD with Professor Dr. G. Verbruggen<br />

involving basic research focusing on metabolic characteristics


Invited Faculty 2012 in alphabetical order (not complete)<br />

of different connective tissue cell types and clinical research<br />

focusing on the outcome of several cartilage repair strategies.<br />

Aad Dhollander is a member of various scientific societies, such<br />

as the ICRS since 2010, the Young Scientist and Orthopaedic<br />

Surgeons (YSOS), the Belgian <strong>Society</strong> for Orthopaedic Surgery<br />

and Traumatology (BVOT), Belgian Knee and Hip <strong>Society</strong> (BKS-<br />

BHS), Osteoarthritis Research <strong>Society</strong> <strong>International</strong> (OARSI),<br />

and vice-president of the Belgian Orthopaedics and Traumatology<br />

Residents Association (BOTRA). Together with Prof. Dr. Peter<br />

Verdonk, he hosted the ICRS travelling fellowship 2011 visiting<br />

Ghent.<br />

Farr Jack, MD<br />

<strong>Cartilage</strong> Restoration Center of Indiana,<br />

Greenwood, USA<br />

Dr. Farr received his undergraduate degree in<br />

Biological Engineering from Rose Hulman Institute of Technology<br />

in Terre Haute, Indiana in 1975, where he also was awarded<br />

an honorary doctorate of Biological Engineering. He earned his<br />

medical degree from Indiana University in 1979. He completed<br />

his Orthopaedic Surgery residency at Indiana University Medical<br />

Center in 1986. Over the past 20 years, Dr. Farr has continued to<br />

focus his practice in sports medicine and knee restoration. His<br />

numerous appointments and affiliations include a voluntary clinical<br />

Professorship in Orthopaedic Surgery at Indiana University<br />

Medical Center (Honorary Doctorate Biological Engineering) and<br />

a board position with the <strong>Cartilage</strong> Research Foundation (Board<br />

Certified Orthopaedic Surgeon). As a leader in US cartilage restoration<br />

advances, Dr. Farr participates in several ongoing articular<br />

and meniscal cartilage clinical trials. He also designed and<br />

received a patent for a Meniscal Allograft Transplant System. For<br />

patients with knee changes too far advanced for restoration, Dr.<br />

Farr worked as a design surgeon for a new partial knee replacement<br />

system. Dr. Farr is actively affiliated with the Indiana Orthopaedic<br />

Hospital. He also has courtesy affiliations with other<br />

Indianapolis area hospitals. He is a member of the American<br />

Academy of Orthopaedic Surgeons, the American Orthopaedic<br />

<strong>Society</strong> of Sports Medicine, the <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong><br />

<strong>Society</strong> and numerous other professional organizations.<br />

Ferkel Richard, MD<br />

Southern California Orthopaedic Institute,<br />

Van Nuys, CA, USA<br />

Richard Ferkel grew up in Illinois and attended<br />

UCLA where he played football for two years. Subsequently,<br />

he graduated from Northwestern University Medical School<br />

in Chicago and accomplished his orthopedic training at UCLA.<br />

He completed fellowship training in Sports Medicine and Reconstructive<br />

Knee and Shoulder Surgery at the Southern California<br />

Orthopedic Institute, and had additional training in foot<br />

and ankle surgery at that time. He is currently the Director of<br />

the Sports Medicine Fellowship Program at Southern California<br />

Orthopedic Institute. Dr. Ferkel has published numerous articles<br />

regarding the shoulder, knee, foot and ankle, and sports medicine.<br />

He co-edited the book, “Prosthetic Ligament Reconstruction<br />

of the Knee,” and authored the text book “Arthroscopic Surgery<br />

of the Foot and Ankle.” He lectures extensively all over the<br />

world. In addition, he has invented numerous surgical devices<br />

and performed live surgery throughout Europe. He has received<br />

numerous awards and was selected to be in the book entitled<br />

“The Best Doctors in America.“ Dr. Ferkel is a Clinical Instructor<br />

of Orthopedic Surgery at UCLA, and was Chief of Arthroscopic<br />

Surgery at Wadsworth Veterans Hospital for fifteen years. He is a<br />

member of Arthroscopy Association of North America, American<br />

Orthopaedic <strong>Society</strong> for Sports Medicine, <strong>International</strong> <strong>Society</strong><br />

for Arthroscopy, Knee Surgery, and Orthopedic Sports Medicine,<br />

and the American Orthopaedic Foot and Ankle <strong>Society</strong>. He is a<br />

Qualified Medical Examiner (Q.M.E.) for the State of California<br />

Industrial Medical Council, Department of Industrial Relations.<br />

Dr. Ferkel started the Athletic Training Program at Southern California<br />

Orthopedic Institute. In addition, he has been the Team<br />

Physician for Crespi, Oaks Christian, and Harvard-Westlake High<br />

Schools, and Los Angeles Valley College for many years. He has<br />

worked with the U.S. Olympic teams, U.S. Soccer Team, Special<br />

Olympics, and is a consultant to the NFL, MLB, and NBA.<br />

Flannery Carl, PhD<br />

Pfyzer <strong>–</strong> Bio Therapeutics Division,<br />

Cambridge, USA<br />

Carl Flannery received a PhD in Medical Sciences<br />

from the University of South Florida in 1995. He has conducted<br />

research and studies on cartilage and joint pathobiology<br />

and therapeutics at Rhode Island Hospital, the Shriners Hospital<br />

in Tampa, Cardiff University, and at Wyeth Research and Pfizer in<br />

Cambridge, Massachusetts.<br />

Fortier Lisa, Ass Prof., DVM, PhD<br />

Cornell University, Ithaca, NY, USA<br />

Lisa Fortier is an Associate Professor of Surgery<br />

at Cornell University in Ithaca, NY. She received<br />

her DVM from Colorado State University and completed her<br />

PhD and surgical residency training at Cornell University. She<br />

is boarded with the American College of Veterinary Surgeons<br />

and is an active equine orthopaedic surgeon at Cornell. Her laboratory<br />

studies the intracellular pathways involved in the pathogenesis<br />

of osteoarthritis, with particular emphasis on posttraumatic<br />

osteoarthritis. In addition, Lisa’s research program<br />

investigates the clinical application of stem cells and biologics<br />

such as PRP for cartilage repair and tendonosis. She has received<br />

the Jaques Lemans Award from the <strong>International</strong> <strong>Cartilage</strong><br />

<strong>Repair</strong> <strong>Society</strong>, the New Investigator Research Award from the<br />

Orthopaedic Research <strong>Society</strong>, and the Pfizer Research Award<br />

for Research Excellence from Cornell University. Lisa is the Vice<br />

President of the <strong>International</strong> Veterinary Regenerative Medicine<br />

<strong>Society</strong> and the immediate Past President of the <strong>International</strong><br />

<strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong>.<br />

31


32<br />

Invited Faculty 2012 in alphabetical order (not complete)<br />

Frisbie David, DVM, PhD Assoc Prof<br />

Equine Surgery CSU’s Orthopaedic Research<br />

Center, Fort Collins, USA<br />

David Frisbie earned an undergraduate biochemistry<br />

degree at the University of Wisconsin as well as a Doctor<br />

of Veterinary medicine. He completed a Surgical Internship at<br />

Cornell University and began his research in joint disease. Dr.<br />

Frisbie went to Colorado State University, where he completed<br />

a Surgical Residency in Large Animal Surgery and a Masters Degree<br />

in Joint Pathobiology. He then began his work on a novel<br />

way to treat joint disease using gene therapy. Dr. Frisbie is an<br />

Associate Professor of Equine Surgery CSU’s Orthopaedic Research<br />

Center. He is a partner in Equine Sports Medicine, LLC,<br />

specializing in orthopaedics and sports medicine. Diplomate of<br />

the American College of Veterinary Surgeons and a Diplomate of<br />

the American College of Veterinary Sports Medicine and Rehabilitation.<br />

Dr. Frisbie specializes in orthopaedic research, equine<br />

lameness, orthopaedic surgery and gene therapy.<br />

Fu Freddie, Prof., MD<br />

Department of Orthopaedic Surgery,<br />

University of Pittsburgh,<br />

School of Medicine, Pittsburgh, USA<br />

Freddie H. Fu is the David Silver Professor and Chairman of the<br />

Department of Orthopaedic Surgery, University of Pittsburgh<br />

School of Medicine. Dr. Fu specializes in Sports Medicine and<br />

holds secondary appointments as Professor of Physical Therapy,<br />

Health & Physical Activity, and Mechanical Engineering and<br />

serves as the Head Team Physician for the University of Pittsburgh<br />

Athletic Department. Dr. Fu graduated summa cum laude<br />

from Dartmouth College in 1974 and received his BMS in 1975<br />

from Dartmouth Medical School. He earned his medical degree<br />

in 1977 from the University of Pittsburgh and completed his residency<br />

training at Pitt in 1982. Dr. Fu’s major research interest lies<br />

in clinical outcomes as well as bioengineering of sports-related<br />

problems. His research efforts have led to over 200 professional<br />

awards and honours, 960 national and international presentations,<br />

and editorship of 29 major orthopaedic textbooks. Dr. Fu<br />

is also the author or co-author of 450 peer-reviewed articles and<br />

116 book chapters on the management of sports injuries. In July<br />

2008 he assumed the Presidency of the American Orthopaedic<br />

<strong>Society</strong> for Sports Medicine and, in April 2009, was named President<br />

of the <strong>International</strong> <strong>Society</strong> of Arthroscopy, Knee Surgery<br />

and Orthopaedic Sports Medicine<br />

Gawlitta Debby, PhD<br />

Orthopaedics, University Medical Center<br />

Utrecht, NL<br />

Her research is part of the strategic impulse of<br />

the UMC Utrecht directed at improvement of current techniques<br />

in regenerative medicine. The special focus she has within the<br />

strategic impulse is on the orthopaedic applications, involving<br />

tissue engineering of cartilage and bone using adult human<br />

stem cells. Other topics within this project are the design of a bioreactor<br />

system for stimulating optimal tissue development and<br />

angiogenesis. Her Key side activities include the supervision of<br />

undergraduate students.<br />

EDUCATION AND CAREER HISTORY: Post-doc (2007 - now),<br />

Dept. Orthopaedics, UMC Utrecht, Dept. Biomedical Engineering,<br />

MaTe, Eindhoven University of Technology, PhD in Biomedical<br />

Engineering, (2007) 'Compression-induced factors influencing<br />

the damage of engineered skeletal muscle.’, Eindhoven<br />

University of Technology , MSc in Biomedical Engineering (2002)<br />

Eindhoven University of Technology<br />

Gersoff Wayne, MD<br />

Advanced Orthopedics and Sports Medicine<br />

Specialists, Denver, USA<br />

Wayne Gersoff is an orthopedic surgeon specializing<br />

in sports medicine and cartilage restoration in Denver,<br />

Colorado. After completing his residency in Orthopedic Surgery<br />

at Yale University in 1986, he went on to complete a fellowship<br />

in Sports Medicine at the University of Wisconsin in 1987. From<br />

1987 <strong>–</strong> 1994, he was an Assistant Professor at the University of<br />

Colorado Health Sciences Center where he also served as Director<br />

of Sport Medicine and Head Team Physician for the University<br />

of Colorado. Upon leaving the university for private practice,<br />

Dr. Gersoff has maintained his active involvement in sports<br />

medicine and cartilage restoration of the knee. He continues to<br />

serve as team physician for professional, collegiate, and high<br />

school athletes. His expertise in articular cartilage restoration<br />

and meniscal transplant has allowed him to lecture and write on<br />

these areas. He is on the general board of the ICRS and is president<br />

of the Major League Soccer physicians’ group.<br />

Getgood Alan, Ass Prof., MD<br />

University Hospital Coventry and Warwickshire<br />

NHS Trust, Coventry, UK<br />

Alan is Associate Professor of Trauma and Orthopaedic<br />

Surgery at the University of Warwick, UK. His clinical<br />

interests include sports injuries of the knee and shoulder, and<br />

the treatment of young arthritics, with a particular focus on osteotomy,<br />

articular cartilage repair and meniscal reconstruction.<br />

His research includes both a clinical and basic science focus,<br />

with an emphasis towards combination biological products,<br />

which can potentially enhance tissue regeneration. Following<br />

graduating from the University of Edinburgh in 2000, Alan moved<br />

to Cambridge to complete his orthopaedic training. In 2009<br />

he completed his Doctor of Medicine thesis entitled ‘Articular<br />

<strong>Cartilage</strong> Tissue Engineering’ from the University of Cambridge.<br />

A one-year fellowship in orthopaedic sports medicine followed<br />

at both the Fowler Kennedy Sport Medicine Clinic (London,<br />

Ontario, <strong>Canada</strong>) and Banff Sport Medicine (Alberta, <strong>Canada</strong>),<br />

where he was involved in the medical care for Alpine <strong>Canada</strong><br />

ski team. On returning to the UK, he then completed a further<br />

knee reconstruction fellowship in Coventry. In September this<br />

year he will return to the Fowler Kennedy Clinic to take up a faculty<br />

position at the University of Western Ontario and continue<br />

his clinical and research interests. When away from work Alan<br />

enjoys skiing, climbing and running marathons, destroying his<br />

own knees.


Invited Faculty 2012 in alphabetical order (not complete)<br />

Giza Eric, Ass Prof., MD<br />

Foot and Ankle Service at UC Davis Department<br />

of Orthopaedics in Sacramento, CA, USA<br />

Eric Giza is an Associate Professor and Chief of<br />

the Foot and Ankle Service at UC Davis Department of Orthopaedics<br />

in Sacramento, CA. Dr. Giza did his undergraduate work at<br />

Haverford College in Philadelphia and received his MD degree<br />

from Temple University, also in Philadelphia. He completed his<br />

residency at Harvard University in Boston. Following residency,<br />

he completed a Foot and Ankle Surgical Fellowship at the Foot<br />

and Ankle Clinic in Sydney, Australia and a Sports Medicine Fellowship<br />

at the Santa Monica Orthopaedic Group. Being a former<br />

collegiate soccer player, Dr. Giza maintains his interest in sports<br />

by caring for athletes on both Professional and amateur soccer<br />

teams. He plays an active role on the Major League Soccer medical<br />

staff and currently serves as an assistant team physician for<br />

the US Soccer Federation.<br />

Gomoll Andreas, Ass Prof., MD<br />

Brigham and Women’s Hospital<br />

Boston, USA<br />

Born and raised in Germany, Dr. Gomoll attended<br />

Ludwig-Maximilians-Medical School in Munich prior to<br />

spending 2 years at Brigham and Women’s Hospital as a research<br />

fellow. He then completed his residency training at the<br />

Harvard Combined Orthopaedic Residency Program in Boston,<br />

MA, and a Sports Medicine Fellowship at Rush University in Chicago,<br />

IL. After his return to Boston, he joined Dr. Tom Minas at<br />

the <strong>Cartilage</strong> <strong>Repair</strong> Center at Brigham and Women’s Hospital,<br />

where he specializes in biologic knee reconstruction, such as<br />

cartilage repair, meniscal transplantation and osteotomy. Dr.<br />

Gomoll has an academic appointment as Assistant Professor<br />

of Orthopaedic Surgery at Harvard Medical School. His main research<br />

interests are clinical outcome studies of existing, as well<br />

as the investigation of new cartilage repair procedures.<br />

Goodrich Laurie, DVM, PhD Assoc Prof.<br />

Equine Surgery, Colorado State University, USA<br />

Laurie completed her DVM in 1991, an internship<br />

in equine surgery in 1992 and surgical residency<br />

in 1996. She became board certified in surgery thereafter.<br />

She was a faculty surgeon at Cornell College of Veterinary Medicine<br />

from 1996 through 2001 and then went on to complete a<br />

PhD in cellular and molecular therapy related to cartilage repair.<br />

She became an assistant Professor in equine surgery at Colorado<br />

State University as well as a member of the research team at<br />

the Equine Orthopedic Research Center at CSU. Her research is<br />

NIH funded and her areas of focus include gene therapy for cartilage<br />

and bone repair, and regenerative therapies as they relate<br />

to musculoskeletal healing in the equine model. She is now an<br />

associate Professor in the department of clinical sciences and<br />

divides her time between being a surgeon in the equine hospital<br />

at CSU and a translational scientist at the Orthopedic Research<br />

Center. She is a member of the ICRS, the Orthopedic Research<br />

<strong>Society</strong> and the American <strong>Society</strong> of Gene and Cell Therapy.<br />

Grande Daniel, Ass Prof., PhD<br />

Feinstein Institute for Medical Research<br />

Manhasset, NY, USA<br />

Daniel Grande is associate investigator and<br />

director of orthopaedic research at the Feinstein Institute for<br />

Medical research. He is also associate Professor at the newly<br />

accredited Hofstra School of Medicine. He completed his PhD<br />

at New York University and his post-doctoral fellowship ion biomechanics<br />

at the Hospital for Special Surgery. He has worked<br />

extensively in the area of regenerative medicine and tissue engineering.<br />

His early work developed the first use of cell based<br />

therapy for cartilage repair, currently known as autologus chondrocyte<br />

transplantation. He has served on committees with the<br />

Orthopaedic Research <strong>Society</strong> as spine topic chair and the basic<br />

science committee. Dr, Grande is significantly involved in mentoring<br />

and teaching of orthopaedic residents for his department.<br />

He has been a reviewer for a number of journals including: Journal<br />

of Orthopaedic Research, Clinical Orthopaedics, Osteoarthritis<br />

and <strong>Cartilage</strong>, American Journal of Sports Medicine, Nature<br />

Reviews Rheumatology and Applied Biomaterials. He has<br />

been awarded eight patents and helped found two companies in<br />

the orthopaedic surgery field of use. He has served as a member<br />

of several companies’ scientific advisory boards. He completed<br />

a five year rotation with OREF to assist in grant reviews. He also<br />

regularly serves on NIH study sections for RO1, R21, and SBIR/<br />

STTR grants specific to musculoskeletal applications.<br />

Hambly Karen, MS, PhD<br />

Senior Lecturer/PhD Research Student,<br />

Centre for Sports Studies -University of Kent, UK<br />

Karen Hambly qualified as a physiotherapist in<br />

1998 from the University of Southampton having already completed<br />

a degree in sports science. Prior to moving into academia<br />

Karen worked as the sports medicine coordinator for both<br />

British Cycling and UK Sport and has worked with Olympic and<br />

Paralympic teams and elite athletes in several sports. Karen's<br />

research interests are cantered around rehabilitation for articular<br />

cartilage repair and in 2011 she completed her PhD on the patient<br />

perspective of outcome measures after articular cartilage<br />

repair of the knee. Karen has published and presented at internationally<br />

and is an editorial reviewer for eleven peer-reviewed<br />

journals. Karen is currently the Co-Chair of the ICRS Sports Injury<br />

and Rehabilitation workgroup.<br />

Hoemann Caroline, Ass. Prof., PhD<br />

Dept. Chemical Engineering,<br />

École Polytechnique <strong>Montreal</strong>, <strong>Canada</strong><br />

Caroline. Hoemann (PhD, MIT, 1992) is an associate<br />

Professor of Chemical Engineering and Biomedical Engineering,<br />

an FRSQ National Research fellow, and has over 45<br />

publications and 6 patents. She has been a member of the ICRS<br />

since 2002, was lead author on the ICRS recommendation paper,<br />

“<strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong> (ICRS) recommended<br />

guidelines for histological endpoints for cartilage repair studies<br />

in animal models and clinical trials <strong>Cartilage</strong> 2011, 2:153-172”<br />

33


34<br />

Invited Faculty 2012 in alphabetical order (not complete)<br />

and serves on the editorial board of <strong>Cartilage</strong>, and The Open Orthopaedics<br />

Journal. She spent 5 years as Director of <strong>Cartilage</strong><br />

<strong>Repair</strong>, in a <strong>Montreal</strong>-based biomedical device company, where<br />

she co-invented and co-developed a novel medical device for<br />

articular cartilage repair, BST-CarGel ® that recently completed<br />

an 80-patient randomized controlled clinical trial. Her current<br />

research program has created bioengineered blood clots, developed<br />

new methods for staining and analyzing human cartilage<br />

repair biopsies, and has made advances in understanding the<br />

role of therapeutic inflammation and subchondral bone remodelling<br />

in cartilage repair responses. Her translational research<br />

program aims to understand the mechanisms of cartilage repair,<br />

in order to bring new treatment options to patients with arthritis<br />

Hollander Anthony, Prof., PhD<br />

School of Medical Sciences University Walk<br />

Clifton Bristol, UK<br />

Anthony Hollander is the Arthritis Research UK<br />

Professor of Rheumatology and Tissue Engineering at the University<br />

of Bristol and Head of The School of Cellular and Molecular<br />

Medicine. He has many years experience in cartilage biology<br />

and his research is particularly focused on osteoarthritis.<br />

He also has a more general expertise in the wider fields of stem<br />

cells and tissue engineering. In 2010 the “Times” newspaper<br />

ranking of Britain’s 100 most important scientists included him<br />

at 39th on the list. Professor Anthony Hollander has been working<br />

in the field of cartilage biology and arthritis research for two<br />

decades. Three of those years were spent at the internationally<br />

recognised cartilage laboratory at McGill University in <strong>Montreal</strong>.<br />

More recently he has focused on tissue engineering and stem<br />

cell biology for cartilage repair. Professor Hollander has received<br />

funding in excess of £5 million of peer-reviewed funding over the<br />

past 10 years from The UK government, medical charities, the<br />

EU framework programmes and from biotechnology companies.<br />

He has been the named inventor on several patents. He is cofounder<br />

and Scientific Director of a University of Bristol spin-out<br />

company, Azellon Cell Therapeutics. His work includes a study<br />

on the regulation of stem cell differentiation for cartilage repair<br />

and has pioneered the development of new assays and methodological<br />

approaches for the measurement of repair tissue quality<br />

in very small biopsies of cartilage from patients with knee<br />

injuries. In 2008, Professor Hollander and a team of scientists<br />

and surgeons successfully created and then transplanted the<br />

first tissue-engineered trachea (windpipe), using a patient's own<br />

stem cells. The bioengineered trachea immediately provided the<br />

patient with a normally functioning airway, thereby saving her<br />

life. Professor is Hollander vice president/president-elect of The<br />

<strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong>. He is Associate Editor of a<br />

leading journal, Osteoarthritis and <strong>Cartilage</strong>. He is an editorial<br />

board member for several other journals.<br />

Hunziker Ernst, Prof., MD<br />

ITI Research Institute, University of Bern,<br />

Switzerland<br />

After completing his training as a medical doctor,<br />

Ernst B. Hunziker launched on a research career in the field<br />

of growth-plate cartilage, which focused on its development,<br />

structure and physiological activity. After his appointment as<br />

Director of the M.E.Müller-Institute of Biomechanics (University<br />

of Bern), his research activities were geared more towards articular<br />

cartilage: its structure, microbiomechanics and repair.<br />

Later, as Director of the ITI Research Institute of Dental and Skeletal<br />

Biology, his interest in the engineering (repair) of articular<br />

cartilage continued. However, he also became involved in the<br />

field of Implantology. Now, as Director of the Center of Regenerative<br />

Medicine for Skeletal Tissues (University of Bern), Ernst<br />

Hunziker`s research activities continue in the same directions.<br />

He was ICRS President from 2004 <strong>–</strong> 2006.<br />

Hurtig Mark, Prof., DVM<br />

Canadian Arthritis Network, Clinic Studies,<br />

University of Guelph, <strong>Canada</strong><br />

Mark Hurtig is the director of the Strategic Research<br />

Resource Laboratory for the Canadian Arthritis Network<br />

at the University of Guelph in <strong>Canada</strong>. He received a DVM at Guelph<br />

in 1978, an MVSc from the University of Saskatchewan in<br />

1983, and completed American College of Veterinary Surgeon<br />

Diplomate status in 1989. While working 15 years as a staff surgeon<br />

for the Ontario Veterinary College at the University of Guelph<br />

he developed a career interest in joint injuries and sports<br />

medicine that has led to collaborations in cell & molecular biology,<br />

biomechanics, engineering, biochemistry, and biophysics.<br />

He has been an ICRS member since 1998 and served as an ICRS<br />

board member, chairing the animal models subcommittee during<br />

2005<strong>–</strong>2008. His working group on risk factors that predict<br />

osteoarthritis after knee injury includes faculty and clinicianscientists<br />

from hospitals across <strong>Canada</strong> with collaborations in<br />

many other countries. His laboratory encourages graduate students<br />

from many other disciplines who wish to focus their career<br />

on articular injuries and tissue repair.<br />

Hutmacher Dietmar, Prof.<br />

Professor and Chair of Regenerative Medicine<br />

at the Institute of Health and Biomedical<br />

Innovation of QUT, Kelvin Grove, AUS<br />

Dietmar W Hutmacher is the Professor and Chair of Regenerative<br />

Medicine at the Institute of Health and Biomedical Innovation of<br />

QUT, where he leads the Regenerative Medicine Group, a multidisciplinary<br />

team of researchers including engineers, cell biologists,<br />

polymer chemists, clinicians, and veterinary surgeons.<br />

Professor Hutmacher is a multidisciplinary biomedical engineer,<br />

an educator, an inventor, and a creator of new intellectual property<br />

opportunities. As a reflection of his pioneering ethos, his<br />

recent research efforts have resulted in traditional scientific/<br />

academic outputs as well as pivotal commercialisation outco-


Invited Faculty 2012 in alphabetical order (not complete)<br />

mes. He is one of very few academics in the field of biomaterials/tissue<br />

engineering who have taken a research programme<br />

from the holistic concept through to clinical application. Professor<br />

Hutmacher’s pre-eminent international standing and impact<br />

on the field are illustrated by his publication record (more than<br />

200 journal articles, edited 3 books, 35 book chapters and some<br />

350 conference papers) and citation record (more than 6200 citations,<br />

h-index of 39). Three of his papers in Materials Science<br />

have received citations in the top 1% for the field, and he is also<br />

ranked by Thomas Reuters 45th world-wide in citations per paper<br />

(54 per paper) in Materials Science over the past decade.<br />

Over the past 10 years in academia he has been lead CI, Co-CI<br />

or collaborator in grants totalling more than AUD$ 35 million.<br />

During the most recent 4 years in Australia, he has been an investigator<br />

on external grants totalling in excess of AUD$ 8 million.<br />

These grants have included ARC Discovery, ARC Linkage, ARC<br />

LIEF NHMRC Projects, NIH, and Prostate Cancer Foundation of<br />

Australia awards. He holds Adjunct Professorships at prestigious<br />

universities e.g. in the USA (Georgia Institute of Technology).<br />

His team is endeavouring to meet the challenge to provide<br />

new bone to replace or restore the function of traumatised bone<br />

or bone lost as a consequence of age or disease. Bone tissue<br />

engineering concepts developed by his group promise to deliver<br />

specifiable replacement tissues and the prospect of efficacious<br />

alternative therapies for orthopaedic applications such as nonunion<br />

fractures, healing of critical-sized segmental defects and<br />

regeneration. Another key project for Professor Hutmacher’s<br />

group has been to develop an animal model for bone repair research.<br />

His group has established and fully characterised a critical-sized<br />

tibial defect model in sheep tibiae to evaluate different<br />

tissue engineering-based treatment concepts. Finally, Professor<br />

Hutmacher’s group is using their expertise in biomaterials & tissue<br />

engineering to tackle a different problem: development of<br />

3D cancer models in the lab from scratch. In comparison to the<br />

2D models currently used, the complex 3D culture model being<br />

developed by Professor Hutmacher’s group is much closer to the<br />

in vivo conditions which will allow researchers testing for example,<br />

drug efficacy, to achieve much more realistic results.<br />

Jakob Roland, Prof., MD<br />

Professor Emeritus of University of Berne<br />

Former Chairman of Orthopaedic Surgery of<br />

Kantonsspital, Fribourg (1995<strong>–</strong>2007), Founding<br />

President of ICRS 1997-98, Past President of Swiss Orthopaedic<br />

<strong>Society</strong> (1994-96), ISAKOS (1999<strong>–</strong>2001), AO Switzerland (2002-<br />

2009), Passed Activity as Member in Editorial Boards of Scientific<br />

Journals: Journal of Bone and Joint Surgery, British Volume,<br />

Journal of Knee Surgery, Sports Traumatology and Arthroscopy,<br />

KSSTA (Board of Trustees), La chirurgia degli organi di movimento,<br />

Author and Co-author of 200 scientific articles and 4 text<br />

books: “The Knee and the Crucial Ligaments”, R.P. Jakob and<br />

H.U. Stäubli, Springer, 1991, “Planning and Reduction Technique<br />

in Fracture Surgery”, J. Mast, R.P. Jakob, R. Ganz, Springer, 1989,<br />

“European Instructional Course Lectures”, R.P. Jakob, P. Fulford,<br />

F. Horan, The British Editorial <strong>Society</strong> of Bone and Joint Surgery,<br />

1999, Osteotomies around the Knee, Ph. Lobenhoffer, R. Heer-<br />

warden, A. Staubli, R.P. Jakob (Thieme, AO Publication, 2008),<br />

Honours: Corresponding member Austrian AO-Chapter (2000),<br />

Godfather Herodicus <strong>Society</strong> USA, 2000, Surgeon in Chief pro<br />

temp. The Hospital For Special Surgery New York, 2002, Honorary<br />

Member of ANA (American <strong>Society</strong> of Arthroscopy, 2004), Honorary<br />

Member of ISAKOS (<strong>International</strong> <strong>Society</strong> of Arthroscopy,<br />

Knee Surgery and Orthopaedic Sports Medicine), 2007, Honorary<br />

Fellow of ICRS (<strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong>), 2007,<br />

Honorary Member of Swiss <strong>Society</strong> of Accident Surgery and Insurance<br />

Medicine, 2008, Honorary Member of Swiss Orthopaedic<br />

<strong>Society</strong>, 2009<br />

Johnstone Brian, Prof., PhD<br />

Oregon Health & Science University<br />

Portland, USA<br />

Brian Johnstone did his predoctoral research at<br />

the Kennedy Institute of Rheumatology, London, England and<br />

postdoctoral work at West Virginia University and the University<br />

of North Carolina at Chapel Hill, USA. His work on intervertebral<br />

disc biology was acknowledged with two Volvo prizes for spine<br />

research. He moved to Case Western Reserve University in 1993<br />

and developed the in vitro system for the chondrogenic induction<br />

of adult stem cells. In 2004, he became Director of Research<br />

in the Department of Orthopaedics at Oregon Health and Science<br />

University, Portland, Oregon where he continues his work<br />

on adult stem cells in skeletal tissue repair and regeneration.<br />

He is a scientific editor for the open access journal eCells and<br />

Materials, and was the President of the Orthopaedic Research<br />

<strong>Society</strong> for 2011<strong>–</strong>2012.<br />

Jurvelin Jukka, MSc, PhD<br />

Dept of medical physics, University of Kuopio,<br />

Finland<br />

Jukka Jurvelin graduated from the Department<br />

of Physics, University of Kuopio 1981. He completed a specialization<br />

in hospital physics in 1991 and received Ph.D degree in<br />

1993. He was a post doc researcher 1993<strong>–</strong>1995 in Mueller Institute<br />

for Biomechanics, Bern, Switzerland. He worked 1995<strong>–</strong>2003<br />

as Physicist and 2004<strong>–</strong>2008 as Chief Physicist, Department of<br />

Clinical Physiology and Nuclear Medicine, Kuopio University<br />

Hospital and as Vice-Dean, Faculty of Natural and Environmental<br />

Sciences, University of Kuopio, Finland. During 2008-9 he was<br />

the Vice-Rector for research in University of Kuopio. Currently he<br />

is a Professor of Medical Physics at the Department of Applied<br />

Physics, University of Eastern Finland. Jukka Jurvelin is the head<br />

of the Biophysics of Bone and <strong>Cartilage</strong> (BBC) research group<br />

(www.luotain.uku.fi). Jukka Jurvelin acts in the editorial board of<br />

Sage journal CARTILAGE. The current research interests include<br />

development of quantitative biomechanical, ultrasound, MRI<br />

and x-ray methods for sensitive diagnostics of osteoporosis and<br />

osteoarthrosis, and monitoring of cartilage repair.<br />

35


36<br />

Invited Faculty 2012 in alphabetical order (not complete)<br />

Kandel Rita, MD<br />

Mount Sinai Hospital<br />

Toronto, <strong>Canada</strong><br />

Rita Kandel is a clinician-scientist and Chief of<br />

Pathology and Laboratory Medicine at Mt. Sinai Hospital, Toronto,<br />

<strong>Canada</strong>. She is a Professor in the Department of Laboratory<br />

Medicine and Pathobiology, cross-appointed to the Dept<br />

of Surgery and a member of the Institute of Biomaterials and<br />

Biomedical Engineering at the University of Toronto. She is the<br />

Director of the Bioengineering of Skeletal Tissues Team, which<br />

consists of a multidisciplinary group of investigators, including<br />

engineers, biologists, stem cell biologists, and clinicians whose<br />

work focuses on regenerative medicine. Her research interest is<br />

in the bioengineering of tissues for articular cartilage repair and<br />

intervertebral disc replacement. She is a long standing member<br />

of the ORS. She is currently an Associate Editor of <strong>Cartilage</strong> and<br />

a member of the Editorial Board of Osteoarthritis and <strong>Cartilage</strong>.<br />

She has published over 170 papers.<br />

Kisiday John, Assoc Prof., PhD<br />

Colorado State University<br />

Fort Collins, USA<br />

John Kisiday is a member in the Equine Orthopaedic<br />

Research Center (EORC) and the Department of Clinical<br />

Sciences at Colorado State University. He received his Ph.D. in<br />

Bioengineering from the Massachusetts Institute of Technology<br />

in 2003, and joined the faculty at Colorado State University in<br />

2005. His research activities have included the characterization<br />

of novel scaffolds for cartilage tissue engineering, the effects<br />

of dynamic compression on chondrocyte biosynthesis and mesenchymal<br />

stem cell differentiation, and techniques for inducing<br />

mesenchymal stem cell chondrogenesis geared towards clinical<br />

translation. He has worked closely with EORC faculty investigating<br />

the use of autologous mesenchymal stem cells in clinical<br />

cases of equine tendonitis and joint disease. He has served as<br />

an organizer for the tissue engineering topic for the annual meeting<br />

of the Orthopaedic Research <strong>Society</strong> since 2010.<br />

Klein Travis, PhD<br />

<strong>Cartilage</strong> Regeneration Laboratory (CRL),<br />

the Institute of Health and Biomedical<br />

Innovation, Queensland University of<br />

Technology, Brisbane, Australia<br />

Travis Klein heads the <strong>Cartilage</strong> Regeneration Laboratory (CRL)<br />

at the Institute of Health and Biomedical Innovation, Queensland<br />

University of Technology, in Brisbane, Australia. Travis<br />

received his PhD in Bioengineering from the University of California,<br />

San Diego in 2005. He is now a Future Fellow of the<br />

Australian Research Council. The ultimate goal of the CRL is<br />

to help develop long-term regenerative therapies for treating<br />

cartilage defects, including osteoarthritis. To help understand<br />

chondrogenesis and joint pathologies, the group is developing:<br />

model systems using human cells (chondrocytes and progenitor<br />

cells); functionalised biomaterials; biofabrication technologies;<br />

and mechanical stimulation techniques. In addition, the CRL<br />

works towards advancing imaging technologies for monitoring<br />

changes in native and tissue-engineered cartilage. One special<br />

area of interest has been in understanding the cellular and extracellular<br />

matrix differences between different depth zones,<br />

and recapitulating these in vitro.<br />

Kon Elizaveta, MD<br />

Department of Orthopaedic Surgery and<br />

Biomechanics Laboratory of Rizzoli<br />

Orthopaedic Institute, Italy<br />

Elizaveta Kon was born in Moscow. Degree in Medicine in 1994 and<br />

specialization in “Orthopaedics and Traumatology” at the University<br />

of Bologna 1999. Since 1993 carry out surgical, clinical, and research<br />

activities at the Rizzoli Orthopaedic Institute in Bologna, Italy. Staff<br />

member of the III Clinic Orthopaedic and Traumatology and Biomechanics<br />

Laboratory (leaded by Prof. Maurilio Marcacci), orthopaedic<br />

surgeon and researcher focusing on clinical and basic research in<br />

the musculoskeletal tissue engineering. Assistant Professor of Motor<br />

Sciences Faculty of the University of Bologna, held the course<br />

of “Bioengineering applying to the locomotor apparatus pathology”<br />

(2000-2005). Since 2010 Director of Nano-Biotechnology Laboratory<br />

and carry on personally numerous research projects and clinical trials<br />

regarding biotechnology applications in orthopaedics (from preclinical<br />

studies to clinical trials). Principal investigator of numerous<br />

research projects funded by the Italian Government and European<br />

Community. President of <strong>Cartilage</strong> Committee of European <strong>Society</strong><br />

of Sports Traumatology Knee Surgery and Arthroscopy (ESSKA).<br />

Chair of the ICRS Fellowship Scholarships and Grants Committee<br />

and member of the ICRS General Board. Co- founder and past president<br />

of Young Surgeons and Orthopaedic Surgeons (YSOS) club of<br />

<strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong> (ICRS). President of the <strong>Cartilage</strong><br />

Committee and Board Member of the Italian <strong>Society</strong> of Knee,<br />

Arthroscopy Sport <strong>Cartilage</strong> and Orthopaedic Technologies (SIGA-<br />

SCOT). Elizaveta Kon has authored over 80 peer-reviewed scientific<br />

articles and over 20 chapters in text books in orthopaedic surgery.<br />

She has presented at over 200 society meetings all over Europe, Asia<br />

and North America. Awards: ICRS Travelling Fellowship in 2004 and<br />

ESSKA <strong>–</strong> AOSSM travelling fellowship in 2009. She is a member of<br />

the Editorial Board of the BMC Musculoskeletal Disorders Journal<br />

and European Journal of Sports Traumatology, reviewer for many<br />

Orthopaedic journals including American Journal of Sports Medicine<br />

(golden reviewer), <strong>Cartilage</strong>, Journal of Arthroscopy, Clinical Orthopaedics<br />

and Related Research, Knee Surgery Sports Traumatology<br />

Arthroscopy Journal, Journal of Tissue Engineering, Biomaterials etc.<br />

Lafeber Floris P.J.G., Prof., PhD<br />

University Medical Centre Utrecht<br />

Netherlands<br />

Professor of Experimental Rheumatology Floris<br />

Lafeber is manager of research of the department of Rheumatology<br />

& Clinical Immunology, at the University Medical Centre Utrecht,<br />

Utrecht the Netherlands. In 1984 he obtained his degree as<br />

Master of Science in Biology and received 4 years later in 1988 his<br />

Ph.D. at the Faculty of Science of the University of Nijmegen, the<br />

Netherlands. In Utrecht he started a new research group at the department<br />

of Rheumatology, originally in the field of cartilage dege-


Invited Faculty 2012 in alphabetical order (not complete)<br />

neration and regeneration, extended with research in immunology<br />

and treatment strategies in rheumatic diseases in general. In 1999<br />

he was installed as full Professor. Floris Lafeber has more than 200<br />

peer reviewed publication half of them in the field of osteoarthritis.<br />

He is member of ICRS, ORS, and OARSI. In addition to organization,<br />

initiation, and facilitation of research in his department,<br />

there is a personal interest in osteoarthritis, more specifically in a<br />

unique treatment of severe osteoarthritis (joint distraction) which<br />

has been developed over the past years, in a unique and internationally<br />

accepted animal model of osteoarthritis (the canine Groove<br />

model) that has been developed and validated, and more recently<br />

in imaging and biochemical markers of joint tissue damage. Topics<br />

which have received great international interest from scientists<br />

and companies (involved in treatment of osteoarthritis).<br />

Lavigne Patrick, MD, PhD<br />

University of <strong>Montreal</strong><br />

<strong>Montreal</strong>, <strong>Canada</strong><br />

Dr Patrick Lavigne received a degree in Biochemistry<br />

from University of Sherbrooke in 1995.<br />

He then completed his medical degree in 1999 and orthopedic<br />

residency training in 2005 at University of <strong>Montreal</strong>. During is<br />

orthopedic training, Dr Lavigne earned a PhD degree in Biomedical<br />

Sciences. He next completed an Arthroscopy and Sports<br />

Medicine fellowship in Australia under the supervision of Mr Ian<br />

Henderson and Mr David Wood. Since 2006, Dr Lavigne is an<br />

Associate Professor of orthopedics at University of <strong>Montreal</strong>. He<br />

specializes in shoulder, knee and ankle surgery with an emphasis<br />

on osteoarthritis treatment in young patients. Dr Lavigne has<br />

both basic science and clinical expertise in cartilage repair and<br />

osteoarthritis research. He is a funded clinician-scientist from<br />

the FRSQ and is actively involved in the development of diagnostic<br />

tools for early stage OA detection and dynamic diagnostic<br />

instruments for knee pathology. He is currently supervising the<br />

clinical introduction of cartilage cell therapy in Quebec.<br />

Little Chris, Ass Prof., PhD, BVSc (DVM equivalent)<br />

Royal North Shore Hospital, St. Leonards,<br />

Australia<br />

Christopher Little is director of the Raymond<br />

Purves Bone and Joint Research Labs at the Kolling Institute<br />

of Medical Research at Royal North Shore Hospital, Australia.<br />

Chris is a qualified Veterinarian with specialist surgery training<br />

and ACVS certification. He was awarded a PhD degree from the<br />

University of Sydney in 1996 for his studies of cartilage degradation<br />

in a sheep model of osteoarthritis (OA). Following a 5-year<br />

postdoctoral position at Cardiff University (UK), he was awarded<br />

an Arthritis Foundation of Australia Fellowship at the University<br />

of Melbourne. In 2004 he moved to his current position in<br />

the University of Sydney Faculty of Medicine. Chris’s research<br />

interests centre on the biochemical and molecular mechanisms<br />

of joint pathology in OA, and tendon and intervertebral disc degeneration.<br />

Chris has extensive experience in developing and<br />

using small and large animal models of bone and joint disease.<br />

He has authored/co-authored 95 scientific papers and 7 book<br />

chapters. Dr Little is an Associate Editor of Osteoarthritis and<br />

<strong>Cartilage</strong>, served on the OARSI histopathology initiative panel to<br />

establish standardised methods for evaluation of animal models<br />

of OA. He received the 2010 Barry Preston Award from the Matrix<br />

Biology <strong>Society</strong> of Australia and New Zealand, presented to an<br />

outstanding leader in the field.<br />

Lohmander Stefan, Prof., MD, PhD<br />

Lund University, Sweden<br />

Stefan Lohmander, is Professor of Orthopaedic<br />

Surgery at Lund University, Sweden. He serves<br />

on the editorial board of several international journals, and is<br />

the editor-in-chief of ‘Osteoarthritis and <strong>Cartilage</strong>’. He has published<br />

more than 250 scientific papers, having received more<br />

than 9000 citations with an H-factor of 54.<br />

Stefan Lohmander is a past president of the Osteoarthritis Research<br />

<strong>Society</strong> <strong>International</strong>, OARSI. In 1994 he received the<br />

OARSI Award for Clinical Osteoarthritis Research, in 2004 the<br />

Orthopaedic Research <strong>Society</strong> USA (ORS) Arthur Steindler Award<br />

for significant international contributions to the understanding of<br />

musculoskeletal disease and injury. In 2006 he received the Marshall<br />

Schiff Lecture Award from the American College of Rheumatology<br />

(ACR): “A special lectureship established to address the<br />

interface between rheumatology and orthopaedics in the area of<br />

musculoskeletal medicine”, and in 2007 the Bone and Joint Decade<br />

2000-2010 Award for Research in Osteoarthritis.<br />

He is the PI of the multidisciplinary Lund University Osteoarthritis<br />

Research Group, focusing on basic and clinical aspects of<br />

osteoarthritis:<br />

• Risk factors and disease mechanisms of OA on gene, molecule<br />

and patient level<br />

• Monitoring OA Outcome by Biomarkers, Imaging and Patient-<br />

reported Outcomes<br />

• Evidence-based treatment of OA <strong>–</strong> Examining Old and New<br />

Interventions for OA in Clinical Trials<br />

Lu Helen, Ass Prof<br />

Columbia University, Biomedical Engineering,<br />

New York, NY, USA<br />

Maden Malcolm, Prof., MD, PhD<br />

Department of Biology at the University of<br />

Florida in Gainesville, Florida, USA<br />

Malcolm Maden is a member of the Department<br />

of Biology at the University of Florida in Gainesville, Florida, USA<br />

and a Joint Professor in the Department of Molecular Geentics<br />

and Microbiology. He received his PhD in Genetics from the University<br />

of Birmingham in 1975 on the subject of Urodele limb<br />

regeneration. He did a post-doctoral fellowship at the University<br />

of Sussex and then was a scientist at the National Institute for<br />

37


38<br />

Invited Faculty 2012 in alphabetical order (not complete)<br />

Medical Research, London for 11 years where his studies centred<br />

on the role of retinoic acid in development and regeneration of<br />

several different vertebrate systems including limb regeneration,<br />

the developing hindbrain and the developing limb. In 1998<br />

he moved to King’s College London to teach Anatomy and continue<br />

his research on retinoic acid where this work became more<br />

translational and concerned the potential roles for retinoic acid<br />

in the treatment of neurological and pulmonary diseases. In<br />

2008 he moved to the University of Florida and returned to study<br />

the regeneration of various systems in Urodeles and mammals.<br />

His work is currently centred on the role of retinoic acid in limb<br />

regeneration, regeneration of the limb and nervous system in<br />

Urodeles, evolutionary aspects of regeneration in Vertebrates,<br />

the relationship between development and regeneration and<br />

possibilities for the induction of regeneration or its appearance<br />

in mammals. He has published over 170 scientific papers on these<br />

subjects, is an associate editor of the <strong>International</strong> Journal of<br />

Developmental Biology and a member of Faculty of 1000.<br />

Madry Henning, Prof., MD<br />

Zentrum für Experimentelle Orthopaedie,<br />

Saarland University, Homburg, Germany<br />

Henning Madry studied Medicine at the Charité<br />

Medical School in Berlin, Germany, supported by the German<br />

Merit Foundation from 1990 <strong>–</strong> 96. Electives at the Universities of<br />

Southampton, Jerusalem, Geneva, Nice and the Baylor College<br />

of Medicine, Houston, USA. M.D. thesis “summa cum laude” at<br />

the Max-Delbrück-Center for Molecular Medicine, Berlin. Resident<br />

at the Department of Trauma and Reconstructive Surgery,<br />

Virchow Hospital, Charité, Berlin. 1996 <strong>–</strong> 98. Postdoctoral Fellow<br />

(supported by the German Academy of Scientists Leopoldina)<br />

from 1998 <strong>–</strong> 2000 at the Massachusetts General Hospital, Harvard<br />

Medical School, Boston and at the Massachusetts Institute<br />

of Technology, Cambridge, USA. In 2000, he founded the<br />

Laboratory for Experimental Orthopaedics at the Department of<br />

Orthopaedic Surgery, Saarland University, Homburg, Germany.<br />

In 2004 board-certified in Orthopaedic Surgery and cumulative<br />

“Habilitation” on the subject: “Therapeutic Gene Transfer in<br />

Chondrocytes”. Since 10/06 Attending Clinician. From 2008 <strong>–</strong><br />

2010 Chairman of the Articular <strong>Cartilage</strong> Committee of ESSKA,<br />

since 2010 Vice-Secretary General of ESSKA. He has received international<br />

and national awards, among which the Heine-Award<br />

of the DGOOC, the AO Research Fund Prize and the New Investigator<br />

Recognition Award of the Orthopaedic Research <strong>Society</strong>.<br />

He currently holds the Endowed Chair of Experimental Orthopaedics<br />

and Osteoarthritis Research at the Saarland University. His<br />

basic scientific interests are the subchondral bone in articular<br />

cartilage repair, regeneration of cartilage defects using genebased<br />

approaches and osteoarthritis. His clinical interests are<br />

reconstructive therapies for articular cartilage defects.<br />

Malda Jos, Ass Prof., MSc, PhD<br />

Orthopaedics, University Medical Center<br />

Utrecht, NL<br />

Jos Malda is an Assistant Professor at the Department<br />

of Orthopaedics at the University Medical Center Utrecht<br />

(The Netherlands) and an adjunct Associate Professor at<br />

the Queensland University of Technology (QUT, Brisbane, Australia).<br />

He received his MSc degree in Bioprocess Engineering<br />

from Wageningen University in 1999 and completed his PhD on<br />

<strong>Cartilage</strong> Tissue Engineering in 2003 (University of Twente). He<br />

subsequently accepted a research fellowship at the Institute of<br />

Health and Biomedical Innovation, (QUT, Brisbane, Australia),<br />

where he still holds an adjunct position. In 2007, he was awarded<br />

a prestigious Veni Grant by STW/NWO for the engineering<br />

of articular cartilage with biomimetic zones using bioprinting<br />

technologies. Since 2010, he is an Assistant Professor at the<br />

Department of Orthopaedics and his research has a particular<br />

focus on implants for regeneration of osteochondral defects.<br />

He has published over 55 peer-reviewed publications and book<br />

chapters and has been actively involved in the organisation of<br />

international conferences in the field of regenerative medicine.<br />

He is currently a board member of the ICRS and has been a<br />

member of the ICRS Programme Committee since 2008. In 2012<br />

he co-chairs this committee for the conference in <strong>Montreal</strong> 2012<br />

with Prof. Wayne McIlwraith.<br />

Mandelbaum Bert, MD<br />

Santa Monica Orthopedic & Sports Medicine<br />

Group, California, USA<br />

Bert Mandelbaum is a medical graduate of<br />

Washington University Medical School in St. Louis in 1980,<br />

which completed his residency in Orthopaedic Surgery at The<br />

Johns Hopkins Hospital and fellowship in Sports Medicine from<br />

UCLA. He served on the faculty at UCLA from 1986<strong>–</strong>89 and subsequently<br />

joined the Santa Monica Orthopaedic and Sports<br />

Medicine Group. He presently practices there and serves as the<br />

Director of the Sports Medicine Fellowship Program and the Research<br />

and Education Foundation and Medical Director for the<br />

FIFA Medical Center of Excellence in Santa Monica. He is also<br />

the Director of Research for Major League Baseball (MLB). Academically,<br />

he is well published including multiple journal articles<br />

(85) and five books. He has received five national awards<br />

for Excellence in Research in the Field of Sports Medicine. Since<br />

1995 he has been on the editorial board of the American Journal<br />

of Sports Medicine and associate editor for Current Concept Reviews.<br />

He also served 1999<strong>–</strong>2001 as executive board member for<br />

the American Orthopaedic <strong>Society</strong> for Sports Medicine. Presently,<br />

he is Past President of the <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong>.<br />

He has also has been awarded a NIH Grant on Prevention<br />

of ACL Injuries in Children and Adolescents in collaboration with<br />

Chris Powers PhD of USC. He was honoured in a distinguished<br />

fashion in 2009 with an Honorary Doctorate of Humane Letters (<br />

DHL) from the State University of New York. As a team physician<br />

Dr. Mandelbaum has worked with UCLA Athletics (1985<strong>–</strong>1989)<br />

and Pepperdine University (1990<strong>–</strong>present, LA Galaxy and Chivas<br />

USA MLS teams. He was the Chief Medical Officer for Women’s


Invited Faculty 2012 in alphabetical order (not complete)<br />

World Cup Soccer 1999 and 2003, US Soccer Men’s National<br />

Teams Physician since 1991, and the assistant Medical Director<br />

for Major League Soccer since 1996, and served as USA Team<br />

Physician for Soccer World Cups ’94 in the USA, ’98 in France,<br />

2002 in Japan and Korea ,Germany in 2006 and South Africa in<br />

2010. He serves on the USA Gymnastics Sports Medicine Advisory<br />

Board. In 2002 he was appointed to FIFA Medical Assessment<br />

and Research Committee (F-MARC). In 2007 he was appointed to<br />

FIFA’s Sports Medicine Committee. He also served on the Sports<br />

Medical Committee and Olympic Medical Officer for the Sydney<br />

2000 , Athens 2004 and Beijing 2008.<br />

Mao Jeremy, DDS, PhD<br />

Columbia University College of Dental Medicine,<br />

New York, NY, USA,<br />

Mardones Rodrigo, MD<br />

Clinica Las Condes, Santiago, Chile<br />

Associate Professor Rodrigo Mardones is the<br />

director of the Hip and Knee reconstructive surgery<br />

fellowship program, Department of Orthopedics Surgery<br />

Clinica Las Condes. Auxiliar Profesor of Orthopedics at Universidad<br />

de Chile. The Director of the Tissue Engineering Laboratory<br />

in Orthopedics Stem Cells based Therapy, at Clínica Las Condes.<br />

President of, the Chilean Hip <strong>Society</strong> and Congress President of<br />

the Chilean Sport Medical <strong>Society</strong> (SOCHMEDEP).<br />

He received his MD at the University of Chile in 1998, with maximal<br />

honors, and completed a specialization in orthopedics<br />

at Pontificia Universidad Católica de Chile in 2002 and a twoyears<br />

fellowship program at Mayo Clinic, Rochester Minesotta,<br />

in 2004. In tissue engineering on <strong>Cartilage</strong> <strong>Repair</strong> under the directions<br />

of Dr Shawn Odriscoll and then Clinical Fellow in Adult<br />

Reconstructive Surgery Lower Extremities. Rodrigo Mardones’<br />

research has been focused on hip reconstructive surgery (arthroscopy<br />

and arthroplasty) and cartilage repair with main focus<br />

on cartilage regeneration with differentiated expanded autologous<br />

mesenchymal stem cells. Today the main interest has been<br />

focused in his resently started Tissue Engineering Laboratory at<br />

Clínica Las Condes, the first center in Chile focused on human<br />

stem cell expansion for cell tehrapies in different clinical uses.<br />

He has over 50 per review articles and book chapters and has<br />

been presented numerous podium and guest speaker’s talks on<br />

Hip surgery and <strong>Cartilage</strong> repair<br />

Rodrigo Mardones has been member of the general board of<br />

ICRS and until this year member of the memebership and bylaws<br />

committee. He is actually board member of the ISHA (<strong>International</strong><br />

<strong>Society</strong> of Hip Arthroscopy). Member of numerous scientific<br />

societies including ISHA, AANA, SLARD, ISAKOS, ICRS, SCHOT,<br />

Sochmedep. Part of the editorial board of Cartiage and reviewer<br />

of different journals including CORR, AJSM and Arthroscopy.<br />

Mauck Robert, Ass Prof, PhD<br />

Orthopaedic Surgery and Bioengineering at the<br />

University of Pennsylvania, Philadelphia, USA<br />

Robert Mauck is an Associate Professor of Orthopaedic<br />

Surgery and Bioengineering at the University of Pennsylvania.<br />

Dr. Mauck’s group focuses on the engineering of musculoskeletal<br />

tissues, with a particular interest in restoring articular<br />

cartilage, the knee meniscus, and the intervertebral disc. They use<br />

rigorous mechanical and molecular analyses to characterize native<br />

tissue structure and function, and employ this information to enhance<br />

the functional properties of engineered constructs through<br />

focused technology development. This work employs stem cells,<br />

custom mechanobiologic culture conditions, and novel cell scaffolding<br />

technologies. Dr. Mauck is an active member of a number of<br />

national and international societies, including the ICRS. He serves<br />

on the editorial board of Tissue Engineering and the Journal of the<br />

Mechanical Behavior of Biomedical Materials, and has published<br />

> 80 manuscripts, > 200 abstracts, multiple book chapters,<br />

and co-edited a book on Biomaterials for Tissue Engineering. Dr.<br />

Mauck is currently chair of the Regenerative Medicine Review Panel<br />

for the Veteran’s Administration, and reviews regularly for the<br />

NIH. Dr. Mauck was recently awarded the ISSLS Prize in Biomechanics<br />

(2008), the YC Fung Young Investigator Award from the ASME<br />

(2009), and was selected as a ‘Rising Star’ at the BMES-SPRBM<br />

Conference on Cellular and Molecular Bioengineering (2011).<br />

McIlwraith Wayne, Prof., DVM, PhD<br />

Colorado State University, Fort Collins, USA<br />

Wayne McIlwraith is a University Distinguished Professor,<br />

Barbara Cox Anthony University Endowed<br />

Chair in Orthopaedics, and Director of the Orthopaedic Research<br />

Center (ORC) at Colorado State University (CSU). He also directs the<br />

Musculoskeletal Research Program which is a CSU Program of Research<br />

and Scholarly Excellence. He is an equine orthopaedic surgeon<br />

as well as a researcher and has received numerous awards for<br />

his contributions to orthopaedic surgery and joint research. Professor<br />

McIlwraith received his veterinary degree in 1971 from Massey<br />

University, New Zealand and after two and a half years in private<br />

veterinary practice in New Zealand he did and internship at the University<br />

of Guelph, <strong>Canada</strong> and his surgical residency and MS and PhD<br />

degrees at Purdue University in the USA. He has been at CSU since<br />

1979. He is the co-author of five textbooks, over 400 textbook chapters<br />

and refereed publications, and has given 600 presentations and<br />

workshops. He is a Diplomate of the American College of Veterinary<br />

Surgeons (ACVS) and, The European College of Veterinary Surgeons<br />

(ECVS) and, the American College of Veterinary Sports Medicine and<br />

Rehabilitation (ACVSMR), as well as, a Fellow of the Royal College<br />

of Veterinary Surgeons. His research focuses included osteoarthritis<br />

therapy (including gene therapy and stem cell therapies), articular<br />

cartilage repair and early diagnosis in osteoarthritis and pre-fracture<br />

disease using imaging and fluid biomarkers. He is a Past President<br />

of the American College of Veterinary Surgeons, the American Association<br />

of Equine Practitioners and the Veterinary Orthopaedic <strong>Society</strong><br />

as well as an Associate Member of the American Academy of<br />

Orthopaedic Surgeons. He is Fellow and current Board Member of<br />

the ICRS. In 2012, he Co-Chairs the Programme Committee for the<br />

conference in <strong>Montreal</strong> with Ass. Prof. Jos Malda.<br />

39


40<br />

Invited Faculty 2012 in alphabetical order (not complete)<br />

Melchels Ferry, MSc, PhD<br />

University Medical Centre Utrecht<br />

Utrecht, Netherlands<br />

Ferry Melchels is a young scientist doing basic<br />

research into biomaterials for tissue engineering and drug delivery.<br />

He received his MSc in chemical engineering and PhD in<br />

biomaterials from the University of Twente (The Netherlands) in<br />

2010. Then he moved on to a post-doc position in Professor Dietmar<br />

W. Hutmacher’s Regenerative Medicine group at the Institute<br />

of Health and Biomedical Innovation, Queensland University<br />

of Technology (Brisbane, Australia). Currently he is a Marie<br />

Curie fellow between University Medical Center Utrecht (NL) and<br />

QUT, under supervision of Dr. Jos Malda and Prof. Dietmar Hutmacher.<br />

His principal research interests lie in the development<br />

of polymeric biomedical materials for additive manufacturing<br />

techniques. During his PhD project he developed biodegradable<br />

materials for preparing sophisticated scaffolds for tissue engineering<br />

using stereolithography, and studied the effect of scaffold<br />

architecture on mechanical properties, and on cell seeding<br />

and culturing. His current topic is additive manufacturing including<br />

biology by printing cell-laden hydrogel tissue engineering<br />

constructs, with a particular focus on developing new hydrogel<br />

systems for cartilage regeneration.<br />

Minas Tom, MD<br />

Brigham and Women’s Hospital and Director of<br />

the <strong>Cartilage</strong> <strong>Repair</strong> Center, Boston, USA<br />

Tom Minas is an Attending Orthopedic Surgeon<br />

at the Brigham and Women’s Hospital and Director of the <strong>Cartilage</strong><br />

<strong>Repair</strong> Center. He is a national and international leader in<br />

his work on cartilage repair and his expertise with autologous<br />

chondrocyte implantation (ACI). He also performs arthroscopic<br />

surgery of the knee and hip, joint preserving osteotomies, unicompartmental<br />

knee replacements, patellofemoral joint replacements,<br />

as well as total joint arthroplasty of the knee and hip. Dr.<br />

Minas completed two fellowships: in pelvis & acetabulum, trauma<br />

and joint reconstruction, at the Sunnybrook Medical Centre,<br />

Toronto, <strong>Canada</strong>, in total joint arthroplasty in the Department<br />

Orthopedic Surgery at the Brigham and Women’s Hospital, Boston,<br />

MA, Dr. Minas has a strong focus on joint preservation in<br />

the young adult, by primarily developing and assessing cartilage<br />

repair and osteotomy techniques. He has performed two of the<br />

only ACI’s in the hip joint in the United States and he has also<br />

performed over 400 ACI’s. He has also designed an interpositional<br />

device (Conformis) and patellofemoral joint prosthesis to<br />

help osteoarthritic patients avoid a total knee replacement.<br />

Mithoefer Kai, MD<br />

Sports Medicine and <strong>Cartilage</strong> Restoration at<br />

Harvard Vanguard Medical Associates in<br />

Boston, USA<br />

Kai Mithoefer is currently the Director of Sports Medicine and<br />

<strong>Cartilage</strong> Restoration at Harvard Vanguard Medical Associates in<br />

Boston and clinical faculty member at Harvard Medical School.<br />

He received his medical degree from Heinrich-Heine University in<br />

Düsseldorf Germany in 1991. He obtained his residency training<br />

in Orthopedic Surgery at Harvard Medical School and completed<br />

a fellowship in Shoulder and Sports Medicine at the Hospital for<br />

Special Surgery in New York. Dr. Mithoefer is board certified in<br />

Orthopedic Surgery and Sports Medicine in both the USA and<br />

Germany. He has been a member of the ICRS since 2003 and is<br />

currently serving as the co-chair of the ICRS Rehabilitation and<br />

Sports Committee. His research interest is in the clinical application<br />

and evaluation of novel tissue engineering techniques for<br />

articular cartilage repair with a special focus on their use in the<br />

high-demand athletic population. Kai Mithoefer has served as<br />

an active member of the Scientific Program Committee for the<br />

2010 ICRS Meeting in Sitges/Barcelona. He is a member of the<br />

editorial board of the journal “<strong>Cartilage</strong>” and has co-edited the<br />

2012 FIFA/ICRS Supplement on Articular <strong>Cartilage</strong> Injury in the<br />

Football (Soccer) Player as well as the 2011 ICRS Newsletter on<br />

<strong>Cartilage</strong> Rehabilitation.<br />

Nehrer Stefan, Prof., MD, PhD<br />

Department for Orthopaedic Surgery at the<br />

hospital in Krems, Austria<br />

Stefan Nehrer is an orthopaedic surgeon at<br />

Department for Orthopaedic Surgery at the hospital in Krems,<br />

Professor for Tissue Engineering at Center for Regenerative<br />

Medicine and since 2011 he is also Dean of Faculty Health and<br />

Medicine at Danube University Krems. He studied at the Medical<br />

University Vienna where he obtained his MD in 1984 and his<br />

PhD in 1999. From 1995 to 1996 he was at the Harvard Medical<br />

School in Boston, USA, at Prof. Myron Spector where he started<br />

his scientific work in cell-based therapies in cartilage regeneration.<br />

From 2000 to 2006 he was head of orthopaedic research<br />

at the Medical University Vienna and leading surgeon in sports<br />

medicine and paediatric orthopaedic. In 2007 he was appointed<br />

Professor for Tissue Engineering at Danube University Krems.<br />

Over the years he has continued his research on experimental<br />

and clinical applications of chondrocyte transplantation and<br />

formed a group for tissue engineering research. Furthermore,<br />

his interests focused on mesenchymal cell differentiation and<br />

design/implementation of tissue culture bioreactors for automated<br />

and controlled manufacturing of cartilage, bone and osteochondral<br />

grafts, based on autologous cells and 3D porous<br />

scaffolds. He has published more than 52 peer reviewed and<br />

81 other articles in national and international journals and 14<br />

book chapters. Prof. Nehrer has presented at many national and<br />

international meetings and he is member of 10 national and international<br />

societies.


Invited Faculty 2012 in alphabetical order (not complete)<br />

Neyret Philippe, Prof.<br />

Department of Orthopaedic and Traumatological<br />

Surgery at the Croix Rousse Hospital<br />

Lyon, France<br />

Philippe Neyret is an orthopaedic surgeon and the head of department<br />

of Orthopaedic and Traumatological Surgery at the<br />

Croix Rousse hospital, Lyon, France. As a Professor, he is responsible<br />

for three academic formations at the University of Lyon,<br />

France. Philippe Neyret received his initial training with Albert<br />

TRILLAT and being a former pupil of Henri DEJOUR, it was quite<br />

natural that in 1988 he devoted his activity on knee surgery. Philippe<br />

Neyret was nominated Professor at the University of Lyon<br />

in 1994, and he is head of the department at the Centre Livet<br />

since 1996. His main activity is clinical. Philippe Neyret treats<br />

an equal number of sports lesions and degenerative pathologies<br />

of the knee. The many publications, books, CD-Roms reflect<br />

this clinical research activity. For his work on the results after<br />

meniscectomy, Philippe Neyret received the “John Joyce Award”<br />

by <strong>International</strong> Arthroscopy in 1989. In 1995 Philippe Neyret<br />

participated in the ESSKA-AOSSM “Traveling Fellowship”. His<br />

participation during the “7th, 9th journées lyonnaises de chirurgie<br />

du genou” dedicated to the degenerative knee and the “15th<br />

Journées” dedicated to the total knee arthroplasty shows his<br />

great interest for the degenerative pathology of the knee. Philippe<br />

Neyret is member of several medical societies and he is an<br />

active participant in the SOFCOT, ESSKA 2000, EFORT as member<br />

at large and ISAKOS as Second Vice President. He is also<br />

Second Vice Chair of the “ACL Study Group”, and member of the<br />

board of the “Patello Femoral Foundation”. In 1999 he was involved<br />

in the development of the subjective IDC form and more<br />

recently under the leadership of Anderson in the development of<br />

the meniscal documentation form. In 2000, Philippe Neyret was<br />

co-organizer of the Journées Lyonnaises de Chirurgie du Genou<br />

held in Porto Allègre, Brasil, in 2004, in Fortaleza (Brasil) and in<br />

2007 Florianapolis. In 2002-2004, 2008 and 2010, together with<br />

Dr Chambat, Philippe Neyret organised the “Journées de Chirurgie<br />

du genou” devoted to the athlete’’ knee or/and prosthesis<br />

knee. In 2004, 2006 and 2008, Philippe Neyret was part of the<br />

scientific committee of the European course of Knee Arthroplasty<br />

in Val d’Isère.<br />

Nixon Alan, Prof., BVSc, MS<br />

Department of Clinical Sciences,<br />

Cornell University<br />

Ithaca, USA<br />

Alan Nixon is Professor of Orthopedic Surgery and Director<br />

of The Comparative Orthopaedics Laboratory and the JD&ML<br />

Wheat Orthopaedic Sports Medicine Laboratory at Cornell University.<br />

He has an adjunct appointment as Professor at Colorado<br />

State University Graduate Field. He obtained his veterinary<br />

degree from the University of Sydney in 1978 and completed a<br />

surgical residency and research degree at Colorado State University<br />

in 1983. After five years in the Department of Surgical<br />

Sciences at the University of Florida, Dr. Nixon moved to New<br />

York in 1988 where he is currently a Professor in the Department<br />

of Clinical Sciences, and served as Chief of Surgery from 1998 to<br />

2002. Dr Nixon has authored over 290 papers and book chapters,<br />

and has written 2 texts on equine orthopedics. Dr Nixon’s<br />

clinical and teaching at Cornell University focus on musculoskeletal<br />

injury and repair, with a specific interest in regenerative<br />

medicine. Research and translational clinical application over<br />

the past 2 decades have included: Joint pathobiology and cartilage<br />

repair with cell grafting, growth factor recombinant protein<br />

and gene-enhanced chondrocyte and stem cell transplantation<br />

techniques, Stem cell propagation, characterization, and application<br />

in musculoskeletal diseases, including bone marrow and<br />

adipose derived stem cells for repair of tendinitis, Clinical application<br />

of growth factor recombinant proteins and gene therapy<br />

for improved joint, tendon, and bone repair, Genetic characterization<br />

of OCD in animals and man using microarray gene chip expression<br />

studies, Arthroscopic approaches and techniques for<br />

enhanced repair in the stifle, shoulder, elbow, and hip in horses.<br />

Dr Nixon’s laboratory group has engaged in over 80 funded research<br />

projects and 8 contracts with industry engaged in musculoskeletal<br />

research, with total budget expenditures of over $17<br />

million. He currently has a 5-year National Institutes of Health<br />

R01 award with total costs of 1.8 million dollars. Dr Nixon has<br />

lectured on his research findings in the US and abroad on over<br />

110 occasions. He serves as a consultant to industry and to the<br />

FDA panel on Cell and Gene Therapy.<br />

Pacifici Maurizio, Prof.<br />

Division of Orthopaedic Surgery,<br />

Children’s Hospital of Philadelphia, USA<br />

Pacifici received his doctorate degree from the<br />

University of Rome and postdoctoral training under the auspices<br />

of a European Molecular Biology Fellowship. He joined the faculty<br />

at the University of Pennsylvania where he rose to the rank of<br />

Professor. He subsequently moved to Jefferson University Medical<br />

School where he served as Director of Research in Orthopaedics.<br />

About two years ago, he and his team were recruited by<br />

the Children’s Hospital of Philadelphia. Dr. Pacifici’s biomedical<br />

research work focuses on mechanisms controlling skeletal development<br />

and growth in fetal and postnatal life. Emphasis is on<br />

the identification of molecular regulators acting at the nuclear<br />

levels that direct commitment, determination and differentiation<br />

of progenitor skeletal cells. The overall goal is to target<br />

those regulators using gene-, cell- and drug-based therapies to<br />

treat skeletal pathologies including congenital skeletal malformations<br />

or growth defects and acquired conditions such as Heterotopic<br />

Ossification. Emphasis is also on signalling diffusible<br />

factors that normally act within developing skeletal elements<br />

to coordinate growth and morphogenesis. When these factors<br />

act abnormally and affect adjacent non-skeletal tissues due to<br />

failure of signalling or restraining mechanisms, they can trigger<br />

pathologies, including benign tumours such as those seen Hereditary<br />

Multiple Exostoses. Experimental therapies are being<br />

tested to restore normal signalling mechanisms and block or<br />

reverse these and related pathologies. Dr. Pacifici’s biomedical<br />

research work has been continuously funded by the NIH for over<br />

25 years.<br />

41


42<br />

Invited Faculty 2012 in alphabetical order (not complete)<br />

Philippon Marc J., MD<br />

The Steadman Clinic<br />

Vail, USA<br />

Marc J. Philippon is a partner at The Steadman<br />

Clinic and is one of the world's leading orthopaedic hip surgeons.<br />

Dr. Philippon joined The Steadman Clinic in 2005 after<br />

a successful time period at the University of Pittsburgh Medical<br />

Center where he served as Director of sports related hip disorders<br />

and was also Director of the UPMC Golf Medicine Program.<br />

Previously, he was chief of orthopaedic surgery at Holy Cross<br />

Hospital in Fort Lauderdale, Florida. Dr. Philippon is internationally<br />

known for performing joint preservation techniques utilizing<br />

arthroscopic hip surgery to treat painful joint injury in highlevel<br />

athletes who constantly use powerful hip rotation, such as<br />

golfers, NFL players and NHL players, making themselves prone<br />

to acute and chronic injuries of the hip joint. He has published<br />

numerous scientific articles in sports medicine and orthopaedic<br />

journals and is a frequently invited presenter at international<br />

sports medicine and orthopaedic meetings. Dr. Philippon earned<br />

his medical degree with an academic scholarship from Mc-<br />

Master University Medical School in Hamilton, Ontario, <strong>Canada</strong><br />

in 1990, and completed an orthopaedic surgery residency at the<br />

University of Miami, Jackson Memorial Hospital in 1995. Dr. Philippon<br />

is a consultant to NHL, NFL, NBA, and MLB Professional<br />

teams and has treated numerous PGA golfers. He is also a consultant<br />

to the NHL Players Association. Some of the Professional<br />

athletes he has treated include golfers Greg Norman and Peter<br />

Jacobsen, hockey player Mario Lemieux, Professional football<br />

player Priest Holmes, and baseball player Alex Rodriquez. Dr.<br />

Philippon is an Active Member with the American Orthopaedic<br />

<strong>Society</strong> for Sports Medicine and the Arthroscopy Association of<br />

North America. He is also a Fellow with the American Academy<br />

of Orthopedic Surgeons and a Master Instructor with the Arthroscopy<br />

Association of North America, Masters Experience Hip<br />

Course, and a Member of the Herodicus <strong>Society</strong>. Dr. Philippon<br />

enjoys spending time with his family and participating in sports<br />

such as skiing, tennis, swimming and cycling.<br />

Poole A. Robin, Prof. Emeritus, BSc<br />

McGill University, Lancaster<br />

Ontario, <strong>Canada</strong><br />

Robin Poole received his BSc degree 1961 and his<br />

PhD degree in 1969 from Reading University, U.K. Following early<br />

research in microbiology (in industry), on membrane fusion and<br />

tissue invasion in cancer, he was invited in 1970 to join a research<br />

group at the Strangeways Research Laboratory in Cambridge, England<br />

working on cartilage degradation in arthritis. In 1977 he moved<br />

to <strong>Montreal</strong> to the Shriners Hospital and McGill University to create<br />

and become director of a new research laboratory (The Joint Diseases<br />

Laboratory) working on arthritis. He became a full Professor<br />

at McGill in 1981.Here he established an international research<br />

group working on skeletal development and cartilage degradation<br />

in arthritis. This was funded by the Medical Research Council of <strong>Canada</strong>/<br />

Canadian Institutes of Health research, Shriner’s Hospitals,<br />

industry and National Institutes of Health, USA (NIH). The successes<br />

of his research on the pathobiology of inflammatory and degenerative<br />

joint disease, focussing on cartilage degradation in health and<br />

disease, has led to numerous awards. These include the Kappa Delta<br />

Award of the American Academy of Orthopaedic Surgeons/Orthopaedic<br />

Research <strong>Society</strong>; the Howard and Martha Holley Research<br />

Prize in Rheumatology and the award of Master, the latter two from<br />

the American College of Rheumatology; the Carol Nachman <strong>International</strong><br />

Research Prize in Rheumatology; Honorary Doctor of Science<br />

degree, Reading University, England; Lifetime Achievement Award<br />

of the Osteoarthritis Research <strong>Society</strong> <strong>International</strong>; President, 6th<br />

World Congress of Inflammation Research Associations; President,<br />

Canadian Orthopaedic Research <strong>Society</strong> and Professor Emeritus<br />

at McGill University on his retirement from the laboratory at the<br />

Shriner’s Hospital in December, 2005. He also worked in an advisory<br />

capacity with the Food and Drugs administration (FDA), USA. He has<br />

personally trained over 40 graduate students and fellows from clinical<br />

and basic research backgrounds and assisted many others. He<br />

continues to mentor a number of trainees and young investigators<br />

in his “retirement”. His research has resulted in 238 peer reviewed<br />

papers and 93 invited reviews/book chapters. His work on molecular<br />

markers of cartilage matrix metabolism and joint damage and<br />

repair in arthritis has led to their commercialization and use in research<br />

and drug development in academia and industry. He wrote<br />

the white paper on biomarkers for the NIH Osteoarthritis Initiative<br />

in 2000 and recently was involved in the preparation for the FDA of<br />

a guidance document on biomarker usage in clinical trials for osteoarthritis.<br />

He was a co-founder and later Scientific Director of the<br />

Canadian Arthritis Network, a National Centre of Excellence composed<br />

of almost 200 principal investigators. This was founded in 1998<br />

to promote collaborative research activities and research training in<br />

the arthritis field within <strong>Canada</strong> and internationally and the translation<br />

of new knowledge to improve the care of patients with arthritis.<br />

He maintains his numerous scientific and research interests being<br />

involved in research collaborations, mentoring, reviewing, lecturing<br />

and editorial and scientific advisory boards, including Osteoarthritis<br />

Research <strong>Society</strong> <strong>International</strong>. He continues to act as a consultant<br />

to biotech and pharmaceutical companies.<br />

Quinn Thomas, Ass Prof., PhD,<br />

McGill University, Lancaster<br />

Ontario, <strong>Canada</strong><br />

Thomas M. Quinn obtained a BSc in Engineering<br />

Physics from Queen’s University at Kingston, <strong>Canada</strong> and a PhD in<br />

Mechanical and Medical Engineering from the Harvard-MIT Division<br />

of Health Sciences and Technology, USA in 1996. After a twoyear<br />

postdoctoral fellowship at the Müller Institute for Biomechanics<br />

in Bern, he established the <strong>Cartilage</strong> Biomechanics Group<br />

at EPFL in Lausanne, Switzerland. He joined the Department of<br />

Chemical Engineering at McGill University, <strong>Canada</strong> in 2007, where<br />

he is currently Associate Professor and <strong>Canada</strong> Research Chair in<br />

Soft Tissue Biophysics. His research aims to clarify and use cell<br />

responses to soft tissue deformation, and has emphasized the<br />

electromechanics of cartilage matrix, cell responses to cartilage<br />

injury, transport of fluids and solutes through soft tissues, and<br />

development of technologies for cell culture on extendable substrates<br />

for improved tissue engineering.


Invited Faculty 2012 in alphabetical order (not complete)<br />

Richardson James, Prof., MD<br />

Institute of Orthopaedics, Robert Jones &<br />

Agnes Hunt Orthopaedic Hospital, Oswestry,<br />

Shropshire, UK<br />

James Richardson is an orthopaedic surgeon who has studied<br />

cells in culture for over 20 years. The development of ACI in Gothenburg<br />

led to him leading a team at Oswestry in developing a<br />

GMP laboratory within the British National Health Service. Eurocell<br />

and MyJoint have been EU funded research projects. He<br />

has treated over 400 patients with ACI and combines a range of<br />

reconstructive techniques in the hip, knee and ankle.<br />

Rodeo Scott, Prof., MD<br />

Orthopaedic Surgery, Weill Medical College of<br />

Cornell University, USA<br />

Scott Rodeo is a clinician-scientist at Hospital<br />

for Special Surgery, with appointments in the Department of Orthopaedic<br />

Surgery (Sports Medicine and Shoulder Service) and<br />

the Research Department (Laboratory for Soft Tissue Research).<br />

He is Professor of Orthopaedic Surgery at Weill Cornell Medical<br />

College and Co-Chief of the Sports Medicine and Shoulder<br />

Service at Hospital for Special Surgery. He specializes in sports<br />

medicine injuries of the knee, shoulder, ankle, and elbow. He<br />

also performs arthritis surgery of the knee and shoulder, including<br />

joint replacement surgery. His research focuses on the basic<br />

biology of tendon and ligament healing, meniscal allograft<br />

transplantation, and rotator cuff repair. He has been Associate<br />

Team Physician of the New York Giants football team since 2000.<br />

In 2004 and 2008, he served as Team Physician for USA Olympic<br />

Swimming, a position he will return to in London for the 2012<br />

games. Dr. Rodeo also holds a Board position at Asphalt Green,<br />

a community-based athletic organization in Manhattan, where<br />

he helps to promote injury prevention and healthy living through<br />

exercise and health education information to its members.<br />

Sah Robert, Prof., MD<br />

Bioengineering, University of California,<br />

San Diego, USA<br />

Robert L. Sah is Professor of the Department of<br />

Bioengineering, Adjunct Professor of the Department of Orthopaedic<br />

Surgery, and co-director of the Center for Musculoskeletal<br />

Research at UCSD. He received the B.S. and M.S. in Electrical<br />

Engineering and the Sc.D. in Medical Engineering from M.I.T.,<br />

and the M.D. from Harvard. He joined UCSD Bioengineering in<br />

1992 and was promoted to Professor in 2001. Dr. Sah has contributed<br />

to the Tissue Engineering and Regenerative Medicine<br />

<strong>International</strong> <strong>Society</strong> as a North America council member, to<br />

the <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong> as a member of the<br />

Executive Board, to a number of journals including Arthritis and<br />

Rheumatism, <strong>Cartilage</strong>, Journal of Orthopaedic Research, Osteoarthritis<br />

and <strong>Cartilage</strong>, and Tissue Engineering in editorial<br />

roles. His research interests are the biomechanics, mechanobiology,<br />

and tissue engineering of articular cartilage, synovial fluid,<br />

and synovial joints with the ultimate goal of improving the treatment,<br />

diagnosis, and prevention of osteoarthritis. He is recipient<br />

of the Van C. Mow medal, a fellow of the American Institute for<br />

Medical and Biological Engineering, and a Professor of the Howard<br />

Hughes Medical Institute.<br />

Sanchez Luis J., MD, DPM<br />

Podiatric Medicine & Surgery of the Foot & Ankle<br />

He received his MD from the CETEC School of<br />

Medicine, Santo Domingo, Dominican Republic<br />

in 1983. He worked from 1991<strong>–</strong>1996 at the Orlando Foot &<br />

Ankle Clinic, Inc, then 1996<strong>–</strong>2000 at Accent Foot, Ankle, & Leg<br />

Clinic and from 2000<strong>–</strong>2009 at the Orlando Foot & Ankle Clinic.<br />

Dr. Sanchez held teaching positions from 1983<strong>–</strong>2008 in Puerto<br />

Rico, Dominican Republic, Mexico as well as in Tallahassee, FL,<br />

USA mainly multiple lectures related to the field of Foot & Ankle,<br />

Leg medicine and surgery.,<br />

Sandell Linda, Prof., PhD<br />

Washington University School of Medicine,<br />

Washington, USA<br />

Linda Sandell is the Mildred B. Simon Professor<br />

and Director of Research in the Department of Orthopaedic Surgery<br />

and Director of the Center for Musculoskeletal Biology and<br />

Medicine at Washington University in St. Louis. She has been a<br />

leader in the field of orthopaedic research, pioneering the use of<br />

molecular biologic techniques, protein biochemistry, large screening<br />

technologies, microscopy and computational biology to<br />

study cell responses to cartilage cell injury and the regulation of<br />

gene expression in connective tissues. Dr. Sandell received her<br />

bachelor’s and master’s degrees from Denver University, her Ph.D.<br />

in biochemistry at Northwestern University, and did postdoctoral<br />

work in molecular biology at the University of Chicago. Dr. Sandell<br />

was a faculty member at Rush Medical College in Chicago<br />

from 1982 to 1987, and at the University of Washington in Seattle<br />

from 1987 to 1997 when she moved to Washington University. Dr.<br />

Sandell has authored more than 200 publications, in addition<br />

to three books and seven patents. She has provided extensive<br />

leadership in the orthopaedic research field, particularly in the<br />

Orthopaedic Research <strong>Society</strong>, as President in 2000, and as a<br />

founder of the Women’s Leadership Forum. Over the past several<br />

years, has chaired three Gordon Conferences and numerous<br />

other conferences including co-founding the <strong>Cartilage</strong> Gordon<br />

Conference. She has been President of the Orthopaedic Research<br />

<strong>Society</strong> (2000), the Histochemical <strong>Society</strong> (2005-2007) and the<br />

<strong>Society</strong> for Matrix Biology (2006-2008). In 2010, she received the<br />

Women’s Leadership Award from the Orthopaedic Research <strong>Society</strong><br />

and is currently director of the OARSI OA Biomarkers Global<br />

43


44<br />

Invited Faculty 2012 in alphabetical order (not complete)<br />

Initiative and President of the Osteoarthritis Research <strong>Society</strong> <strong>International</strong><br />

(2010- 2013). Dr. Sandell has been awarded the Kappa<br />

Delta Award for Basic Science Research by the American Association<br />

for Orthopaedic Surgeons, and has served on the Advisory<br />

Council of the National Institute for Arthritis, Musculoskeletal and<br />

Skin Diseases. She is Deputy Editor for the Journal of Orthopaedic<br />

Research, Associate Editor for the Journal of Histochemistry and<br />

Cytochemistry, and is a member of the editorial boards for many<br />

other journals including Arthritis Research and Care, the Journal<br />

of Biological Chemistry, and Osteoarthritis and <strong>Cartilage</strong>.<br />

Saris Daniël, Prof., MD, PhD<br />

Orthopaedic Department, University Medical<br />

Center Utrecht, NL<br />

Graduated University of Amsterdam Medical<br />

School in 1992. During orthopaedic residency he did a fellowship<br />

at the Mayo Clinic in Rochester MN USA under Prof. Shawn<br />

O’Driscoll of the <strong>Cartilage</strong> and Connective Tissue Research Laboratory<br />

and Prof. Kai-Nan An of the orthopaedic biomechanics<br />

laboratory. His PhD thesis completed in 2002 at the University<br />

of Utrecht was titled “Joint Homeostasis in Tissue Engineering<br />

for <strong>Cartilage</strong> <strong>Repair</strong>”. It first introduced the now generally accepted<br />

concept of joint homeostasis. In 2000 Daniël joined as<br />

staff member in the department of Orthopaedics at the UMC Utrecht.<br />

In March of 2010 Dr Saris was appointed as Professor of<br />

Reconstructive Medicine at the University of Twente. Prof. Dr Saris<br />

is co director of the Biological Joint Reconstruction research<br />

program and head of the orthopaedic residency program at the<br />

University of Utrecht. He describes his goal and driving force as<br />

a wish to satisfy natural curiosity into optimizing the regenerative<br />

biological capacity of the musculoskeletal system, improve<br />

treatment and understanding of knee afflictions and to help in<br />

building an international network for ICRS with high potential<br />

and productive group dynamics.<br />

Schmidt Tannin, Ass Prof., PhD<br />

Centre for Bioengineering Research and<br />

Education, University of Calgary, Calgary, <strong>Canada</strong><br />

Tannin Schmidt is also an Associate Director of<br />

the Biomedical Engineering Graduate Program. He received his<br />

B.A.Sc. in Engineering Science from the University of Toronto in<br />

2000, then his MS and PhD in Bioengineering from the University<br />

of California San Diego in 2002 and 2006, respectively. He<br />

then completed a post doctoral fellowship in the Department<br />

of Biochemistry at Rush University Medical Center, Chicago.<br />

His bioengineering research interests lie within cartilage biomechanics,<br />

biotribology, and biochemistry. Long term cartilage<br />

research interests include mechanistic based, multidisciplinary<br />

study of articular cartilage boundary lubrication through clinical,<br />

biochemical and engineering collaborations. Relevant areas<br />

include the study of normal, injured, and diseased cartilage and<br />

synovial fluid, where composition, structure, and interactions of<br />

mechanically relevant biomolecules, such as proteoglycan 4 (lubricin)<br />

and hyaluronan, can be altered, as well as other tissues<br />

in the joint affected by injury and disease.<br />

Shive Matthew, BSc, MSc, PhD<br />

Piramal Healthcare,<br />

Laval, <strong>Montreal</strong>, CA<br />

Matthew Shive is currently a consultant specializing<br />

in research and development of orthopaedic medical devices.<br />

Dr Shive received his B.S. from the Johns Hopkins University, and<br />

his M.Sc. and Ph.D. from Case Western Reserve University, all in Biomedical<br />

Engineering with a focus on Biomaterials. His scientific expertise<br />

relates to the interactions between implanted biomaterials<br />

and biological systems supported by over 100 publications, scientific<br />

articles and conference proceedings in the fields of biomaterials,<br />

tissue and cartilage repair. From 2000<strong>–</strong>2009, Dr Shive oversaw<br />

product development as Chief Scientific Officer at BioSyntech <strong>Canada</strong><br />

Inc, a medical device company in <strong>Montreal</strong> that developed and<br />

manufactured regenerative products for tissue repair. Notably, Dr.<br />

Shive designed and implemented the clinical development program<br />

for BST-CarGel, a device for cartilage repair, that was the subject of<br />

a multicenter, randomized clinical trial in <strong>Canada</strong>, Spain and Korea<br />

completed in Feb 2010 and which will now support commercialization<br />

applications. The Company's assets were recently acquired<br />

(June 2010) by Indian pharmaceutical giant, Piramal Healthcare. Dr.<br />

Shive’s current efforts as a primary consultant to Piramal Healthcare<br />

work towards completion of previous product development, clinical<br />

and regulatory activities. He is responsible for analysis and interpretation<br />

of efficacy data resulting from multicenter BST-CarGel clinical<br />

trial and subsequent reporting and publications.<br />

Spalding Tim, Ass Prof.<br />

University of Warwick Leamington, UK<br />

Tim Spalding is a specialist knee surgeon at<br />

University Hospitals Coventry and Warwickshire<br />

NHS Trust and Honorary Associate Professor at the University<br />

of Warwick. His specialist interest in reconstructive knee surgery<br />

includes meniscal transplantation, articular cartilage repair,<br />

ligament reconstruction including multi-ligament injuries, and<br />

osteotomy. Training took place in Oxford and at Royal Hospital<br />

Haslar, prior to a specialist arthroscopy and knee surgery fellowship<br />

in Toronto in 1994<strong>–</strong>1995. He qualified in 1982 from Charing<br />

Cross Hospital, London and spent the first part of his medical<br />

career with the Royal Marines and the Royal Navy. He joined Coventry<br />

in 2000 after 5 years as a Consultant in the armed forces.<br />

He has a busy sports knee surgery practise, runs a knee fellowship<br />

program and continues to be very active in teaching and<br />

research, pioneering several new techniques. His hobbies are<br />

his family and his competitive sailing.


Invited Faculty 2012 in alphabetical order (not complete)<br />

Steinwachs Matthias, Prof. h.c. PD MD<br />

Schulthess Klinik<br />

Zurich, Switzerland<br />

Consultant Orthopaedics, Traumatology & Sports<br />

Medicine, Head of the Center of Orthobiologics & <strong>Cartilage</strong> <strong>Repair</strong>,<br />

Schulthess Klinik, Zürich, (FIFA Medical Centre of Excellence, Swiss<br />

Olympic Base, Member of ISOC). Study of Medicine, University of<br />

Heidelberg & Göttingen, 1990 Research study at the Dept. of Pharmacology<br />

and Toxicology, University of Göttingen, 1/1991<strong>–</strong>3/1992<br />

Ass. Doctor Dept. of Trauma Surgery, Bernward-Hospital Hildesheim,<br />

1992 Dissertation MD/PhD University of Göttingen, 1997<br />

Specialist education, Dept. for Orthopaedic Surgery, University<br />

of Freiburg, 1995 Training by Prof. L. Peterson, GMC Gothenburg,<br />

Sweden, 1997 Specialist exam for Orthopaedic Surgery, University<br />

of Freiburg, 1997<strong>–</strong>2007 Specialist for <strong>Cartilage</strong> <strong>Repair</strong> and Tissue<br />

Engineering, Knee Surgery, Dept. of Orthopaedic and Trauma<br />

Surgery, University Hospital Freiburg, 1998<strong>–</strong>2007 Member of the<br />

Steering Committee Valley Tissue Engineering Centre University<br />

Freiburg, 1998<strong>–</strong>2007 Head of the <strong>Cartilage</strong> Research Group, Valley<br />

TEC Freiburg, 12/2004 “Privatdozent” (Habilitation), Venia legendi<br />

for Orthopaedic Surgery, University Freiburg, 2003<strong>–</strong>2007 Head of<br />

the first <strong>Cartilage</strong> Transplantation Unit, Dept. of Orthopaedic and<br />

Trauma Surgery, University Hospital Freiburg, 2003- Expert of<br />

“Bundesausschuss Krankenhaus (G-BA), Methodenbewertung<br />

ACT”, 2005-Member of the Expert board BMBF Project „Regenerative<br />

Medizin, 2006 Professional Development for Sports Medicine,<br />

2006 Honorary Professor Inst. of Sports medicine, Peking University<br />

2007, 2008 Specialist for Orthopaedic & Traumatology 2008<br />

Member of Expert board EMEA, London and Paul Erlich-Inst. (PEI,<br />

Langen), Congress President AGA 2012, Zürich. ICRS Board Member<br />

2004<strong>–</strong>2008, Chair of the outcome committee of the ICRS 2008.).<br />

Van Osch Gerjo, Prof., PhD<br />

Erasmus MC, University Medical Center<br />

Rotterdam, Netherlands<br />

Gerjo van Osch (1967) studied medical biology at<br />

the University of Utrecht (MSc 1990) and received her PhD in 1994<br />

at the University of Nijmegen on animal models for osteoarthritis.<br />

Since then she became involved in cartilage tissue engineering and<br />

especially the use of different cell types and growth factors. She is<br />

currently appointed as associate Professor at the Erasmus MC, University<br />

Medical Center in Rotterdam the Netherlands where she is<br />

leading a research group of approx. 12 people that is part of the departments<br />

of Orthopaedics and Otorhinolaryngology. Her research<br />

focuses on cellular aspects of connective tissue degeneration and<br />

regeneration. (www.erasmusmc.nl/orthopaedie/research/labor/<br />

CTCRgroup). Gerjo van Osch has been working in the field of cartilage<br />

since 1990. She is co-author on over 100 international peer-reviewed<br />

publications. She has been active in various committees of the<br />

<strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong> (ICRS). She served as council<br />

member of the European chapter of the TERMIS and chaired the<br />

TERMIS-EU meeting in Rotterdam in 2006. Presently she is board<br />

member of ICRS and European Science Foundation network on Regenerative<br />

Medicine (REMEDIC). She is associate editor of <strong>Cartilage</strong><br />

and editorial board member of Tissue Engineering and Journal of<br />

Tissue Engineering and Regenerative medicine.<br />

Van Roermund Peter, MD<br />

University Medical Center Utrecht, Netherlands<br />

Peter van Roermund was born on 30 April 1949 in Kockengen,<br />

The Netherlands. His Professional experience:<br />

1985 <strong>–</strong> today staff member, department of orthopaedics ,UMC Utrecht.<br />

2001<strong>–</strong>2003: President Dutch Orthopaedic <strong>Society</strong>, 2005-present: President<br />

Bone and Joint Decade the Netherlands. Reports and documents written:<br />

Thesis in 1994: Tibial lengthening by distraction epiphysiolysis. Clinical<br />

and experimental studies. University of Utrecht, Award 2002: The Kenneth<br />

A.Johnson Memorial Award and Lecturer AOFAS winter meeting Dallas USA:<br />

joint distraction as treatment of OA ankle joints.<br />

Walsh David, Prof.<br />

Arthritis Research UK Pain Centre<br />

University of Nottingham, UK<br />

David Walsh is a clinical academic at the University of<br />

Nottingham Medical School and Director of the Arthritis Research UK Pain<br />

Centre. The Centre undertakes a translational programme of research exploring<br />

the mechanisms by which changes within the joint and in the nervous<br />

system interact with psychosocial factors to result in the pain of arthritis in<br />

order to develop new treatments and to enhance the effectiveness of current<br />

therapies. His preclinical research has focused on structural changes<br />

that contribute to joint pain, in particular angiogenesis, nerve growth and<br />

inflammation in the synovium and subchondral bone. This research benefits<br />

from a repository of joint tissues donated by > 2500 people with arthritis<br />

and > 400 post mortem cases, complemented by a range of preclinical inflammatory<br />

and osteoarthritis models. He is also co-Chief Investigator of<br />

the Early Rheumatoid Arthritis Network (ERAN), a registry for patients with<br />

Early RA recruiting from centres across the UK and Eire. Professor Walsh is<br />

Consultant Rheumatologist at Sherwood Forest Hospitals NHS Foundation<br />

Trust, where he developed an Early Rheumatoid Arthritis service and is Clinical<br />

Director of the Back Pain Unit, providing multidisciplinary Pain Management<br />

Programmes for people with chronic low back pain.<br />

Welsch Goetz, Ass Prof.<br />

University of Erlangen, Department of Trauma<br />

Surgery, Erlangen, Germany<br />

Goetz H. Welsch is a German board certified orthopaedic<br />

and trauma surgeon with the speciality of knee surgery and sports medicine.<br />

Furthermore he is team physician of a Professional German soccer<br />

team. His clinical and scientific focus is since more than 10 years regenerative<br />

cartilage therapy including different techniques of cartilage repair procedures.<br />

Besides this, Dr. Welsch is a well known musculoskeletal imaging<br />

expert in close collaboration with the Medical University of Vienna, Austria.<br />

He has been involved in several studies mainly concerning advanced<br />

MR imaging technologies in cartilage repair. In this field he has published<br />

many studies about the MOCART score and recently the 3D MOCART score.<br />

Furthermore he works very intensely in biochemical cartilage imaging techniques<br />

like T2 mapping, dGEMRIC, T1rho and Diffusion Weighted Imaging.<br />

Especially within the field of quantitative T2 mapping, Dr. Welsch published<br />

many studies and is one of the most active researchers world-wide.<br />

Dr. Welsch is member of different orthopaedic and radiological societies,<br />

authored more than 60 peer-reviewed papers and about 20 book chapters,<br />

is an active reviewer for more than 15 journals and in the advisory board of<br />

Archives of Orthopaedic and Trauma Surgery (AOTS).<br />

45


46<br />

Industry Exhibition & Sponsoring<br />

Technical Exhibit Opening Hours<br />

Saturday 12.00 <strong>–</strong> 20.00 hrs.*<br />

Sunday 09.00 <strong>–</strong> 18.00 hrs.<br />

Monday 09.00 <strong>–</strong> 18.00 hrs.<br />

Tuesday 09.00 <strong>–</strong> noon.<br />

Sufficient time during intermissions is reserved for visiting<br />

the booths of leading manufacturers which present<br />

the latest achievements and give competent information.<br />

For detailed information on the exhibiting companies,<br />

please consult the exhibit guide on the following pages.<br />

ICRS 2012 and the ICRS <strong>Society</strong> express their gratitude<br />

to all collaborators and volunteers for this meeting.<br />

Particularly the participation of the following companies<br />

is much appreciated and gratefully acknowledged:<br />

Exhibitors (Alphabetical Order)<br />

Company Booth Nr.<br />

Anika Therapeutics Inc. 15<br />

Arthrex, Inc. 13<br />

ArthroCare 19<br />

Bacterin 14<br />

Biomomentum Inc 18<br />

BioPoly 20<br />

BioTissue AG 26<br />

Croma-Pharma GesmbH 5<br />

DePuy Mitek 23<br />

DJO Global 2<br />

Fin-Ceramica Faenza S.p.A 11<br />

Geistlich Pharma AG 8<br />

Harvest Technologies 14A<br />

Histogenics 25<br />

Knee Creations, LLC 22<br />

Moximed, Inc. 10<br />

Piramal Healthcare Ltd. 7<br />

REGEN Lab 17<br />

Regentis Biomaterials LTD 16<br />

SAGE Publications 9<br />

Sanofi Genzyme Biosurgery 6<br />

SBM Inc. Science & Bio Materials 12<br />

Smith & Nephew Inc. 4<br />

Tigenix 1<br />

Zimmer Inc. 3<br />

Donors and Sponsors<br />

Platinum Sponsors<br />

Geistlich Surgery, Wolhusen, CH<br />

Sanofi Biosurgery, Cambridge, USA<br />

Zimmer Inc., Austin, USA<br />

Piramal Healthcare Ltd., <strong>Montreal</strong>, <strong>Canada</strong><br />

Smith & Nephew Inc., Andover, USA<br />

Silver Sponsor<br />

Tigenix, Leuven, BE<br />

Other Sponsors<br />

Anika Therapeutics, Inc., Bedford, USA<br />

DJO <strong>International</strong>, Guildford, UK<br />

Exhibitors (Numerical Order)<br />

Company Booth Nr.<br />

Tigenix 1<br />

DJO Global 2<br />

Zimmer Inc. 3<br />

Smith & Nephew Inc. 4<br />

Croma-Pharma GesmbH 5<br />

Sanofi Genzyme Biosurgery 6<br />

Piramal Healthcare Ltd. 7<br />

Geistlich Pharma AG 8<br />

SAGE Publications 9<br />

Moximed, Inc. 10<br />

Fin-Ceramica Faenza S.p.A 11<br />

SBM Inc. Science & Bio Materials 12<br />

Arthrex, Inc. 13<br />

Bacterin 14<br />

Harvest Technologies 14A<br />

Anika Therapeutics Inc. 15<br />

Regentis Biomaterials LTD 16<br />

REGEN Lab 17<br />

Biomomentum Inc 18<br />

ArthroCare 19<br />

BioPoly 20<br />

Knee Creations, LLC 22<br />

DePuy Mitek 23<br />

Histogenics 25<br />

BioTissue AG 26


Situation Plan<br />

47


48<br />

Exhibitor’s Guide (a <strong>–</strong> z)<br />

Anika Therapeutics Inc. Booth Nr. 15<br />

32 Wiggins Avenue<br />

Bedford, MA 01730, USA<br />

Phone +1 781 457 9000<br />

jointhealth@anikatherapeutics.com<br />

www.anikatherapeutics.com<br />

Anika Therapeutics is a pioneer in developing therapeutic<br />

products for tissue protection, healing and repair. These<br />

products are based on hyaluronic acid (HA), a naturally<br />

occurring polymer found throughout the body. With the<br />

recent acquisition of Fidia Advanced Biopolymers, S.r.l.,<br />

Anika has expanded its range of orthopedic HA products<br />

from pain relief to cartilage repair and regeneration.<br />

Hyalograft ® C autograft is the first bio-engineered cartilage<br />

for minimally invasive surgical procedures. More than<br />

5,000 patients have been treated to date with Hyalograft<br />

C autograft. Hyalofast is a HA matrix for arthroscopic<br />

use in single step cartilage repair procedures. It is highly<br />

conformable and can be used without fixation in most<br />

cases.<br />

Anika’s portfolio of viscosupplements includes MONO-<br />

VISC, ORTHOVISC ® and ORTHOVISC ® mini. MONOVISC<br />

is formulated specifically for treatment with a singleinjection.<br />

FDA approved ORTHOVISC is a three injection<br />

regimen, and ORTHOVISC mini is designed to treat small<br />

joints.<br />

To learn more about Anika’s cartilage regeneration products,<br />

please visit us at Booth 15.<br />

Arthrex Inc. Booth Nr. 13<br />

1370 Creekside Blvd.<br />

Naples, FL 34108<br />

www.arthrex.com<br />

Please stop by booth #13 for a hands-on demonstration<br />

using our new iBalance HTO & UKA Systems, BioMatrix<br />

<strong>Cartilage</strong> <strong>Repair</strong> Device and ArthroFlex Systems, just to<br />

name a few.<br />

ArthroCare Corporation Booth Nr. 21<br />

Sports Medicine<br />

Austin, TX 78735, USA<br />

www.arthrocare.com<br />

Bacterin <strong>International</strong> Holdings Inc. Booth Nr. 14<br />

732 Cruiser Lane<br />

Belgrade, MT 59714<br />

Phone: 406.388.0480<br />

Fax: 406.388.0915<br />

mkoch@bacterin.com<br />

www.bacterin.com<br />

Bacterin develops innovative biologics designed for specific<br />

surgical procedures. Proprietary scaffolds, such as<br />

OsteoSponge ® and OsteoSelect ® DBM Putty, provide superior<br />

handling characteristics and osteoinductive properties<br />

for optimal surgical outcomes.<br />

Biomomentum Inc. Booth Nr. 18<br />

970 Michelin, Suite 200<br />

Laval, Quebec, H7L 5C1, <strong>Canada</strong><br />

Tel: (450) 667-2299<br />

info@biomomentum.com<br />

www.biomomentum.com<br />

Biomomentum specializes in providing solutions for the<br />

biomechanical evaluation of biomaterials and cartilage.<br />

The Arthro-BST is a hand-held medical device used in<br />

conjunction with arthroscopic procedures for the nondestructive<br />

measurement of compression-induced streaming<br />

potentials of articular cartilage. The instrument<br />

measures streaming potentials generated during gentle<br />

compression of the articular cartilage and calculates a<br />

quantitative parameter reflecting its electromechanical<br />

properties. Scientific literature has indicated that streaming<br />

potentials are not only a function of the stiffness<br />

of cartilage, but also of its composition, structure, and<br />

thickness. In addition to its clinical applications, the Arthro-BST<br />

offers a vast array of research opportunities<br />

in the field of cartilage repair.<br />

The company also commercializes the Mach-1, a modular,<br />

multiple-axis mechanical tester capable of performing<br />

compression, tension, shear, and torsion tests for<br />

the precise characterization and mechanical stimulation<br />

of cartilage and other soft tissue or materials. Biomomentum<br />

also offers biomechanical testing services using<br />

its unique instrumentation.


Exhibitor’s Guide (a <strong>–</strong> z)<br />

BioPoly, LLC Booth Nr. 20<br />

a Schwartz Biomedical Company<br />

3201 Stellhorn Rd.<br />

Fort Wayne, Indiana 46815, USA<br />

Phone: 260-399-1694<br />

Fax: 260-492-0452<br />

info@BioPolyortho.com<br />

www.BioPolyortho.com<br />

BioPoly, LLC is committed to advancing materials and<br />

device designs to address orthopaedic problems. The<br />

BioPoly ® RS Knee System is the leading product in the<br />

company’s pipeline. This product is a family of femoral<br />

condyle partial resurfacing devices uniquely designed<br />

to restore the knee’s articulating surfaces. Made from a<br />

patented microcomposite of Hyaluronic Acid and UHM-<br />

WPE, the BioPoly ® RS Knee implant provides a smooth,<br />

hydrophilic and lubricous surface to interface favorably<br />

with native joint tissues while at the same time supporting<br />

anatomical loads. Stop by our booth to learn more<br />

about this new technology and other applications under<br />

development.<br />

BioTissue AG Booth Nr. 26<br />

Seefeldstrasse 279 A<br />

CH-8008 Zurich, Switzerland<br />

Phone: +41 43 222 4004<br />

customerservice@biotissue.ch<br />

www.biotissue.ch<br />

For almost 15 years BioTissue has been the pioneer par<br />

excellence in the field of cartilage regeneration treatments.<br />

BioTissue combines state of the art applied science<br />

with the growing demands of orthopaedic treatment<br />

methods. This is made possible by the BioTissue´s<br />

experienced and visional R&D team, a hi-tech GMP Lab<br />

and by the continuous scientific exchange with clinicians.<br />

This year´s booth is dedicated to chondrotissue ® , the<br />

cell-free implant for joint cartilage repair.<br />

• chondrotissue ® is used in a one-step procedure in<br />

combination with microfracturing.<br />

• chondrotissue ® ´s capacity to form highly advanced<br />

hyaline-like cartilage tissue has been proven in ex-<br />

tensive preclinical and clinical studies, confirmed by<br />

detailed histological stainings.<br />

• chondrotissue ® has unique shape and mechanical sta-<br />

bility, allowing a stable subchondral fixation with nails<br />

and anchors.<br />

Croma-Pharma GmbH Booth Nr. 5<br />

Stockerauerstr. 181<br />

2100 Korneuburg, Austria<br />

Phone: +43/2262/684 68-0<br />

Fax +43/2262/ 68468-4<br />

www.croma.at<br />

Contact person: Mr. Arthur Fleischmann<br />

Croma-Pharma, an internationally operating pharmaceutical<br />

company headquartered in Austria, is a leading<br />

manufacturer of sterilized ready-to-use syringes of viscoelastic<br />

hyaluronic acid for intra-articular use. Full dedication<br />

to and achievement of highest quality is guaranteed.<br />

DePuy Mitek Booth Nr. 23<br />

a Johnson & Johnson Company<br />

325 Paramount Drive<br />

Raynham, MA 02767, USA<br />

www.depuymitek.com<br />

DePuy Mitek, Inc., a Johnson & Johnson company, is the<br />

leading developer and manufacturer of innovative surgical<br />

sports medicine and soft tissue repair devices. The<br />

company offers a vast array of surgical solutions to assist<br />

in addressing the challenges of soft tissue repair.<br />

Through the on-going process of improving upon and<br />

creating new, technologically advanced materials, instruments<br />

and techniques, DePuy Mitek looks for continued<br />

growth in developing the means to advance procedural<br />

solutions in the field of sports medicine.<br />

DJO Global Inc. Booth Nr. 2<br />

1430 Decision St<br />

Vista, CA 92081, USA<br />

Phone: 760.734.3125<br />

Fax: 760.734.3595<br />

www.djoglobal.com<br />

DJO is a leading global developer, manufacturer and<br />

distributor of high-quality medical devices that provide<br />

solutions for musculoskeletal health, vascular health<br />

and pain management. The Company’s products address<br />

the continuum of patient care from injury prevention to<br />

rehabilitation after surgery, injury or from degenerative<br />

disease.<br />

Our products are used by orthopedic specialists, spine<br />

surgeons, primary care physicians, pain management<br />

49


50<br />

Exhibitor’s Guide (a <strong>–</strong> z)<br />

specialists, physical therapists, podiatrists, chiropractors,<br />

athletic trainers and other healthcare professionals.<br />

In addition, many of the Company’s medical devices and<br />

related accessories are used by athletes and patients for<br />

injury prevention and at-home physical therapy treatment.<br />

The Company’s product lines include rigid and soft orthopedic<br />

bracing, hot and cold therapy, bone growth stimulators,<br />

vascular systems, electrical stimulators used for<br />

pain management and physical therapy products. The<br />

Company’s surgical division offers a comprehensive suite<br />

of reconstructive joint products for the hip, knee and<br />

shoulder. DJO’s products are marketed under the brands<br />

Aircast ® , DonJoy ® , ProCare ® , CMF, Empi ® , Saunders ® ,<br />

Chattanooga Group, DJO Surgical, Cefar ® - Compex ®<br />

and Ormed ® . For additional information on the Company,<br />

please visit www.DJOglobal.com.<br />

FIN-CERAMICA Faenza S.p.A Booth Nr. 11<br />

Via Granarolo 177/3<br />

48018 Faenza (RA) ITALY<br />

www.finceramica.com<br />

ddonati@finceramica.it<br />

www.finceramica.it<br />

Finceramica develops and produces propietary bone and<br />

cartilage substitutes, including custom-made solutions<br />

for particular clinical needs. Research activity focuses on<br />

innovative bioceramic materials, ceramic-polymer composites<br />

and a new generation of biologial joint replacements<br />

and knee resurfacings.<br />

Geistlich Pharma AG Booth Nr. 8<br />

Business Unit Surgery<br />

Bahnhofstrasse 40<br />

6110 Wolhusen, Switzerland<br />

Phone: +41 41 492 55 55<br />

Fax: +41 41 492 56 39<br />

surgery@geistlich.com<br />

www.geistlich-surgery.com<br />

Geistlich Surgery develops premier global solutions for<br />

the formation of bone and cartilage in the rapidly advancing<br />

field of regenerative medicine.<br />

Chondro-Gide ® is the leading natural collagen matrix in<br />

cartilage regeneration. This standardised, easy to handle<br />

matrix can be used to treat cartilage defects using the<br />

innovative AMIC ® technique or using ACI.<br />

AMIC ® is a single-step, cost efficient and effective procedure<br />

for treating traumatic cartilage defects. Chondro-<br />

Gide ® provides a suitable cell carrier and positively influences<br />

chondrogenic differentiation of mesenchymal<br />

stem cells to form a mostly hyaline-like cartilaginous<br />

repair tissue.<br />

Orthoss ® is a natural bone graft substitute. Its inorganic<br />

bone matrix has a macro- and microporous structure similar<br />

to human spongeous bone. It is structurally integrated<br />

into the surrounding bone and incorporated into the<br />

natural remodelling process. As a result of the excellent<br />

biofunctionality, Orthoss ® is an ideal bone graft substitute<br />

which can be used alone or during composite bone<br />

grafting using autologous bone or bone marrow aspirate<br />

when treating large defects. This includes the repair of<br />

defects following trauma, reconstruction in orthopaedics<br />

and in spinal surgery. Over 25 years of clinical experience<br />

show a high degree of safety and efficacy.<br />

Harvest Technologies Corp. Booth Nr. 14a<br />

40 Grissom Road, Suite 100<br />

Plymouth, MA 02360, USA<br />

Phone: 508-732-7500<br />

Toll Free (U.S.): +1-877-842-7837<br />

Fax: 508-732-0400<br />

info@harvesttech.com<br />

www.harvesttech.com<br />

Developing Technologies for Accelerating Healing, NaturallyÒ,<br />

Harvest manufactures the SmartPRePÒ BMACÒ<br />

(Bone Marrow Aspirate Concentrate) System for Concentrating<br />

Autologous Adult Stem Cells and the SmartPReP<br />

APC+Ò System for Concentrating Platelets.<br />

Harvest’s BMACÒ System is the first technology that rapidly<br />

concentrates clinically significant amounts of stem<br />

and precursor cells from a small aspirate of autologous<br />

bone marrow at patient point-of-care. As little as 30-60ml<br />

of bone marrow aspirate can be concentrated at point of<br />

care in just 15 minutes. The SmartPReP BMAC2 series<br />

efficiently concentrates and recovers regenerative cells<br />

while retaining their cellular viability and proliferative<br />

potential. With minimal processing time, concentrated<br />

autologous adult stem cells are now available to the clinician<br />

at patient point-of-care.<br />

Harvest’s APC+Ò System for concentrating platelets<br />

starts with only 20ml of patient blood and produces<br />

a platelet concentrate enriched with multiple growth


Exhibitor’s Guide (a <strong>–</strong> z)<br />

factors and white blood cells, at 4X or greater above<br />

baseline--the clinical requirement needed to accelerate<br />

wound healing.<br />

The SmartPReP BMAC and APC+ Systems make concentrated<br />

autologous adult stem cells and concentrated platelets<br />

available to the clinician as needed, with minimal<br />

processing time, at patient point-of-care.<br />

Histogenics Corp. Booth Nr. 25<br />

830 Winter Street<br />

Waltham, MA 02451, USA<br />

www.histogenics.com<br />

Histogenics is a leading regenerative medicine company<br />

that combines cell therapy and tissue engineering technologies<br />

to develop highly innovative products for tissue<br />

repair and regeneration. The company’s flagship products<br />

focus on the treatment of active patients suffering<br />

from articular cartilage derived pain and immobility.<br />

Histogenics has developed technology and products<br />

to reverse or prevent cartilage damage, regenerating<br />

healthy hyaline cartilage tissue. NeoCart ® , autologous<br />

engineered neocartilage grown outside the body using<br />

the patient’s own cells, has been developed for regeneration<br />

of full thickness cartilage lesions. The VeriCart<br />

single-step cartilage matrix is a cell-free matrix designed<br />

to be used in conjunction with marrow-stimulating procedures,<br />

or alternatively, reconstituted with the patient’s<br />

bone marrow, thereby initiating the repair process. The<br />

goal: solutions upstream to Osteoarthritis that restore<br />

patients to an active lifestyle that can last all life long.<br />

Knee Creations Booth Nr. 22<br />

West Chester, PA, USA<br />

customerservice@kneecreations.com<br />

www.kneecreations.com<br />

Knee Creations develops innovative bone-based technologies,<br />

procedures, and instrumentation. Knee Creations<br />

pioneered the Subchondroplasty (SCP ® ) procedure, a<br />

minimally invasive treatment of defects associated with<br />

bone marrow edema. Bone marrow edema is a known,<br />

painful element of osteoarthritis (OA), a disease which affects<br />

one in two Americans. SCP ® effectively targets bone<br />

defects that are not addressed by current arthroscopic<br />

and conservative care treatments.<br />

Moximed ® Inc. Booth Nr. 10<br />

26460 Corporate Ave, Suite 100<br />

Hayward, CA 94545 USA<br />

Phone: +1 510 887 3300<br />

Fax: +1 510 887 3499<br />

www.moximed.com<br />

info@moximed.com<br />

Moximed ® , Inc. is dedicated to improving the standard of<br />

care for patients with osteoarthritis (OA) and is initially<br />

focused on developing joint sparing solutions for patients<br />

with knee OA.<br />

The KineSpring ® System is an implantable load absorber<br />

designed to relieve pain and improve function in patients<br />

suffering from medial knee OA and who might be<br />

too young, too active, or otherwise disinclined for HTO<br />

or UKA. The concept of joint unloading is a proven treatment:<br />

orthotics, braces, and HTO can relieve OA pain,<br />

and recent studies indicate that reducing joint load can<br />

potentially delay OA progression or regenerate damaged<br />

cartilage.<br />

The KineSpring System consists of an implantable, extracapsular<br />

load absorber and femoral and tibial base components.<br />

The device is implanted in a standard orthopaedic<br />

procedure that does not require any bone, ligament,<br />

or cartilage removal. The extra-capsular positioning and<br />

structure preserving technique creates an essentially reversible<br />

procedure.<br />

The KineSpring System has been evaluated by leading<br />

surgeons in multiple, international clinical studies over<br />

the past four years. It has been granted CE Mark and is<br />

currently available for sale in select centers in Europe<br />

and Australia.<br />

To learn more about Moximed and the KineSpring System,<br />

please visit us at Stand 10.<br />

51


52<br />

Exhibitor’s Guide (a <strong>–</strong> z)<br />

Piramal Healthcare Ltd Booth Nr. 7<br />

475 Boulevard Armand Frappier<br />

Laval, <strong>Canada</strong><br />

www.piramal.com<br />

Piramal Healthcare Ltd, a Piramal Group company, is a<br />

globally integrated healthcare company with a diverse<br />

product portfolio spanning several therapeutic areas,<br />

and a growing commitment in Bio-Orthopedics. Piramal<br />

Healthcare Ltd is one of the largest custom healthcare<br />

manufacturing companies with a footprint across North<br />

America, Europe and Asia.<br />

Since 2005, Piramal Healthcare has devoted itself to<br />

bio-orthopedic research through strategic investments<br />

in Canadian innovations such as BST-CarGel ® , a novel bioscaffold<br />

for advanced cartilage regeneration which was<br />

shown clinically to result in consistently higher quality<br />

cartilage repair while at the same time being economically<br />

viable within challenging reimbursement healthcare<br />

systems. Being introduced to the European and Canadian<br />

orthopedic communities in 2012, BST-CarGel ® offers a<br />

new standard in cartilage regeneration.<br />

At Piramal Healthcare, our core values of Knowledge, Action<br />

and Care propel us to improve the quality of lives by<br />

democratizing healthcare. We aim to attain leadership in<br />

market share and innovation by:<br />

• Partnering with the medical fraternity<br />

• Building strong capabilities to deliver product and process<br />

innovations<br />

• Attracting and developing the best in class talent<br />

Piramal Healthcare, a proud Platinum sponsor of ICRS 2012<br />

Regen Lab Booth Nr. 17<br />

En Budron B2<br />

CH - 1052 Le Mont-sur-Lausanne<br />

Switzerland<br />

Phone: +41 (0)21 864 01 11<br />

Fax +41 (0)21 864 01 10<br />

www.regenlab.com<br />

Regen Lab is recognized as the leading provider of patented<br />

technologies to produce Autologous Platelet Rich<br />

Plasma (A-PRP) and other cell concentrates. The technology<br />

is processing the patient´s own blood, fat or bone<br />

marrow to use the concentrated cells for indications in<br />

various medical disciplines such as sports medicine,<br />

wound healing and aesthetics. Regen Lab is an independent<br />

biotech company from Lausanne (Switzerland)<br />

that has been established in 2003 and is marketing its<br />

products worldwide. Its Medical Devices for the close<br />

circuit preparation of Autologous Platelet Rich Plasma<br />

are known under the RegenACR ® and RegenKit ® brand<br />

names.<br />

Regentis Biomaterials Ltd. Booth Nr. 16<br />

2 Ha'Ilan Street, Northern Industrial Zone<br />

P.O.Box 260, Or-Akiva 30600, Israel<br />

Livnat@regentis.co.il<br />

www.regentis.co.il<br />

Regentis Biomaterials is a tissue repair company developing<br />

innovative biodegradable hydrogels for local repair<br />

of soft and hard tissue. Our platform technology is<br />

a family of hydrogels called Gelrin. These gels can be<br />

injected or applied to a specific local site and offer beneficial<br />

properties for the local repair of damaged tissue<br />

such as cartilage and bone.<br />

The Gelrin technology offers off-the-shelf products<br />

that are designed to be suitable for both open surgery<br />

and minimally invasive procedures. An ideal solution for<br />

physicians and their patients, the products are easy to<br />

implant and have been shown to stimulate the regeneration<br />

of healthy cartilage and bone tissue.<br />

Regentis’ first orthopedic product is GelrinC, a biodegradable<br />

hydrogel for articular cartilage regeneration, providing<br />

a controlled environment for gradual tissue repair<br />

and formation of hyaline-like cartilage. GelrinC is comprised<br />

of polyethylene glycol diacrylate (PEG-DA) and<br />

denatured fibrinogen, a natural substrate for tissue regeneration.<br />

These materials form a matrix for tissue repair<br />

<strong>–</strong> combining the stability and versatility of a synthetic<br />

material with the bio-functionality of a natural material.<br />

CAUTION: GelrinC is an investigational product, not<br />

available in Europe and in the US


Exhibitor’s Guide (a <strong>–</strong> z)<br />

SAGE Booth Nr. 9<br />

2455 Teller Road<br />

Thousand Oaks, CA 91320, USA<br />

lisa.lamont@sagepub.com<br />

www.sagepub.com<br />

SAGE is a leading international publisher of journals,<br />

books, and electronic media for academic, educational,<br />

and professional markets. Since 1965, SAGE has helped<br />

educate a global community spanning a wide range of<br />

subject areas including business, humanities, social sciences,<br />

and science, technology, and medicine. Visit us at<br />

www.sagepub.com.<br />

Sanofi Biosurgery Booth Nr. 6<br />

55 Cambridge Parkway<br />

Cambridge, MA 02142, USA<br />

www.genzyme.com<br />

Sanofi Biosurgery is a global strategic business unit of<br />

Sanofi. It develops and markets innovative, biologically<br />

based products for osteoarthritis relief, adhesion prevention,<br />

cartilage repair, and severe burn treatment.<br />

Sanofi Biosurgery’s products include: Synvisc ® , Synvisc-<br />

One (hylan G-F 20), Carticel ® (autologous cultured<br />

chondrocytes), MACI ® (Matrix-induced Autologous Chondrocyte<br />

Implantation), Seprafilm ® and Epicel ® (cultured<br />

epidermal autografts). Sanofi Biosurgery is committed<br />

to transforming disease management through innovative<br />

medical interventions.<br />

About Sanofi<br />

Sanofi, a global and diversified healthcare leader, discovers,<br />

develops and distributes therapeutic solutions<br />

focused on patients’ needs. Sanofi has core strengths in<br />

the field of healthcare with seven growth platforms: diabetes<br />

solutions, human vaccines, innovative drugs, consumer<br />

healthcare, emerging markets, animal health and<br />

the new Genzyme. Sanofi is listed in Paris (EURONEXT:<br />

SAN) and in New York (NYSE: SNY).<br />

SBM Inc. Booth Nr. 12<br />

19 Hancock St<br />

Winchester, MA, USA<br />

gboue@s-b-m.us<br />

www.s-b-m.us<br />

Since 1991, SBM innovates in the field of orthopaedic biosurgery<br />

by working closely with renowned surgeons to<br />

design and manufacture unique solutions based on bioresorbable<br />

materials, e.g. Bone void fillers, ACL composite<br />

fixations. In 1996, SBM has been the first company to<br />

manufacture a resorbable and load-bearing implant for<br />

Opening Wedge High Tibial Osteotomies.<br />

Today, SBM is perfecting its HTO system with a very<br />

unique locking dual-compression plate that promotes<br />

faster Bone healing, causes less post-operative pain and<br />

leads to quicker return to function.<br />

Smith & Nephew Booth Nr. 4<br />

150 Minuteman Road<br />

Andover, MA 01810, USA<br />

www.smith-nephew.com<br />

Smith & Nephew is a global medical technology business<br />

with global leadership positions in Orthopaedic<br />

Reconstruction, Advanced Wound Management, Sports<br />

Medicine, Trauma and Clinical Therapies. The Company<br />

has distribution channels, purchasing agents and buying<br />

entities in over 90 countries worldwide. Annual sales in<br />

2010 were nearly $4.0 billion.<br />

Smith & Nephew is dedicated to helping improve people's<br />

lives. The Company prides itself on the strength of its relationships<br />

with its surgeons and professional healthcare<br />

customers, with whom its name is synonymous with<br />

high standards of performance, innovation and trust.<br />

In established markets such as the United States, <strong>Canada</strong>,<br />

Europe, Japan, Australia and New Zealand we operate<br />

Advanced Surgical Devices and Advanced Would<br />

Management.<br />

53


54<br />

Exhibitor’s Guide (a <strong>–</strong> z)<br />

Our Advanced Surgical Devices Division consists of:<br />

- Orthopaedic Reconstruction in the form of hip/knee/<br />

shoulder implants<br />

- Trauma products that help heal broken bones and cor-<br />

rect deformities<br />

- Biologics Clinical Therapies including next generati-<br />

on tissue repair treatments and therapies that provide<br />

everything from bone growth stimulation to relief from<br />

back pain<br />

- Sports Medicine instruments used to access, visualize,<br />

resect and repair injuries through minimally invasive<br />

surgery (arthroscopy)<br />

Our Advanced Wound Management Division develops<br />

innovative dressings and systems that provide faster healing<br />

and infection protection for patients with chronic,<br />

hard-to-heal wounds. Our Emerging Markets Organization<br />

focuses on Brazil, Russia, India and China. Our <strong>International</strong><br />

Organization focuses on all the other countries<br />

and regions, such as South Africa, the Middle East, South<br />

East Asia and Central and Latin America<br />

Across the Group as a whole, we benefit from being a<br />

truly global company, with approximately 45% of our revenue<br />

being in the US, 30% in Europe and 25% in rest<br />

of world.<br />

TiGenix NV Booth Nr. 1<br />

Researchpark Haasrode 1724<br />

Romeinse straat 12 bus 2<br />

3001 Leuven, Belgium<br />

Phone: +32 (0) 16 39 60 60<br />

Fax: +32 (0) 16 39 79 70<br />

info@tigenix.com<br />

www.tigenix.com<br />

TiGenix NV (NYSE Euronext: TIG) is a leading European<br />

cell therapy company with two marketed products and a<br />

strong clinical stage pipeline of adult stem cell programs.<br />

The company’s lead product, ChondroCelect ® , for cartilage<br />

repair in the knee, is the first and only approved cellbased<br />

product in Europe, and is currently being launched<br />

in different European countries.<br />

TiGenix’s stem cell programs are based on a validated<br />

platform of allogeneic expanded adipose-derived stem<br />

cells (eASCs) targeting autoimmune and inflammatory<br />

diseases.<br />

Built on solid pre-clinical and CMC packages, they are<br />

being developed in close consultation with the European<br />

Medicines Agency. The company is slated to start a<br />

Phase III clinical trial in complex perianal fistulas in patients<br />

with Crohn’s disease, is conducting a Phase IIa trial<br />

in rheumatoid arthritis, and in early 2012 will initiate a<br />

Phase I trial to investigate the potential of intra-lymphatic<br />

administration of eASCs for autoimmune disorders.<br />

TiGenix is based out of Leuven (Belgium), and has operations<br />

in Madrid (Spain), Cambridge (UK) and Sittard-<br />

Geleen (the Netherlands). For more information please<br />

visit www.tigenix.com.<br />

Zimmer Inc. Booth Nr. 3<br />

9301 Amberglen Blvd<br />

Austin, Tx 78729, USA<br />

www.zimmer.com<br />

Founded in 1927 and headquartered in Warsaw, Indiana,<br />

Zimmer designs, develops, manufactures and markets<br />

orthopaedic reconstructive, spinal and trauma devices,<br />

dental implants, and related surgical products. Zimmer<br />

has operations in more than 25 countries around the<br />

world and sells products in more than 100 countries.<br />

Zimmer's 2011 sales were approximately $4.5 billion. The<br />

Company is supported by the efforts of more than 8,500<br />

employees worldwide. For more information about Zimmer,<br />

visit www.zimmer.com


10 th World Congress of the<br />

<strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong><br />

ICRS 2012<br />

May 12 <strong>–</strong> 15, 2012<br />

Agenda<br />

Scientific Programme<br />

“Advancing science & education in cartilage repair worldwide!”<br />

55


56<br />

Programme: Saturday, May 12, 2012<br />

ICRS Editorial Board Meeting Room: Chaudiere<br />

07:30 <strong>–</strong> 08:30<br />

ICRS Executive Board Meeting Room: Chaudiere<br />

09:00 <strong>–</strong> 10:30<br />

ICRS General Board Meeting Room: Chaudiere<br />

10:30 <strong>–</strong> 12:00<br />

Session 1.0 Plenary<br />

Sports Injury: ICRS - FIFA Session<br />

13:00 <strong>–</strong> 14:00 Room: Grand Salon<br />

Moderators: Bert Mandelbaum (US), Stefano Della Villa (IT)<br />

1.1 Articular <strong>Cartilage</strong> Mechanisms, Management and Outcome<br />

B. Mandelbaum, Santa Monica/US<br />

1.2 Basic science mechanisms & animal models of articular cartilage injury<br />

M.B. Hurtig, Guelph/CA<br />

1.3 <strong>Cartilage</strong> problems in the athlete: The scope of the problem<br />

K. Mithoefer, Chestnut Hill/US<br />

1.4 Injury repair options in the athlete - Special concerns<br />

B. Mandelbaum, Santa Monica/US<br />

Session 2.1 Special<br />

Growth Factors, Morphogens & Cytokines<br />

14:15 <strong>–</strong> 15:15 Room: Grand Salon<br />

Moderators: Gerjo Van Osch (NL), Maurizio Pacifici (US)<br />

2.1.1 Growth factors pathways as regulators of chondrogenesis<br />

F. Beier, London/CA<br />

2.1.2 Signaling proteins and transcription factors in joint development and degeneration<br />

M. Pacifici, M. Iwamoto, Philadelphia/US<br />

2.1.3 Growth factors in cartilage homeostasis in vitro / in vivo<br />

S. Chubinskaya, Chicago/US<br />

Session 2.2 Special<br />

Nerve Dependence on <strong>Cartilage</strong> Development, <strong>Repair</strong> and Joint Pain<br />

14:15 <strong>–</strong> 15:15 Room: Marquette<br />

Moderators: Mats Brittberg (SE), Norimasa Nakamura (JP)<br />

2.2.1 Nerve dependence and cartilage development during Urodele limb regeneration and its<br />

relevance to mammals.<br />

M. Maden, Gainesville/US


Programme: Saturday, May 12, 2012<br />

2.2.2 Neurogenic factors in the etiopathogenesis of osteoarthritis<br />

D. Walsh, Nottingham/UK<br />

2.2.3 <strong>Cartilage</strong> lesions; which lesions are painful and what is the cause of pain ?<br />

M. Brittberg, Kungsbacka/SE<br />

Session 2.3 Special<br />

Femoropatellar Joint<br />

14:15 <strong>–</strong> 15:15 Room: Jolliet<br />

Moderators: Christoph Erggelet (CH), Tim Spalding (UK)<br />

2.3.1 <strong>Cartilage</strong> defects in the femoropatellar joint<br />

B.J. Cole, Chicago/US<br />

2.3.2 Clinical decisions for cartilage repair in the femoropatellar joint<br />

T. Spalding, Coventry/UK<br />

2.3.3 Surgical treatment and results<br />

T. Minas, Chestnut Hill/US<br />

Coffee Break/Industry Exhibition<br />

15:15 <strong>–</strong> 15:45 Room: Exhibition Hall<br />

Session 3.1 Special<br />

Opportunities of Bioprinting in <strong>Cartilage</strong> Regeneration<br />

15:45 <strong>–</strong> 16:45 Room: Jolliet<br />

Moderators: Jos Malda (NL), Lawrence Bonassar (US)<br />

3.1.1 Basic concepts and potential application of tissue/organ printing<br />

D. Hutmacher, Brisbane/AU<br />

3.1.2 Potential of natural and synthetic biomaterials for bioprinting<br />

F.P.W. Melchels 1, 2 , W.J.A. Dhert 2 , D. Hutmacher 1 , J. Malda 1, 2 , 1 Kelvin Grove/AU, 2 Utrecht/NL<br />

3.1.3 Bioprinting for osteochondral repair<br />

L. Bonassar, Ithaca/US<br />

Session 3.2 Special<br />

Medication and <strong>Cartilage</strong><br />

15:45 <strong>–</strong> 16:45 Room: Marquette<br />

Moderators: John Tarlton (UK), Wayne McIlwraith (US)<br />

3.2.1 Effect of medications on articular cartilage health and repair<br />

W. McIlwraith, Fort Collins/US<br />

57


58<br />

Programme: Saturday, May 12, 2012<br />

3.2.2 In vitro models for development and screening of cartilage repair strategies<br />

G.J.V.M. Van Osch, Rotterdam/<br />

3.2.3 Nutraceuticals & Osteoarthritis: Do omega-3 fatty acids, glucosamine & chondroitin sulphates<br />

have chondro-protective actions?<br />

B. Caterson, Cardiff/UK<br />

Session 3.3 Special<br />

Meniscus<br />

15:45 <strong>–</strong> 16:45 Room: St. François<br />

Moderators: Peter Verdonk (BE), Andreas Gomoll (US)<br />

3.3.1 Can meniscus reconstruction prevent or even cure OA?<br />

W. Gersoff, Denver/US<br />

3.3.2 Clinical options for meniscus reconstruction and regeneration<br />

R. Jakob, Fribourg/CH<br />

3.3.3 How critical is the meniscus to the pathology of OA?<br />

P. Lavigne, <strong>Montreal</strong>/CA<br />

Session 4.0 Plenary<br />

Opening Ceremony & Award Session<br />

17:00 <strong>–</strong> 17:45 Room: Grand Salon<br />

Session 5.0 Plenary<br />

Honorary Lectures<br />

17:45 <strong>–</strong> 18:45 Room: Grand Salon<br />

5.1 Advances in understanding of post-traumatic osteoarthritis <strong>–</strong> Implications for treatment of joint injuries<br />

J. Buckwalter, Iowa City/US<br />

5.2 <strong>Cartilage</strong> <strong>Repair</strong>: Where are we now? Where are we going?<br />

D. Grande, Manhasset/US<br />

Welcome Reception Exhibition Hall<br />

19:00 <strong>–</strong> 20:30


Programme: Sunday, May 13, 2012<br />

Session 6.1 Instructional Course - Imaging of <strong>Cartilage</strong> Defects<br />

07:30 <strong>–</strong> 08:15 Room: Jolliet<br />

Moderators: Goetz Welsch (DE), Sebastian Apprich (AT)<br />

Session 6.2 Meet the Experts (Basic Scientists)<br />

07:30 <strong>–</strong> 08:15 Room: Marquette<br />

Moderators: Sally Roberts (UK), Bruce Caterson (UK)<br />

Session 7.0 Plenary<br />

Clinical Studies using <strong>Cartilage</strong> Fragments<br />

08:30 <strong>–</strong> 09:30 Room: Grand Salon<br />

Moderators: Anthony Hollander (UK), Allan Gross (CA)<br />

7.1 The use of autologous cartilage fragments <strong>–</strong> clinical studies<br />

J. Farr, Greenwood/US<br />

7.2 The use of allogenic cartilage fragments <strong>–</strong> clinical studies<br />

D. Caborn, Louisville/US<br />

Session 8.1 Special<br />

<strong>Cartilage</strong> <strong>Repair</strong> in the Foot and Ankle Joint<br />

09:45 <strong>–</strong> 10:45 Room: Marquette<br />

Moderators: James Richardson (UK), Roland Jakob (CH)<br />

8.1.1 Treatment of Osteochondral Lesions of the Talus<br />

E. Giza, Sacramento/US<br />

8.1.2 Clinical options for cartilage reconstruction in the ankle.<br />

R. Ferkel, Van Nuys/US<br />

8.1.3 Dermal graft application in Hallux Limitus / Rigidus for salvage of the 1st metatarsophallangeal joint<br />

L.J. Sanchez, Orlando/US<br />

Session 8.2 Special<br />

Culture Techniques for Controlling Cell Differentiation<br />

09:45 <strong>–</strong> 10:45 Room: Grand Salon<br />

Moderators: Jan Schageman (US), Tim Welting (NL)<br />

8.2.1 In vitro culture of chondrocytes - what are the limitations?<br />

B. Johnstone, Portland/US<br />

8.2.2 Expansion of chondrocyte populations on high extension culture surfaces for improved<br />

retention of phenotype<br />

T.M. Quinn 1 , B. Hinz 2 , M. Matmati 1 , D.H. Rosenzweig 1 , 1 <strong>Montreal</strong>/CA, 2 Toronto/CA<br />

8.2.3 2D and 3D cultures of chondrocytes<br />

T.J. Klein, Kelvin Grove/AU<br />

59


60<br />

Programme: Sunday, May 13, 2012<br />

Session 8.3 Special<br />

Intervertebral Disc<br />

09:45 <strong>–</strong> 10:45 Room: St. François<br />

Moderators: Rita Kandel (CA), Susan Chubinskaya (US)<br />

8.3.1 Intervertebral Disc Tissue Engineering: Is it ready for weight bearing?<br />

R. Kandel, Toronto/CA<br />

8.3.2 Current clinical treatment strategies and future concepts<br />

S. Daisuke, Kanagawa, Isehara/JP<br />

8.3.3 Materials for intervertebral disc tissue engineering<br />

R. Mauck, M.B. Fisher, D.M. Elliott, Philadelphia/US<br />

Coffee Break / Industry Exhibition<br />

10:45 <strong>–</strong> 11:15 Room: Exhibition Hall<br />

Session 9.1 Special<br />

ICRS <strong>–</strong> ISAKOS Symposium "Joint Preservation"<br />

11:15 <strong>–</strong> 12:45 Room: Grand Salon<br />

Moderators: Norimasa Nakamura (JP), Rocky Tuan (US)<br />

9.1.1 <strong>Cartilage</strong> repair to prevent osteoarthritis. <strong>–</strong> Fact or fiction?<br />

T.S. De Windt, D.B.F. Saris, Utrecht/NL<br />

9.1.2 What is the key pathway to prevent post-traumatic arthritis for future molecule-based therapy?<br />

S. Chubinskaya, Chicago/US<br />

9.1.3 ACL reconstruction and osteoarthritis: Evidence from long-term follow-up and potential solutions<br />

R.A. Magnussen 1 , V. Duthon, E. Servien 2 , P. Neyret 2 , 1 Columbus/US, 2 Lyon Cedex 04/FR<br />

9.1.4 Anatomic ACL Reconstruction <strong>–</strong> Current concept and future perspective<br />

F.H. Fu, Pittsburgh/US<br />

Session 9.2 Free Papers<br />

Chondrocytes 1<br />

11:15 <strong>–</strong> 12:45 Room: Marquette<br />

Moderators: Stefan Marlovits (AT), William Bugbee (US)<br />

9.2.1 Overview & Introduction<br />

A. Nixon, Ithaca/US<br />

9.2.2 Redifferentiation of Human Articular Chondrocytes in 2D versus 3D culture<br />

M.M.J. Caron, P. Emans, M.M.E. Coolsen, L. Voss, D.A.M. Surtel, L.W. Van Rhijn,<br />

T.J.M. Welting, Maastricht/NL<br />

9.2.3 Effect of TGF <strong>–</strong> on Sox9 Function and Chondrocyte Phenotype<br />

J. Perez 1 , H. Seo 2 , R. Serra 1 , 1 Birmingham/US, 2 Vancouver/CA


Programme: Sunday, May 13, 2012<br />

9.2.4 Development of an intracellular flow cytometry assay of S100 for quantitative assessment of<br />

chondrogenic potency<br />

J. Diaz Romero, A. Quintin, D. Nesic, Bern/CH<br />

9.2.5 An Unique Metabolic Profile of Human Acetabular Labrum Cells.<br />

A.A.M. Dhollander 1 , S. Lambrecht 2 , P.C. Verdonk 1 , E.A. Audenaert 1 , K.F. Almqvist 1 , C. Pattyn 1 ,<br />

R. Verdonk 1 , D. Elewaut 2 , G. Verbruggen 2 , 1 Ghent/BE, 2 Gent/BE<br />

9.2.6 Advances in the Development of Engineered Ear<br />

M.A. Randolph, A. Tseng, I. Pomerantseva, E. Bassett, J. Vacanti, C.A. Sundback, Boston/US<br />

9.2.7 Monitoring COMP Expression during Articular Chondrocyte Compression<br />

D. Amanatullah 1 , J. Lu 1 , J. Yik 1 , J. Hecht 2 , K. Posey 2 , P. Di Cesare 1 , D. Haudenschild 1 ,<br />

1 Sacramento/US, 2 Houston/US<br />

9.2.8 OA-chondrons outperform OA-chondrocytes in late-stage OA: relevance for biological therapy?<br />

M. Rothdiener 1 , Q. Wang 1 , T. Uynuk-Ool 1 , T. Felka 1 , B.G. Ochs 1 , C. Bahrs 1 , U. Stoeckle 1 ,<br />

A.J. Grodzinsky 2 , W.K. Aicher 1 , B. Rolauffs 1 , 1 Tuebingen/DE, 2 Cambridge/US<br />

Session 9.3 Free Papers<br />

Biomaterials & Scaffolds 1<br />

11:15 <strong>–</strong> 12:45 Room: St. François<br />

Moderators: Tim Woodfield (NZ), Jeremy Mao (US)<br />

9.3.1 Overview & Introduction<br />

J. Mao, New York/US<br />

9.3.2 Low oxygen tension increases the tensile properties of engineered articular cartilage through<br />

enzyme mediated collagen cross-linking.<br />

E.A. Makris, K. Athanasiou, Davis/US<br />

9.3.3 Hypoxia Promotes Phenotype Preservation of Human Chondrocytes Embedded within<br />

Collagen Matrices<br />

J. Sernik, R. Croutze, C.C. Secretan, L. Laouar, N.M. Jomha, A.B. Adesida, Edmonton/CA<br />

9.3.4 Mesenchymal Stem Cells Show Improved <strong>Cartilage</strong> Formation on Extracellular <strong>Cartilage</strong><br />

Matrix-derived Scaffolds for <strong>Cartilage</strong> Defect <strong>Repair</strong><br />

K.E.M. Benders 1 , W. Boot 1 , P.R. Van Weeren 1 , D.B.F. Saris 1 , 2 , W.J.A. Dhert 1 , J. Malda 1 , 1 Utrecht/NL,<br />

2 Enschede/NL<br />

9.3.5 Natural polymer blends processed using ionic liquids can initiate chondrogenic differentiation<br />

of mesenchymal stem cells<br />

N. Singh 1 , S.S. Rahatekar 1 , K.K. Koziol 2 , A.J. Patil 1 , T.H.S. Ng 1 , S. Mann 1 , A.P. Hollander 1 ,<br />

W. Kafienah 1 , 1 Bristol/UK, 2 Cambridge/UK<br />

9.3.6 Chitosan modulates the ability of U937 macrophage-like cells to secrete chemokines and<br />

attract human bone marrow-derived mesenchymal stem cells<br />

D. Fong, M.B. Ariganello, S. Bolduc Beaudoin, C.D. Hoemann, <strong>Montreal</strong>/CA<br />

61


62<br />

Programme: Sunday, May 13, 2012<br />

9.3.7 Use of SDF and sphingosine impregnated scaffolds in addressing isolated cartilaginous defects<br />

of the knee: can we stimulate hyaline cartilage regeneration?<br />

N. Chinitz 1 , A. Catanzano 1 , P. Razzano 2 , N.V. Shah 2 , Z. Klapholz 2 , N.A. Sgaglione 2 , D. Grande 2 ,<br />

1 New Hyde Park/US, 2 Manhasset/US<br />

9.3.8 Scaffold and cell combination influence chondrogenesis in 3D coculture of chondrocytes with<br />

mononuclear fraction cells<br />

J.E.J. Bekkers, A.I. Tsuchida, A. Kolk, W.J.A. Dhert, D.B.F. Saris, L.B. Creemers, Utrecht/NL<br />

9.3.9 A Self-Setting Hydrogel Mechanically reinforced with a marine polysaccharide as a Scaffold for<br />

<strong>Cartilage</strong> Tissue Engineering<br />

E. Rederstorff, C. Vinatier, S. Sourice, M. Masson, S. Colliec-Jouault, B.H. Fellah, P. Weiss,<br />

J. Guicheux, Nantes/FR<br />

Session 9.4 Free Papers<br />

Other Musculoskeletal Tissues<br />

11:15 <strong>–</strong> 12:45 Room: Jolliet<br />

Moderators: Stefan Nehrer (AT), Sally Roberts (UK)<br />

9.4.1 Decreased hypertrophic differentiation accompanies enhanced matrix formation in cocultures<br />

of outer meniscus cells with bone marrow mesenchymal stromal cells<br />

D. Saliken, A. Sierra-Mulet, N.M. Jomha, A. Adesida, Edmonton/CA<br />

9.4.2 The effect of arthroscopic partial medial meniscectomy on tibiofemoral stability<br />

S. Arno, S. Hadley, K.A. Campbell, C. Bell, M. Hall, L. Beltran, M.P. Recht, O. Sherman, P. Walker,<br />

New York/US<br />

9.4.3 Microarray analysis of human meniscus cell dedifferentiation<br />

S.P. Grogan, S.F. Duffy, C. Pauli, S. Das, M.K. Lotz, D.D. D'Lima, La Jolla/US<br />

9.4.4 Clinical and Immunoassay evaluation in meniscal allograft transplant at eighteen months follow up.<br />

F. Cruz 1 , M. Acuña 1 , 2 , F.E. Villalobos Córdova 1 , C. Ibarra 1 , A. Almazan 1 , F. Pérez 1 , L. Sierra 1 ,<br />

A. Izaguirre 1 , 1 Mexico City/MX, 2 Mexico/MX<br />

9.4.5 MRI mid-term outcome after partial meniscus substitution using the collagen meniscal implant (CMI)<br />

M.T. Hirschmann 1 , M.P. Arnold 1 , R. Berbig 2 , U. Luethi 3 , 1 Bruderholz/CH, 2 Zuerich/CH, 3 Zürich/CH<br />

9.4.6 Comparison between arthroscopic meniscal allograft transplantation and polyurethane scaffold<br />

implantation: Clinical outcome, reinterventions and implant intergrity after a minimum followup<br />

period of 2 years. A prospective clinical trial.<br />

P.C. Verdonk, K. Moens, L. Willemot, T. Tampere, K.F. Almqvist, R. Verdonk, Gent/BE<br />

9.4.7 Quantitative MRI of degenerated lumbar caprine discs correlates with biomechanical and<br />

histological changes.<br />

C.P.L. Paul 1 , G.J. Strijkers 2 , M. De Graaf 1 , A. Bisschop 1 , A.J. Veen Van Der 1 , T.H. Smit 1 ,<br />

M.N. Helder 1 , B.J. Royen Van 1 , M.G. Mullender 1 , 1 Amsterdam/NL, 2 Eindhoven/NL<br />

9.4.8 Cell models for the human intervertebral disc: nucleus pulposus and annulus fibrosis<br />

G. Van Den Akker, T.J.M. Welting, D.A.M. Surtel, A. Cremers, J.-W. Voncken, L. Van Rhijn,<br />

Maastricht/NL<br />

9.4.9 Anterior Cruciate Ligament and <strong>Cartilage</strong> injury of the knee: A descriptive study<br />

K.K.V. Acharya, V. Pandey, Manipal/IN


Programme: Sunday, May 13, 2012<br />

Session 10.1 Industry Symposium<br />

Geistlich <strong>–</strong> Chondro-Gide® enhanced marrow stimulation <strong>–</strong> also for Ankle and Hip?<br />

13:00 <strong>–</strong> 14:00 Room: Marquette<br />

Moderator: Markus Walther (DE)<br />

10.1.1 Blood clot biology after marrow stimulation<br />

C.D. Hoemann, <strong>Montreal</strong>/CA<br />

10.1.2 AMIC Talus <strong>–</strong> Surgical technique and first clinical results<br />

M. Walther, Munich/DE<br />

10.1.3 AMIC Hip <strong>–</strong> Surgical approach and 5-year clinical outcome<br />

A. Fontana, Monza/IT<br />

Session 10.2 Industry Symposium<br />

Smith & Nephew <strong>–</strong> From Simple to Complex Meniscal Injury - How and When to Resect,<br />

<strong>Repair</strong> and Replace<br />

13:00 <strong>–</strong> 14:00 Room: Grand Salon<br />

Moderators: Peter Verdonk (BE), Tim Spalding (UK)<br />

10.2.1 How and When to Resect, <strong>Repair</strong> and Replace I<br />

T. Spalding, Coventry/UK<br />

10.2.2 How and When to Resect, <strong>Repair</strong> and Replace II<br />

P.C. Verdonk, Gent/BE<br />

Session 10.3 Industry Symposium<br />

Piramal <strong>–</strong> BST-CarGel ® : A novel bioscaffold for enhanced tissue regeneration in cartilage<br />

repair procedures<br />

13:00 <strong>–</strong> 14:00 Room: Jolliet<br />

Moderators: Matthias Steinwachs (DE), Pierre Ranger (CA)<br />

10.3.1 Underlying Principles of Enhancing Bone Marrow Stimulation with BST-CarGel ®<br />

M.S. Shive, Laval/CA<br />

10.3.2 MRI-based Imaging Biomarkers for <strong>Cartilage</strong><br />

T. Mosher, Hershey/US<br />

10.3.3 Final Outcomes from the BST-CarGel ® Randomized Clinical Trial<br />

W.D. Stanish, Halifax/CA<br />

63


64<br />

Programme: Sunday, May 13, 2012<br />

Session 10.4 Industry Symposium<br />

Anika <strong>–</strong> <strong>Cartilage</strong> regeneration with Hyalograft ® C autograft: Long term clinical results<br />

and future perspectives<br />

13:00 <strong>–</strong> 14:00 Room: St. François<br />

Moderator: Mats Brittberg (SE)<br />

10.4.1 Second Generation ACI 2012: An update on methodology and clinical results using Hyalograft C<br />

autograft transarthroscopically<br />

M. Brittberg, Kungsbacka/SE<br />

10.4.2 Experience with Hyalograft C autograft in the arthroscopic treatment of cartilage lesions;<br />

Long term results in athletes<br />

E. Kon, Bologna/IT<br />

10.4.3 Successful patient management with Hyalograft C autograft.<br />

S. Nehrer, Krems/AT<br />

Session 11.1 Free Paper<br />

Animal Models<br />

14:15 <strong>–</strong> 15:45 Room: Grand Salon<br />

Moderators: David Frisbie (US), Ernst Hunziker (CH)<br />

11.1.1 Overview & Introduction<br />

E. Hunziker, Bern/CH<br />

11.1.2 Versatility of cartilage limits flexibility in composition<br />

J. Malda, J.C. De Grauw, K.E.M. Benders, M. Kik, L.B. Creemers, W.J.A. Dhert, P.R. Van Weeren,<br />

Utrecht/NL<br />

11.1.3 Disruption of SIRT1 in chondrocytes causes accelerated development of osteoarthritis induced<br />

by joint instability in the mouse<br />

T. Matsuzaki, T. Matsushita, S. Kubo, T. Matsumoto, K. Takayama, H. Sasaki, M. Kurosaka,<br />

R. Kuroda, Kobe/JP<br />

11.1.4 In vivo effect of bone-specific EphB4 overexpression in mice on subchondral bone and cartilage<br />

during osteoarthritis<br />

G. Valverde-Franco 1 , M. Kapoor 1 , D. Hum 1 , K. Matsuo 2 , B. Lussier 3 , J.-P. Pelletier 1 ,<br />

J. Martel-Pelletier 1 , 1 <strong>Montreal</strong>/CA, 2 Tokyo/JP, 3 Saint-Hyacinthe/CA<br />

11.1.5 Four-day continuous blood exposure leads to prolonged joint damage in a canine in vivo model,<br />

whereas intermittent blood exposure does not<br />

M.E.R. Van Meegeren, G. Roosendaal, K. Van Veghel, N.W.D. Jansen, S.C. Mastbergen,<br />

F.P.J.G. Lafeber, Utrecht/NL<br />

11.1.6 <strong>Repair</strong> of Critical-Sized Defects with Engineered Constructs Generated Without Cell Expansion<br />

J. Brenner, Y. Tse, J. Pang, A. Winterborn, D. Bardana, S. Pang, S. Waldman, Kingston/CA<br />

11.1.7 Evidence for a role of the Wnt signalling pathway in osteochondral repair<br />

F. Henson, J. Power, A. Getgood, N. Rushton, Cambridge/UK


Programme: Sunday, May 13, 2012<br />

11.1.8 Distribution of laminin in early osteochondral repair in a goat model<br />

C.B. Foldager 1,2 , T. Cheriyan 2 , H.-P. Hsu 2 , M. Lind 1 , M. Spector 2 , 1 Aarhus/DK, 2 Boston/US<br />

11.1.9 The In Vivo Performance of Osteochondral Allografts in the Goat is Diminished with Extended<br />

Storage and Decreased <strong>Cartilage</strong> Cellularity<br />

A.L. Pallante 1 , S. Görtz 2 , A.C. Chen1, S.T. Ball 2 , D. Amiel 1 , K. Masuda 1 , R.L. Sah 1 , W.D. Bugbee 1 ,<br />

1 La Jolla/US, 2 San Diego/US<br />

Session 11.2 Free Paper<br />

Chondrocytes 2<br />

14:15 <strong>–</strong> 15:45 Room: St. François<br />

Moderators: Laurie Goodrich (US), Pieter Emans (NL)<br />

11.2.1 Human platelet lysate successfully promotes proliferation and subsequent chondrogenic<br />

differentiation of adipose-derived stem cells: a comparison to articular chondrocytes<br />

F. Hildner 1, 2 , M.J. Eder 1 , J. Aberl 1 , H. Redl 2 , C. Gabriel 1, 2 , M. Van Griensven 2 ,<br />

A. Peterbauer-Scherb 1,2 , 1 Linz/AT, 2 Vienna/AT<br />

11.2.2 Platelet Rich-Plasma Improves The Formation of Tissue Engineered <strong>Cartilage</strong><br />

M. Petrera 1 , J.A. Decroos 1 , J. Iu 1 , M.B.Hurtig 2 , R. Kandel 1 , J.Theodoropoulos 1 , 1 Toronto/CA, 2 Guelph/CA<br />

11.2.3 Activation of NF-KB/p65 Initiates Chondrogenic Differentiation of progenitor cells<br />

M.M.J. Caron, P. Emans, D.A.M. Surtel, A. Cremers, J.-W. Voncken, T.J.M. Welting, L.W. Van Rhijn,<br />

Maastricht/NL<br />

11.2.4 Modulation of NF-KB-dependent production of PGE2, IL-8, and MCP-1 in transfected chondrocytes<br />

S.L. Ownby, K.E. Walker, M.W. Grzanna, A.C. Mrozinski, L.F. Heinecke, A.Y. Au, C.G. Frondoza,<br />

Edgewood/US<br />

11.2.5 Egr-1 mediates the suppressive effect of IL-1 on PPARg expression in human OA chondrocytes<br />

S.S. Nebbaki, F. El Mansouri, N. Zayed, M. Benderdour, J. Martel-Pelletier, J.-P. Pelletier,<br />

H. Fahmi, <strong>Montreal</strong>/CA<br />

11.2.6 Development of chondrocytes resistant to IL-1beta and TNF-alpha through integrating<br />

transposon based RNA interference improve autologous chondrocyte implantation<br />

A.J. Nixon 1 , M. Scimeca 1 , K. Ortved 1 , J. Rossi 2 , 1 Ithaca/US, 2 Duarte/US<br />

11.2.7 Human Articular Chondrocytes Induce IL-2 Non-responsiveness to Allogeneic Lymphocytes<br />

S. Abe, H. Nochi, T. Ruike, T. Matsuno, Asahikawa/JP<br />

11.2.8 Spatial and Temporal Distribution of MAP Kinase Activity in Mechanically Injured <strong>Cartilage</strong> Explants<br />

D.H. Rosenzweig, M. Djap, T.M. Quinn, <strong>Montreal</strong>/CA<br />

11.2.9 Molecular Hydrogen Scavenges Hydroxyl Radicals and Protects Human Chondrocytes In Vitro<br />

K. O'Shaughnessey, A. Matuska, J. Woodell-May, Warsaw/US<br />

65


66<br />

Programme: Sunday, May 13, 2012<br />

Session 11.3 Free Paper<br />

Biomaterials & Scaffolds 2<br />

14:15 <strong>–</strong> 15:45 Room: Marquette<br />

Moderators: Kenneth Zaslav (US), Marcel Karperien (NL)<br />

11.3.1 Novel Scaffold-Based BST-CarGel ® Treatment Results in Superior <strong>Cartilage</strong> <strong>Repair</strong> in a<br />

Randomized Controlled Trial Compared to Microfracture. Better Structural <strong>Repair</strong> at 12 Months<br />

in Terms of <strong>Repair</strong> Tissue Quantity and Quality<br />

W.D. Stanish 1 , R. McCormack 2 , F. Forriol Campos 3 , N. Mohtadi 4 , S. Pelet 5 , J. Desnoyers 6 ,<br />

A. Restrepo 7 , M.S. Shive 7 , 1 Halifax/CA, 2 Vancouver/CA, 3 Madrid/ES, 4 Calgary/CA, 5 Quebec<br />

City/CA, 6 Greenfield Park/CA, 7 Laval/CA<br />

11.3.2 Comparison of safety and efficacy using synthetic scaffolds for treatment of medial and lateral<br />

painful irreparable partial meniscal defects; 24 months follow-up<br />

P.C. Verdonk 1 , R. Verdonk 2 , P. Beaufils 3 , J. Bellemans 4 , P. Colombet 5 , R. Cugat 6 , P. Djian 7 ,<br />

H. Laprell 8 , P. Neyret 9 , H. Paessler 10 , R. Siebold 10 , 1 Gent/BE, 2 Ghent/BE, 3 Versailles/FR,<br />

4 Leuven/BE, 5 Bordeaux/FR, 6 Barcelona/ES, 7 Paris/FR, 8 Kiel/DE, 9 Lyon/FR, 10 Heidelberg/DE<br />

11.3.3 A Nano-Composite Multilayered Biomaterial (MaioRegen) for the Treatment of Osteochondritis<br />

Dissecans in the Knee<br />

A.A.M. Dhollander 1 , K.F. Almqvist 2 , R. Verdonk 2 , G. Verbruggen 1 , P.C. Verdonk 3 , 1 Ghent/BE,<br />

2 Gent/BE, 3 Gent-Zwijnaarde/BE<br />

11.3.4 Synergistic Action of Fibroblast Growth Factor-2 and Transforming Growth Factor-beta1<br />

Enhances Human Neocartilage Formation in Bioprinted 3D Hydrogels<br />

X. Cui, M.K. Lotz, D.D. D'Lima, La Jolla/US<br />

11.3.5 Overall cytokine production correlates with cartilage matrix formation of expanded but not<br />

primary chondrocytes: a comparison of four biomaterials<br />

A.I. Tsuchida 1 , J.E.J. Bekkers 1 , M. Beekhuizen 1 , L. Vonk 1 , D.B.F. Saris 1, 2 , W.J.A. Dhert 1 ,<br />

L.B. Creemers 1 , 1 Utrecht/NL, 2 Enschede/NL<br />

11.3.6 Anti-inflammatory and anabolic effects of a new alginate-chitosan hydrogel beads on human<br />

chondrocytes<br />

F. Oprenyeszk 1 , C. Sanchez 1 , J.-E. Dubuc 2 , V. Maquet 3 , Y. Henrotin 1 , 1 Liege/BE, 2 Brussels/BE,<br />

3 Herstal/BE<br />

11.3.7 Effect of Irradiation on the Strength and Lubricity of PVA-PAA Hydrogels for <strong>Cartilage</strong> <strong>Repair</strong><br />

D. Ling, H. Bodugoz Senturk, H.L. Kluk, O.K. Muratoglu, Boston/US<br />

11.3.8 Development of a novel collagen-glycosaminoglycan scaffold for cartilage repair applications:<br />

Compositional, micro-structural and mechanical optimisation<br />

A. Matsiko, T.J. Levingstone, F.J. O'Brien, J.P. Gleeson, Dublin/IE<br />

11.3.9 Hydrogel-Nanofiber Scaffold System for Integrative Osteochondral <strong>Repair</strong><br />

N.T. Khanarian, R.A. Burga, N.M. Haney, E. Strauss, H. Lu, New York/US


Programme: Sunday, May 13, 2012<br />

Session 11.4 Free Paper<br />

Imaging<br />

14:15 <strong>–</strong> 15:45 Room: Jolliet<br />

Moderators: Sebastian Apprich (AT), Jukka Jurvelin (FI)<br />

11.4.1 Overview & Introduction<br />

J.S. Jurvelin, Kuopio/FI<br />

11.4.2 Delayed Gadolinium Enhanced MRI of <strong>Cartilage</strong> (dGEMRIC) demonstrates how cartilage<br />

regeneration influences other knee compartments<br />

J.E.J. Bekkers 1 , W. Bartels 1 , L.B. Creemers 1 , R. Benink 2 , A.I. Tsuchida 1 , K.L. Vincken 1 ,<br />

W.J.A. Dhert 1 , D.B.F. Saris 1 , 1 Utrecht/NL, 2 Den Helder/NL<br />

11.4.3 Biochemical MRI of Transplanted Osteochondral Allograft (OCA) cartilage with delayed<br />

Gadolinium-Enhanced MRI of <strong>Cartilage</strong> (dGEMRIC) and zonal T2 mapping at 1 and 2 years.<br />

D.S. Brown, M.G. Durkan, U. Szumowski, D.C. Crawford, Portland/US<br />

11.4.4 Standardized quantitative 3D MRI can detect superior cartilage repair in clinical trials and is<br />

correlated with collagen architecture in biopsies assessed by polarized light microscopy<br />

M.S. Shive 1 , W.D. Stanish 2 , R. McCormack 3 , F. Forriol Campos 4 , N. Mohtadi 5 , S. Pelet 6 ,<br />

J. Desnoyers 7 , J. Tamez-Pena 8 , S.M. Totterman 9 , A. Changoor 10 , A. Yaroshinsky 11 , S. Trattnig 12 ,<br />

A. Restrepo 1 , 1 Laval/CA, 2 Halifax/CA, 3 Vancouver/CA, 4 Madrid/ES, 5 Calgary/CA,<br />

6 Quebec City/CA, 7 Greenfield Park/CA, 8 Monterrey/MX, 9 Rochester/US, 10 <strong>Montreal</strong>/CA,<br />

11 San Andreas/US, 12 Vienna/AT<br />

11.4.5 In-vivo evaluation of biomechanical properties after cartilage repair in the patellofemoral joint<br />

by means of quantitative T2-mapping<br />

M. Pachowksi 1 , 2 , S. Trattnig 2 , B. Wondrasch 2 , S. Apprich 2 , S. Marlovits 2 , G.H. Welsch 1 , 2 ,<br />

1 Erlangen/DE, 2 Vienna/AT<br />

11.4.6 <strong>Cartilage</strong> Defects And Joint Problems In Morbidly Obese Children And Adolescents<br />

H.K. Widhalm, A. Neuhold, G.H. Welsch, G. Vekszler, S. Arbes, M. Hamboeck, S. Aldrian,<br />

S. Hajdu, S. Marlovits, K. Widhalm, Vienna/AT<br />

11.4.7 Does the mechanical alignment correlate with the tracer uptake pattern and intensity in<br />

SPECT/CT? A retrospective series on 104 knees<br />

M.T. Hirschmann, M.P. Arnold, N.F. Friederich, H. Rasch, Bruderholz/CH<br />

11.4.8 Quantitative Detection of <strong>Cartilage</strong> and Bone Tissue Quality via Spectral MARS-CT Imaging<br />

T. Woodfield, A.P.H. Butler, A. Siegert, G.J. Hooper, Christchurch/NZ<br />

11.4.9 Early <strong>Cartilage</strong> Injury Quantified and Characterized with Multiphoton Microscopy Imaging<br />

K. Novakofski, R. Williams, L.A. Fortier, L. Bonassar, Ithaca/US<br />

Session 12.0 ICRS General Assembly (For Members Only)<br />

15:45 <strong>–</strong> 17:00 Room: Jolliet<br />

Moderators: Daniël Saris (NL), Anthony Hollander (UK)<br />

Session 12.1 Poster Viewing Cocktail<br />

15:45 <strong>–</strong> 18:00 Room: Duluth/Richelieu<br />

Canadian Sugar Shack & Maple Forest Party<br />

18:30: Meeting point Fairmont hotel lobby<br />

18:45: Bus departure<br />

23:00: Return<br />

67


68<br />

Programme: Monday, May 14, 2012<br />

Session 13.1 Instructional Course 13.1<br />

Rehabilitation after <strong>Cartilage</strong> <strong>Repair</strong><br />

07:30 <strong>–</strong> 08:15 Room: Jolliet<br />

Instructors: Kai Mithoefer (US), Matthias Steinwachs (DE), Stefano Della Villa (IT)<br />

13.1.1 The Evolution of <strong>Cartilage</strong> <strong>Repair</strong> Rehabilitation<br />

K. Mithoefer, Cambridge/US<br />

13.1.2 Current Practice of <strong>Cartilage</strong> Rehabilitation<br />

M. Steinwachs, Zürich/CH<br />

13.1.3 On-Field Rehabilitation and Return to Sport<br />

S. Della Villa, Bologna/IT<br />

Session 13.2 Meet the Experts (for Clinicians)<br />

07:30 <strong>–</strong> 08:15 Room: Marquette<br />

Moderators: Brian Cole (US), Christoph Erggelet (CH)<br />

Session 14.0 Plenary<br />

Cell Free Approaches for <strong>Cartilage</strong> <strong>Repair</strong><br />

08:30 <strong>–</strong> 09:30 Room: Grand Salon<br />

Moderators: Daniël Saris (NL), Jos Malda (NL)<br />

14.1 Homing of MSC as response to Growth factor<br />

J. Mao, New York/US<br />

14.2 The current prospects for gene therapy as a non-cellular therapy<br />

L. Goodrich 1 , W. McIlwraith 1 , J. Samulski 2 , 1 Fort Collins/US, 2 Chapel Hill/US<br />

Session 15.1 Special<br />

Platelet Rich Plasma and Joint Tissue <strong>Repair</strong><br />

09:45 <strong>–</strong> 10:45 Room: Grand Salon<br />

Moderators: Maurilio Marcacci (IT), Elizaveta Kon (IT)<br />

15.1.1 Platelet rich plasma: Overview of current knowledge: hope, hype and reality.<br />

L.A. Fortier 1 , T. McCarrell 1 , B.J. Cole 2 , S. Boswell 1 , L.V. Schnabel 1 , 1Ithaca/US, 2 Chicago/US<br />

15.1.2 Platelet rich plasma and joint tissue repair<br />

E. Kon, G. Filardo, B. Di Matteo, A. Di Martino, M. Marcacci, Bologna/IT<br />

15.1.3 Clinical experiences with Platelet-Rich Plasma<br />

S. Rodeo, New York/US


Programme: Monday, May 14, 2012<br />

Session 15.2 Special<br />

Development of new Biomaterials & Scaffolds<br />

09:45 <strong>–</strong> 10:45 Room: Marquette<br />

Moderators: Michael Buschmann (CA), Dietmar Hutmacher (AU)<br />

15.2.1 Biomaterials and the osteochondral interface<br />

H. Lu N.T. Khanarian E. Hunziker E. Strauss,<br />

15.2.2 Hydrogels in cartilage repair<br />

J.D. Kisiday, D.D. Frisbie, L. Goodrich, W. McIlwraith, Fort Collins/US<br />

15.2.3 Clinical application of scaffolds for cartilage repair<br />

S. Nehrer, Krems/AT<br />

Session 15.3 Special<br />

Subchondral Bone & <strong>Cartilage</strong> <strong>Repair</strong><br />

09:45 <strong>–</strong> 10:45 Room: St. François<br />

Moderators: Henning Madry (DE), Mario Ferretti (BR)<br />

15.3.1 Importance of subchondral bone in cartilage repair<br />

H. Madry, Homburg/DE<br />

15.3.2 Digital imaging of subchondral bone density<br />

F.P.J.G. Lafeber, M. Kinds, F. Intema, S.C. Mastbergen, Utrecht/NL<br />

15.3.3 Fresh Osteochondral Allografting in the Knee<br />

W.D. Bugbee, La Jolla/US<br />

Coffee Break/Industry Exhibition<br />

10:45 AM <strong>–</strong> 11:15 AM Room: Exhibition Hall<br />

Session 16.1 Free Paper<br />

Osteoarthritis 1<br />

11:15 <strong>–</strong> 12:45 Room: St. François<br />

Moderators: Linda Sandell (US), Anthony Hollander (UK)<br />

16.1.1 Overview & Introduction<br />

A.P. Hollander, Bristol/UK<br />

16.1.2 Biochemical characterization of osteoarthritis and osteochondritis dissecans in the ankle<br />

H. Schmal, R. Henkelmann, I.H. Pilz, A.T. Mehlhorn, N.P. Südkamp, P. Niemeyer, Freiburg/DE<br />

16.1.3 Osteoarthritis of lower extremities in former skiers<br />

M.I. Iosifidis, I. Melas, E. Iliopoulos, K. Apostolidis, A. Kyriakidis, Thessaloniki/GR<br />

16.1.4 Genetic correlation between articular cartilage regeneration and ear-wound healing:<br />

implications in osteoarthritis<br />

M.F. Rai, S. Hashimoto, J. Fitzgerald, E. Heber-Katz, J.M. Cheverud, L.J. Sandell, St. Louis/US<br />

69


70<br />

Programme: Monday, May 14, 2012<br />

16.1.5 A Promising Triad in the Biological Prevention of Experimental Post-Traumatic Osteoarthritis<br />

R. Olewinski, A.A. Hakimiyan, L. Rappoport, C. Pacione, M.A. Wimmer, S. Chubinskaya,<br />

Chicago/US<br />

16.1.6 Leptin and adiponectin are differentially regulated in cartilage from patients with osteoarthritis<br />

P.-J. Francin, C. Guillaume, P. Gegout-Pottie, P. Netter, D. Mainard, N. Presle, Vandoeuvre Les<br />

Nancy Cedex/FR<br />

16.1.7 Inflammatory factors released by osteoarthritic chondrocytes and synoviocytes are downmodulated<br />

by adipose stromal cells<br />

C. Manferdini 1 , A. Piacentini 1 , E. Gabusi 1 , J.A. Peyrafitte 2 , P. Bourin 2 , C. Jorgensen 3 ,<br />

M. Maumus 3 , D. Noël 3 , A. Facchini 1 , G. Lisignoli 1 , 1 Bologna/IT, 2 Toulouse/FR, 3 Montpellier/FR<br />

16.1.8 MicroRNA-125b Regulates the Expression of Aggrecanase-1 (ADAMTS4) in Human Osteoarthritic<br />

Chondrocytes<br />

T. Matsukawa, T. Sakai, H. Hiraiwa, T. Hamada, T. Omachi, M. Nakashima, S. Ishizuka, T. Oda, A.<br />

Takamatsu, S. Yamashita, N. Ishiguro, Nagoya/JP<br />

16.1.9 Human articular cartilage metabolism as modulated by collagen hydrolysates<br />

S. Schadow, J. Kordelle, G. Lochnit, J. Steinmeyer, Giessen/DE<br />

Session 16.2 Free Paper<br />

<strong>Cartilage</strong> / Cell Transplantation (Pre-clinical)<br />

11:15 <strong>–</strong> 12:45 Room: Marquette<br />

Moderators: Alberto Gobbi (IT), Daniel Grande (US)<br />

16.2.1 Overview & Introduction<br />

D. Grande, Manhasset/US<br />

16.2.2 An osteochondral culture model to study mechanisms involved in articular cartilage repair<br />

M.L. De Vries - Van Melle 1 , E. Mandl 1,2 , N. Kops 1 , W. Koevoet 1 , J. Verhaar 1 , G.J.V.M. Van Osch 1 ,<br />

1 Rotterdam/NL, 2 Dordrecht/NL<br />

16.2.3 Simulated autologous chondrocyte transplantation within arthritic surroundings<br />

J.D. Erggelet 1, 2 , G.M. Salzmann 1 , P. Niemeyer 1 , G. Pattappa 2 , S. Grad 2 , M. Alini 2 , 1 Freiburg/DE,<br />

2 Davos Platz/CH<br />

16.2.4 Hypoxic expansion of MSCs improves chondrogenicity and cell yield <strong>–</strong> implications for the<br />

optimal use of MSCs in cartilage tissue engineering<br />

N. Georgi, C. Van Blitterswijk, M. Karperien, Enchede/NL<br />

16.2.5 The effects of an in vitro low oxygen tension preconditioning of MSC on their in vivo<br />

chondrogenic potential: application for cartilage tissue engineering<br />

S. Portron, C. Merceron, O. Gauthier, J. Lesoeur, S. Sourice, M. Masson, B.H. Fellah, O. Geffroy,<br />

E. Lallemand, P. Weiss, J. Guicheux, C. Vinatier, Nantes/FR<br />

16.2.6 Chromosomal stability of human chondrocytes in the process of scaffold-assisted cartilage<br />

tissue engineering.<br />

M. Endres 1 , M. Trimborn 1 , C. Bommer 1 , J.-P. Krüger 1 , U. Freymann 1 , L. Morawietz 2 , P.C. Kreuz 3 ,<br />

C. Kaps 1 , 1 Berlin/DE, 2 Stuttgart/DE, 3 Rostock/DE


Programme: Monday, May 14, 2012<br />

16.2.7 A Novel Approach For Preserving The Chondrogenic Phenotype Of Expanded Primary And Bone<br />

Marrow-Derived Chondrocytes: Implications For Cell-Based <strong>Cartilage</strong> <strong>Repair</strong><br />

C.M. Simonaro, S. Sachot, E.H. Schuchman, New York/US<br />

16.2.8 Mechanical Stimulation Enhances Integration in an in vitro model of <strong>Cartilage</strong> <strong>Repair</strong><br />

J. Theodoropoulos, J.A. Decroos, M. Petrera, S. Park, R. Kandel, Toronto/CA<br />

16.2.9 CAIS: European Multicenter Results of a Single Stage Procedure for cell-based <strong>Cartilage</strong> <strong>Repair</strong>:<br />

36-month follow up<br />

M. Brittberg 1 , S. Nehrer 2 , A.B. Imhoff 3 , T. Spalding 4 , C.S. Winalski 5 , J. Bothos 6 , B.A. Byers 7 ,<br />

K.F. Almqvist 8 , 1 Kungsbacka/SE, 2 Krems/AT, 3 Munich/DE, 4 West Midlands/UK, 5 Cleveland/<br />

US, 6 Somerville/US, 7 Raynham/US, 8 Gent/BE<br />

Session 16.3 Free Paper<br />

Microfracture / Bone Marrow Stimulation<br />

11:15 <strong>–</strong> 12:45 Room: Jolliet<br />

Moderators: William Rodkey (US), Marc Philippon (US)<br />

16.3.1 Overview & Introduction<br />

M. Philippon, Vail/US<br />

16.3.2 Stereological analysis of subchondral angiogenesis in bone marrow stimulated cartilage repair<br />

C. Mathieu 1 , G. Chen 1 , A. Chevrier 1 , V. Lascau-Coman 1 , C. Marchand 1 , J. Sun 2 , G.-É. Rivard 1 ,<br />

C.D. Hoemann 1 , 1 <strong>Montreal</strong>/CA, 2 Laval/CA<br />

16.3.3 4-Hole microfracture (MFX) with platelet rich plasma (PRP) soaked synovial sponges<br />

A.B. Anderson 1 , J.J. Rodrigo 2 , J. Desjardins 3 , M.C. Ware 3 , D. Wyland 1 , M. Suri 4 , G.W. Bennett 1 ,<br />

P.H. Wessinger 1 , P.C. Siffri 1 , R.J. Hawkins 1 , 1 Greenville/US, 2 Waco/US, 3 Clemson/US, 4 Harahan/US<br />

16.3.4 Bone marrow stimulation induces greater chondrogenesis in trochlear versus condylar cartilage<br />

defects in skeletally mature rabbits<br />

H. Chen 1 , A. Chevrier 1 , C.D. Hoemann 1 , J. Sun 2 , V. Lascau-Coman 1 , M.D. Buschmann 1 ,<br />

1 <strong>Montreal</strong>/CA, 2 Laval/CA<br />

16.3.5 Microfracture Treatment In Athletes With Knee Grade IV Chondral Lesion <strong>–</strong> A 10 Year Follow Up.<br />

A. Gobbi, A. Kumar, G. Karnatzikos, Milan/IT<br />

16.3.6 Does a meta-analysis reveal differences in the clinical outcome achieved by microfracture and<br />

autologous condrocyte implantation?<br />

L.L. Negrin, V. Vecsei, Vienna/AT<br />

16.3.7 Microfracture or AMIC for arthroscopic repair of acetabular cartilage defects in<br />

femoroacetabular impingement.<br />

A. Fontana, Milano/IT<br />

16.3.8 Incidence, grading and clinical significance of subchondral bone changes after microfracture of<br />

chondral defects in the knee<br />

K. Mithoefer, V. Venugopal, Cambridge/US<br />

16.3.9 The reaction of subchondral bone to ex vivo microfracture using awls with distinct shape and<br />

symptomatic human OA knee condyles with and without prior steroid therapy<br />

C.D. Hoemann 1 , Y. Gosselin 1 , P. Cazelais 1 , J. Sun 2 , H. Chen 1 , V. Lascau-Coman 1 , M.B. Hurtig 3 ,<br />

A. Carli 1 , W.D. Stanish 4 , 1 <strong>Montreal</strong>/CA, 2 Laval/CA, 3 Guelph/CA, 4 Halifax/CA<br />

71


72<br />

Programme: Monday, May 14, 2012<br />

Session 16.4 Free Paper<br />

Stem Cells 1<br />

11:15 <strong>–</strong> 12:45 Room: Grand Salon<br />

Moderators: Lisa Fortier (US), Barry Oakes (AU)<br />

16.4.1 Overview & Introduction<br />

W. Kafienah, Bristol/UK<br />

16.4.2 Direct generation of mature chondrocytes from human induced pluripotent stem cells<br />

A. Owaidah 1 , S.C. Dickinson 1 , H. Jia 1 , L. Carpenter 2 , S.M. Watt 2 , A.P. Hollander 1 , W. Kafienah 1 ,<br />

1 Bristol/UK, 2 Oxford/UK<br />

16.4.3 Comparison of gene-specific DNA methylation patterns in equine induced pluripotent stem cell<br />

lines with cells derived from equine adult and fetal tissues<br />

C. Hackett 1 , L. Greve 2 , K.D. Novakofski 1 , L.A. Fortier 1 , 114853/US, 2 Copenhagen/DK<br />

16.4.4 Multiscale Approach to Stem Cell-based Chondrogenesis for <strong>Cartilage</strong> <strong>Repair</strong><br />

C.-L. Chou, A.L. Rivera, A. Caplan, V.M. Goldberg, J.F. Welter, H. Baskaran, Cleveland, Oh/US<br />

16.4.5 Mesenchymal stem cells exert paracrine effects on osteoarthritic cartilage and synovium<br />

G. Van Buul 1 , E. Villafuertes 1, 2 , J. Waarsing 1 , P.K. Bos 1 , N. Kops 1 , R. Narcisi 1 , J. Verhaar 1 ,<br />

H. Weinans 1, 3 , M. Bernsen 1 , G.J.V.M. Van Osch 4 , 1 Rotterdam/NL, 2 Madrid/ES, 3 Delft/NL, 4 Rotterdam/<br />

16.4.6 Hypoxia Delays Hypertrophy In Human Multipotent Stromal Cells<br />

D. Gawlitta, M.H.P. Van Rijen, E. Schrijver, J. Alblas, W.J.A. Dhert, Utrecht/NL<br />

16.4.7 Liposome-mediated transfection of microRNA-145 into mesenchymal stem cells and articular<br />

chondrocytes induce unwanted upregulation of immune genes<br />

T.A. Karlsen, T. Küntziger, J.E. Brinchmann, Oslo/NO<br />

16.4.8 Structured 3D Co-culture of Mesenchymal Stem Cells with Fibrochondrocytes Promotes<br />

Meniscal Phenotype without Hypertrophy<br />

X. Cui, M.K. Lotz, D.D. D'Lima, La Jolla/US<br />

16.4.9 Implantation of Allogenic Synovial Stem Cells Promotes Meniscal Regeneration in a Rabbit<br />

Meniscal Defect Model<br />

M. Horie 1 , M.D. Driscoll 2 , H..W. Sampson 2 , I. Sekiya 1 , C. Caroom 2 , D. Prockop 2 , D. Thomas 2 ,<br />

1 Tokyo/JP, 2 Temple/US


Programme: Monday, May 14, 2012<br />

Session 17.1 Industry Symposium<br />

Sanofi Biosurgery <strong>–</strong> The Evolution of Autologous Chondrocyte Implantation and <strong>Cartilage</strong> <strong>Repair</strong><br />

13:00 <strong>–</strong> 14:00 Room: Grand Salon<br />

Moderators: Sven Kili (US)<br />

17.1.1 The Genesis of ACI and the Significance of the Chondrocyte in <strong>Cartilage</strong> <strong>Repair</strong><br />

L. Peterson, Västra Frölunda/SE<br />

17.1.2 Evolving Standards in Clinical Research<br />

M. Brittberg, Kungsbacka/SE<br />

17.1.3 Evolving Standards in the Manufacturing of Cell <strong>–</strong> Based Therapies<br />

S.J. Duguay, Cambridge/US<br />

Session 17.2 Industry Symposium<br />

Zimmer Inc. <strong>–</strong> Innovative Solutions for Joint Preservation with DeNovo ® NT Natural Tissue<br />

Graft and Chondrofix ® Osteochondral Allograft<br />

13:00 <strong>–</strong> 14:00 Room: Marquette<br />

17.2.1 Strategies for Articular <strong>Cartilage</strong> Restoration<br />

D. Caborn, Louisville/US<br />

17.2.2 Clinical Applications of <strong>Cartilage</strong> <strong>Repair</strong><br />

J. Farr, Greenwood/US<br />

17.2.3 <strong>Cartilage</strong> Technologies for Talus OCD<br />

E. Giza, Sacramento/US<br />

17.2.4 New Concepts in <strong>Cartilage</strong> <strong>Repair</strong><br />

N. Marcus, Springfield/US<br />

Session 17.3 Industry Symposium<br />

Tigenix <strong>–</strong> ChondroCelect: When evidence makes a difference<br />

13:00 <strong>–</strong> 13:30 Room: Jolliet<br />

Moderators: Karl Almqvist (BE)<br />

17.3.1 ChondroCelect <strong>–</strong> When evidence makes a difference<br />

K.F. Almqvist, Gent/BE<br />

73


74<br />

Programme: Monday, May 14, 2012<br />

Session 17.4 Industry Symposium<br />

BioTissue <strong>–</strong> BioTissue’s stable implants for cartilage repair: up to 10 years clinical data,<br />

biomechanics and histologies<br />

13:00 <strong>–</strong> 14:00 Room: St. François<br />

17.4.1 Clinical application of BioTissue products for cartilage regeneration: A decade of experience<br />

J. Holz, Hamburg/DE<br />

17.4.2 Pin fixation of BioSeed ® -C and chondrotissue ® <strong>–</strong> Biomechanical properties and pitfalls of this<br />

arthroscopic procedure<br />

M. Herbort, Muenster/DE<br />

17.4.3 Knee articular cartilage resurfacing using synthetic scaffolds and fresh chondral autografts<br />

J. Fernandes, <strong>Montreal</strong>/CA<br />

17.4.4 Four <strong>–</strong> year results after polymer-based ACI with BioSeed ® -C for the treatment of chondral<br />

defects in the knee.<br />

P.C. Kreuz, Rostock/DE<br />

Session 18.0 Plenary<br />

Stem Cells for <strong>Cartilage</strong> <strong>Repair</strong><br />

14:15 <strong>–</strong> 15:15 Room: Grand Salon<br />

Moderators: Anthony Hollander (UK)<br />

18.1 Stem Cell Therapy for Joint <strong>Repair</strong><br />

F. Barry, Galway/IE<br />

18.2 Stem cell - based therapeutic approaches to joint surface repair<br />

C. De Bari, Aberdeen/UK<br />

Session 19.1 Special<br />

Imaging Technologies for MS Tissue <strong>Repair</strong><br />

15:30 <strong>–</strong> 16:30 Room: Marquette<br />

Moderators: Jukka Jurvelin (FI), Carl Winalski (US)<br />

19.1.1 Basics on ultrastructural-multiparametric MR techniques<br />

S. Apprich 1 , G.H. Welsch 1, 2 , S. Zbyn 1 , B. Schmitt 1 , S. Trattnig 1 , 1 Vienna/AT, 2 Erlangen/DE<br />

19.1.2 Clinical application of ultrastructural-multiparametric MR techniques in patients after repair surgery<br />

G.H. Welsch 1, 2 , 1 Erlangen/DE, 2 Vienna/AT<br />

19.1.3 Quantitative X-ray and ultrasound methodology for cartilage repair<br />

J.S. Jurvelin 1 , T. Viren 1 , H. Kokkonen 1 , J. Liukkonen 1 , K. Kulmala 1 , A. Joukainen 1 , I. Kiviranta 2 ,<br />

J. Salo 1 , H. Kröger 1 , J. Töyräs 1 , 1 Kuopio/FI, 2 Helsinki/FI


Programme: Monday, May 14, 2012<br />

Session 19.2 Special<br />

YSOS Symposium “One-Stage Procedures for <strong>Cartilage</strong> <strong>Repair</strong>”<br />

15:30 <strong>–</strong> 16:30 Room: Jolliet<br />

Moderators: Simon Görtz (US), Joris Bekkers (NL)<br />

19.2.1 Current Treatment Paradigm for Single-Stage <strong>Cartilage</strong> <strong>Repair</strong><br />

A.H. Gomoll, Boston/US<br />

19.2.2 Emerging technologies<br />

A.A.M. Dhollander 1 , K.F. Almqvist 2 , P.C. Verdonk 2 , R. Verdonk 2 , G. Verbruggen 2 , J. Victor 2 ,<br />

1 Gent/BE, 2 Ghent/BE<br />

19.2.3 Concomitant procedures<br />

A. Getgood, Coventry/UK<br />

Session 19.3 Special<br />

Biomarkers<br />

15:30 <strong>–</strong> 16:30 Room: Grand Salon<br />

Moderators: Bruce Caterson (UK), Anthony Poole (CA)<br />

19.3.1 Biomarkers for diagnosis, screening and monitoring of treatment in cartilage repair and<br />

osteoarthritis<br />

S. Lohmander, Lund/SE<br />

19.3.2 Emerging molecular biomarker technologies and the way forward<br />

A.R. Poole, <strong>Montreal</strong>/CA<br />

19.3.3 Genetic influence on cartilage repair<br />

L.J. Sandell, St. Louis/US<br />

Session 20.0 Poster Viewing / Coffee Break<br />

16:30 <strong>–</strong> 17:30 Room: Duluth/Richelieu<br />

Session 21.1 Special<br />

Joint Lubrication & <strong>Cartilage</strong> Health<br />

17:30 <strong>–</strong> 18:30 Room: Grand Salon<br />

Moderators: Robert Sah (US), Carl Flannery (US)<br />

21.1.1 Role of lubrication and joint homeostasis: <strong>Cartilage</strong> Boundary Lubricating Ability of Full-Length<br />

Recombinant Human PRG4 <strong>–</strong> Alone and In Combination with Hyaluronan<br />

S. Abubacker 1 , N. Masala 1 , S. Morrison 1 , G.D. Jay 2 , T.A. Schmidt 1 , 1 Calgary/CA, 2 Providence/US<br />

21.1.2 Opportunities and challenges for joint lubrication therapies<br />

R.L. Sah, M.M. Temple-Wong, La Jolla/US<br />

21.1.3 New frontiers in joint lubrication therapy<br />

C.R. Flannery, Acton/US<br />

75


76<br />

Programme: Monday, May 14, 2012<br />

Session 21.2 Special<br />

Rehabilitation<br />

17:30 <strong>–</strong> 18:30 Room: Marquette<br />

Moderators: Kai Mithoefer (US), Moreno Morelli (CA)<br />

21.2.1 Rehabilitation after cartilage repair<br />

M. Steinwachs, Zürich/CH<br />

21.2.2 Return to sports after articular cartilage repair<br />

K. Mithoefer, Chestnut Hill/US<br />

21.2.3 Challenges in Standardizing Rehabilitation in <strong>Cartilage</strong> <strong>Repair</strong> RCTs<br />

M.S. Shive, <strong>Montreal</strong>/CA<br />

Session 21.3 Special<br />

ICRS / Stryker Clinical Scientist Programme<br />

17:30 <strong>–</strong> 18:30 Room: Jolliet<br />

Moderators: Lisa Fortier (US), Lars Peterson (SE)<br />

21.3.1 The Evolution of Osteochondral Storage and Preservation Techniques<br />

A.L. Pallante, La Jolla/US<br />

21.3.2 The Immunology of Osteochondral Grafting<br />

W.D. Bugbee, La Jolla/US<br />

President's Dinner (Upon invitation only)<br />

19:15: Meeting point Fairmont lobby<br />

19:30 <strong>–</strong> 23:00: Dinner


Programme: Tuesday, May 15, 2012<br />

Session 22.0 Plenary<br />

Biomechanics & Stability of <strong>Cartilage</strong> <strong>Repair</strong><br />

08:30 <strong>–</strong> 09:30 Room: Grand Salon<br />

Moderators: Tom Minas (US), Wayne Gersoff (US)<br />

22.1 Contribution of other tissues to cartilage degeneration<br />

C.B. Little, St. Leonards, Nsw/AU<br />

22.2 <strong>Cartilage</strong> repair; what does it mean and how to assess cartilage repair outcome?<br />

L.E. Dahlberg, Malmö/SE<br />

Session 23.0 Plenary<br />

Strategic Outlines ICRS<br />

09:30 <strong>–</strong> 10:00 Room: Grand Salon<br />

Moderators: Anthony Hollander (UK), Daniël Saris (NL)<br />

23.1 Where are we now?<br />

D.B.F. Saris, Utrecht/NL<br />

23.2 Where do we want to go?<br />

A.P. Hollander, Bristol/UK<br />

Coffee Break/Industry Exhibition<br />

10:00 <strong>–</strong> 10:15 Room: Exhibition Hall<br />

Session 24.1 Special<br />

<strong>Cartilage</strong> <strong>Repair</strong> in the Hip Joint<br />

10:30 <strong>–</strong> 11:30 Room: Marquette<br />

Moderators: Christoph Erggelet (CH), Marc Philippon (US)<br />

24.1.1 Change of thinking in treating cartilage defects in the hip<br />

M. Philippon, Vail/US<br />

24.1.2 ACI in the hip joint<br />

J.B. Richardson, Oswestry/UK<br />

24.1.3 <strong>Cartilage</strong> repair in the hip joint<br />

R.M. Mardones, Santiago/CL<br />

Session 24.2 Special<br />

Animal Models for <strong>Cartilage</strong> Tissue Regeneration<br />

10:30 <strong>–</strong> 11:30 Room: Grand Salon<br />

Moderators: Wayne McIlwraith (US), Ernst Hunziker (CH)<br />

24.2.1 ICRS consensus on animal models<br />

M.B. Hurtig, Guelph/CA<br />

24.2.2 Rabbit cartilage repair models: a review<br />

C.D. Hoemann, <strong>Montreal</strong>/CA<br />

24.2.3 Equine models: a review<br />

D.D. Frisbie, W. McIlwraith, Fort Collins/US<br />

77


78<br />

Programme: Tuesday, May 15, 2012<br />

Session 24.3 Special<br />

Distraction Arthroplasty<br />

10:30 <strong>–</strong> 11:30 Room: Jolliet<br />

Moderators: Simon C Mastbergen (NL), Annunziato Amendola (US)<br />

24.3.1 Does unloading of joint surfaces affect cartilage healing in patients with end-stage ankle<br />

osteoarthritis; an overview of the literature<br />

A.N. Amendola 1 , M. Nguyen 1 , C. Saltzman 2 , 1 Iowa City/US, 2 Salt Lake City, Ut/US<br />

24.3.2 <strong>Cartilage</strong> and subchondral bone repair by joint distraction in clinical and experimental models<br />

of joint degeneration<br />

P.M. Van Roermund, Utrecht/NL<br />

24.3.3 Panel Discussion<br />

S.C. Mastbergen, Utrecht/NL<br />

Session 25.1 Free Paper<br />

Osteoarthritis 2<br />

11:30 <strong>–</strong> 13:00 Room: St. François<br />

Moderators: Elizaveta Kon (IT), Anthony Poole (CA)<br />

25.1.1 PRP injections versus viscosupplementation for early knee osteoarthritis: a randomized<br />

double-blind study<br />

G. Filardo, E. Kon, A. Di Martino, S. Patella, B. Di Matteo, F. Perdisa, M.L. Merli, M. Marcacci,<br />

Bologna/IT<br />

25.1.2 Structural Tissue <strong>Repair</strong> and Prolonged Clinical Improvement by Joint Distraction in Treatment of<br />

End-stage Knee Osteoarthritis; Two Years Follow-up<br />

K. Wiegant 1 , F. Intema 1 , P.M. Van Roermund 1 , A.C.A. Marijnissen 1 , S. Cotofana 2 , F. Eckstein 2 ,<br />

S.C. Mastbergen 1 , F.P.J.G. Lafeber 1 , 1 Utrecht/NL, 2 Salzburg/AT<br />

25.1.3 Early stages of knee OA alter type 2b fibers of the vastus lateralis muscle and decrease<br />

eccentric knee extensor torque<br />

P.R. Serrão 1 , K. Gramani-Say 1 , F.A. Vasilceac 1 , G.C. Lessi 1 , R. Reiff 2 , A.C. Mattiello-Sverzut 3 ,<br />

S.M. Mattielo 1 , 1 São Carlos/BR, 2 Sao Carlos/BR, 3 Ribeirao Preto/BR<br />

25.1.4 Relationship between serum hyaluronan level and progression of knee osteoarthritis <strong>–</strong><br />

A two-year longitudinal study in a Japanese general population <strong>–</strong><br />

E. Sasaki, Y. Ishibashi, E. Tsuda, Y. Yamamoto, R. Inoue, I. Takahashi, M. Matsuzaka, T. Umeda,<br />

S. Nakaji, S. Toh, Hirosaki/JP<br />

25.1.5 Relationship between cartilage wear and meniscal contact in medial osteoarthritis of the knee<br />

S. Arno, P. Walker, C. Bell, S. Krasnokutsky, J. Samuels, S. Abramson, R. Regatte, M.P. Recht,<br />

New York/US<br />

25.1.6 Specific absence of PPAR-gamma in mouse cartilage results in early cartilage developmental<br />

defects and spontaneous osteoarthritis<br />

F. Vasheghani Farahani 1 , R. Monemjou 1 , H. Fahmi 1 , G. Perez 1 , M. Blati 1 , N. Taniguchi 2 , M.K.<br />

Lotz 2 , R. St-Arnaud 1 , J.-P. Pelletier 1 , J. Martel-Pelletier 1 , F. Beier 3 , M. Kapoor 1 , 1 <strong>Montreal</strong>/CA,<br />

2 La Jolla/US, 3 London/CA


Programme: Tuesday, May 15, 2012<br />

25.1.7 Inhibition of Interleukin-1 Prevents Post-Traumatic Arthritis Following Articular Fracture in the<br />

Mouse Knee<br />

D.S. Mangiapani, E.M. Zeitler, B.D. Furman, J.L. Huebner, V.B. Kraus, F. Guilak, S.A. Olson,<br />

Durham, Nc/US<br />

25.1.8 In Vivo Localization, Persistence, and External Control of Adeno-Associated Virus (AAV)-<br />

Mediated Transgene Expression in Injured Knees<br />

H.H. Lee 1 , M. O'Malley 1 , N.A. Friel 1 , K.A. Payne 1 , X. Xiao 2 , C. Chu 1 , 1 Pittsburgh/US, 2 Chapel Hill/US<br />

25.1.9 The relationship between HTRA1 and type VI collagen in a single impact load model of cartilage<br />

damage<br />

P. Hernandez, A. Getgood, N. Rushton, F. Henson, Cambridge/UK<br />

Session 25.2 Free Paper<br />

<strong>Cartilage</strong> / Cell Transplantation (Clinical)<br />

11:30 AM <strong>–</strong> 13:00 Room: Marquette<br />

Moderators: Barry Oakes (AU), Patrick Lavigne (CA)<br />

25.2.1 Overview & Introduction<br />

A. Gobbi, Milano/IT<br />

25.2.2 Evidence of Response Shift in Patient Reported Outcomes (PROs) following Autologous<br />

Chondrocyte Implantation (ACI)<br />

J.S. Howard 1 , C.G. Mattacola 1 , D.R. Mullineaux 2 , R.A. English 1 , C. Lattermann 1 , 1 Lexington/US,<br />

2 Lincoln/UK<br />

25.2.3 <strong>Repair</strong> of focal cartilage defects with polymer-based autologous chondrocyte grafts: clinical,<br />

functional and biomechanical results 48 months after transplantation<br />

P.C. Kreuz 1 , S. Müller 2 , U. Freymann 3 , C. Erggelet 2 , P. Niemeyer 2 , C. Kaps 3 , A. Hirschmüller 2 ,<br />

1 Rostock/DE, 2 Freiburg/DE, 3 Berlin/DE<br />

25.2.4 Autologous Chondrocyte Implantation and Anteromedialization of Isolated Patella Articular<br />

<strong>Cartilage</strong> Lesions: 5 to 12 Year Follow-up<br />

R.M. Arnold 1 , S.D. Gillogly 2 , 1 Omaha/US, 2 Atlanta/US<br />

25.2.5 Incidence of Osteophyte regrowth in Patients treated with autologous chondrocyte<br />

implantation and previous bone marrow stimulating techniques: a radiographic study.<br />

A. Von Keudell, S. Sodha, K. Small, T. Minas, A.H. Gomoll, Boston/US<br />

25.2.6 Clinical and radiologic long-term outcome following first genereation autologous chondrocyte<br />

implantation for cartilage defects across the knee joint<br />

G.M. Salzmann 1 , S. Porichis 1 , N. Ghanem 1 , M. Uhl 1 , M. Steinwachs 2 , N.P. Südkamp 1 ,<br />

P. Niemeyer 1 , 1 Freiburg/DE, 2 Zürich/CH<br />

25.2.7 In vivo evaluation of four surgical variants for autologous chondrocyte implantation<br />

M. Maréchal 1 , H. Vanhauwermeiren 2 , J. Neys 1 , T. Van De Putte 1 , 1 Leuven/BE, 2 Diest/BE<br />

25.2.8 Novel surgical treatment for cartilage reconstruction in the foot and ankle<br />

H. Thermann, F. Süzer, Heidelberg/DE<br />

25.2.9 Second Generation ACI: Mid-term Results and Prognostic Factors<br />

G. Filardo, E. Kon, A. Di Martino, S. Patella, F. Perdisa, B. Di Matteo, F. Vannini, M. Marcacci,<br />

Bologna/IT<br />

79


80<br />

Programme: Tuesday, May 15, 2012<br />

Session 25.3 Free Paper<br />

Rehabilitation<br />

11:30 <strong>–</strong> 13:00 Room: Jolliet<br />

Moderators: Jack Farr (US), Mario Ferretti (BR)<br />

25.3.1 Overview & Introduction<br />

K. Hambly, Chatham/UK<br />

25.3.2 Weekly Changes in Serum <strong>Cartilage</strong> Oligomeric Matrix Protein (sCOMP) Levels for Soccer<br />

Athletes Over a 10 Week Spring Soccer Season<br />

J.M. Hoch, J.L. Mateer, C.G. Mattacola, C. Lattermann, Lexington/US<br />

25.3.3 Polarized Light Microscopy and ICRS Histological Assessments as Validated Methodologies for<br />

the Analysis of Tissue Quality in <strong>Cartilage</strong> <strong>Repair</strong> Randomized Controlled Trials<br />

S. Méthot 1 , W.D. Stanish 2 , R. McCormack 3 , F. Forriol Campos 4 , N. Mohtadi 5 , S. Pelet 6 ,<br />

J. Desnoyers 7 , A. Changoor 8 , N. Tran-Khanh 8 , S. Roberts 9 , M.S. Shive 1 , A. Restrepo 1 ,<br />

1 Laval/CA, 2 Halifax/CA, 3 Vancouver/CA, 4 Madrid/ES, 5 Calgary/CA, 6 Quebec City/CA, 7 Greenfield<br />

Park/CA, 8 <strong>Montreal</strong>/CA, 9 Shropshire/UK<br />

25.3.4 Nd:YAG laser therapy for the treatment of human chondral defects.<br />

B. Grigolo 1 , A. Zati 1 , G. Desando 1 , C. Cavallo 1 , D. Fortuna 2 , A. Facchini 1 , S. Giannini 1 , R.E.<br />

Buda 1 , 1 Bologna/IT, 2 Calenzano (Florence)/IT<br />

25.3.5 Recovery of isokinetic knee strength following matrix-induced autologous chondrocyte<br />

implantation (MACI)<br />

K. Hambly, J. Ebert, D.G. Lloyd, T.R. Ackland, D.J. Wood, Perth/AU<br />

25.3.6 Long term survival of autologous chondrocyte implantation - predictive factors of the<br />

survivorship.<br />

T. Paatela 1 , A.I. Vasara 1 , H. Nurmi 2 , I. Kiviranta 1, 3 , 1 Hus/FI, 2 Jyväskylä/FI, 3 Helsinki/FI<br />

25.3.7 Cost-Effectiveness Analysis of Autologous Chondrocyte Implantation: A Comparison of<br />

Periosteal Patch Versus Type I/III Collagen Patch<br />

E. Samuelson 1 , D.E. Brown 2 , 1 Ralston/US, 2 Omaha/US<br />

25.3.8 MR imaging results of particulated juvenile cartilage allograft for repair of chondral lesions in<br />

the knee<br />

J. Farr 1 , B.J. Cole 2 , S.K. Tabet 3 , G. Gold 4 , S. Vasanawala 4 , P. Reischling 5 , 1 Indianapolis/US,<br />

2 Chicago/US, 3 Albuquerque/US, 4 Stanford/US, 5 Austin/US<br />

25.3.9 Systematic Review and Meta-Analysis of Patient Reported Outcome Instruments Following<br />

Autologous Chondrocyte Implantation<br />

J.S. Howard, C. Lattermann, J.M. Hoch, C.G. Mattacola, J.M. Medina McKeon, Lexington/US


Programme: Tuesday, May 15, 2012<br />

Session 25.4 Free Paper<br />

Stem Cells 2<br />

11:30 <strong>–</strong> 13:00 Room: Grand Salon<br />

Moderators: Charles Archer (UK), Wael Kafienah (UK)<br />

25.4.1 Lysophoshatidic acid inhibits chondrogenic differentiation of human mesenchymal cells<br />

F. Petrigliano, D. McAllister, J.S. Adams, D. Evseenko, Los Angeles/US<br />

25.4.2 Equine mesenchymal stem cells lose their angiogenic properties when differentiated toward<br />

chondrogenic and osteogenic lineages.<br />

J.J. Bara 1 , E. Humphrey 1 , H. McCarthy 2 , W.E.B. Johnson 3 , S. Roberts 1 , 1 Oswestry/UK, 2 Cardiff/<br />

UK, 3 Birmingham/UK<br />

25.4.3 MicroRNA profiles during chondrocyte dedifferentiation and chondrogeneic differentiation of<br />

bone marrow derived MSC<br />

T.A. Karlsen, R.B. Jakobsen, J.H. Stendal, J.E. Brinchmann, Oslo/NO<br />

25.4.4 Comparison between progenitor cells from infrapatellar fat pad and subcutaneous adipose<br />

tissue: selecting a specific cell type for knee chondral applications<br />

S. Lopa, A. Colombini, L.J. Turner, L. De Girolamo, V. Sansone, M. Moretti, Milan/IT<br />

25.4.5 Genetic background affects induced pluripotent stem (iPS) cell generation<br />

L.V. Schnabel, C.M. Abratte, J.C. Schimenti, L.A. Fortier, Ithaca/US<br />

25.4.6 The importance of cell-cell interactions during in vitro differentiation of human<br />

chondroprogenitor cells<br />

K. Schrobback 1 , S. Stroebel 1 , B.S. Schon 1 , D. Hutmacher 2 , T.J. Klein 2 , T. Woodfield 1 ,<br />

1 Christchurch/NZ, 2 Brisbane/AU<br />

25.4.7 Sox Transcription Factors and TGF-Beta Gene Transduction Drives Pre-Transplant<br />

Chondrogenesis in Adult Bone-Marrow Derived Stromal Cells<br />

A.J. Nixon, J. Lui, A.E. Watts, Ithaca/US<br />

25.4.8 Genome-wide mapping of epigenetic changes during in vitro 3D chondrogenic differentiation of<br />

human bone marrow-derived mesenchymal stem cells<br />

S.R. Herlofsen 1 , J.C. Bryne 1 , T. Høiby 1 , L. Meza-Zepeda 1 , P. Collas 1 , T.S. Mikkelsen 2 ,<br />

J.E. Brinchmann 1 , 1 Oslo/NO, 2 Cambridge Massachusetts/US<br />

25.4.9 Development of scaffold-free tissue-engineered construct (TEC) with chondrogenic<br />

differentiation capacity using rabbit embryonic stem cell-derived mesenchymal stem cells<br />

Y. Moriguchi 1 , K. Shimomura 1 , T. Teramura 2 , W. Ando 3 , M. Sakaue 1 , H. Hasegawa 1 , N. Sugita 1 ,<br />

A. Myoui 1 , H. Yoshikawa 1 , N. Nakamura 1, 4 , 1Suita/JP, 2 Osaka Sayama/JP, 3 Hyogo/JP, 4 Osaka/JP<br />

ICRS General Board Meeting (New GB 2012/2013)<br />

13:30 <strong>–</strong> 14:30 Room: Chaudiere<br />

81


82<br />

Poster Sessions (Electronic & Traditional)<br />

Sunday, May 13, 2012 from 16:45 <strong>–</strong> 18:00 Rooms: Duluth/Richelieu<br />

Monday, May 14, 2012, from 16:30 <strong>–</strong> 17:30 Rooms: Duluth/Richelieu<br />

Poster presenters are required to stay near their poster boards during both poster sessions. Authors should encourage<br />

discussions with interested participants. The poster presenters should introduce themselves and be prepared<br />

to answer questions and initiate discussions.<br />

Allografts<br />

P1 The role of surgical insertion and pro-inflammatory cytokines in graft survival and metabolism<br />

S. Gitelis, R. Olewinski, S. Kirk, A.A. Hakimiyan, L. Rappoport, C. Pacione, B.J. Cole, M.A. Wimmer,<br />

S. Chubinskaya, Chicago/US<br />

P2 Chondrofix ® Osteochondral Allograft has Reduced Immunogenicity and Inflammatory Stimulation<br />

Z. Zhao, G. Ofek, T. Jiang, D.M. Squillace, R. Garrett, J. Gao, Austin/US<br />

P3 Outcome of Failed Osteochondral Allografts Treated with Revision Osteochondral Allografting<br />

M.T. Horton 1 , P.A. Pulido 1 , J.C. McCauley 2 , W.D. Bugbee 2 , 1 La Jolla, Ca/US, 2 La Jolla/US<br />

P4 Long Term Follow-Up of Fresh Osteochondral Allografting of the Femoral Condyle<br />

Y.D. Levy 1 , S. Görtz 1 , P.A. Pulido 2 , J.C. McCauley 2 , W.D. Bugbee 2 , 1 San Diego/US, 2 La Jolla/US<br />

P5 Functional evaluation of patients treated with bipolar fresh osteochondral allograft transplantation<br />

for post-traumatic ankle arthritis<br />

L. Berti, F. Vannini, M. Cavallo, G. Lullini, D. Luciani, A. Ruffilli, S. Giannini, Bologna/IT<br />

P6 Mid-term Results of the Treatment of <strong>Cartilage</strong> Defects in the Knee using Alginate Beads containing<br />

Human Mature Allogenic Chondrocytes<br />

A.A.M. Dhollander 1 , P.C. Verdonk 2 , S. Lambrecht 1 , R. Verdonk 2 , D. Elewaut 1 , G. Verbruggen 1 ,<br />

K.F. Almqvist 1 , 1 Gent/BE, 2 Gent-Zwijnaarde/BE<br />

P7 Effect of Allogenic Serum Addition to UW Solution for Prolonged Cold Preservation of Osteochondral Allografts<br />

K. Onuma 1 , K. Urabe 1 , K. Naruse 1 , K. Uchida 1 , K. Sukegawa 1 , T. Kenmoku 1 , R. Higashiyama 1 , M. Takaso 1 ,<br />

M. Itoman 2 , 1 Sagamihara/JP, 2 Kitakyushu/JP<br />

P8 Subchondral bone cysts increase buckling and stress formation along the cartilage-bone interface of<br />

osteochondral allografts<br />

G. Ofek, T. Jiang, Z. Zhao, D.M. Squillace, R. Garrett, J. Gao, Austin/US<br />

P9 Bipolar Fresh Osteochondral Allografting of the Tibiotalar Joint<br />

G. Khanna 1 , M.E. Brage 2 , M. Cavallo 3 , J.C. McCauley 4 , S. Görtz 5 , W.D. Bugbee 4 , 1 Baldwin Park, Ca/US,<br />

2 Seattle, Wa/US, 3 Bologna/IT, 4 La Jolla/US, 5 San Diego/US<br />

P10 Bipolar Fresh Total Osteochondral Allograft: Why, Where, When<br />

S. Giannini, R. Buda, M. Cavallo, A. Ruffilli, S. Neri, F. Vannini, Bologna/IT<br />

P11 Bone formation and osteointegration of the bone portion of Chondrofix ® Osteochondral Allografts: an<br />

experimental study in rabbits<br />

J. Gao, Z. Zhao, G. Ofek, D.M. Squillace, T. Jiang, R. Garrett, Austin/US<br />

P12 Chondrofix ® Osteochondral Allograft Does Not Affect Chondrocyte Viability in an In Vitro Tissue Culture Model<br />

T. Jiang, G. Ofek, Z. Zhao, D.M. Squillace, R. Garrett, J. Gao, Austin/US


Poster Sessions (Electronic & Traditional)<br />

P13 Hyaluronic acid (HA) Enhances Human Chondrocyte Viability in Cold-Stored Allografts<br />

A. Pearsall, A. Mates, K. Reed, G.L. Wilson, V. Grishko, Mobile/US<br />

P14 Mitochondrial Oxidative Stress Enhances MMP Activity in OA Chondrocytes<br />

A. Pearsall, K. Reed, G. Wilson, V. Grishko, Mobile/US<br />

P15 Treatment of knee defects with fresh-frozen massive osteochondral allografts<br />

A. Safi, R. Hart, Znojmo/CZ<br />

Animal Models<br />

P16 A simplified method to determine joint loading as a surrogate marker of pain/disability in a canine mo<br />

del of osteoarthritis, validated using force plate analyses as a gold standard.<br />

K. Wiegant, H.A.W. Hazewinkel, A. Doornenbal, A.D. Barten-Van Rijbroek, S.C. Mastbergen,<br />

F.P.J.G. Lafeber, Utrecht/NL<br />

P18 Intra Articular Therapy of Chondroitin Sulphate with Hydrogel Scaffold in Porcine <strong>Cartilage</strong> Defect Model<br />

K. Kannan 1 , R. Xiafei 1 , J.H.P. Hui 1 , E.H. Lee 1 , Z. Yang 1 , J. Gao 2 , H. Afizah 1 , G. Call 1 , J.Q. Yao 2 , T. Jiang 2 ,<br />

1 Singapore/SG, 2 Austin/US<br />

P19 Histomorphometric analysis of subchondral bone repair in a large animal osteochondral defect model<br />

J. Power, A. Getgood, N. Rushton, F. Henson, Cambridge/UK<br />

P20 Developed an animal model for intervertebral disc degeneration and regeneration<br />

C.-C. Niu 1 , L.-J. Yuan 1 , S.-S. Lin 1 , W.-J. Chen 2 , 1 Kweishan/TW, 2 Taoyuan/TW<br />

P21 Efficacy of common surgical compounds in the prevention of chondrocyte death in a bovine knee model<br />

A. Von Keudell, H. Syed, J. Canseco, A.H. Gomoll, Boston/US<br />

P22 A tissue engineering strategy for replacing the meniscus in a sheep model of OA.<br />

A. Izaguirre 1 , R. Gomez-Garcia 1 , G. Gonzalez 1 , H. Lecona 1 , H. Garcia-Campillo 1 , A. Ortiz 1 , I. Quiñones-<br />

Uriostegui 1 , L. Solis-Arrieta 1 , C. Velasquillo 2 , C. Pineda 1 , C. Ibarra 1 , 1 Mexico City/MX, 2 México/MX<br />

P23 A novel nano-structured porous polycaprolactone scaffold improves hyaline cartilage repair in a rabbit<br />

model compared to a collagen type I/III scaffold: in vitro and in vivo studies<br />

B.B. Christensen 1 , O.M. Hansen 2 , C.B. Foldager 1 , A. Krstiansen 2 , D.Q.S. Le 2 , J.V. Nygaard 2 ,<br />

A.H. Nielsen 3 , C. Bünger 1 , M. Lind 2 , 1 Aarhus/DK, 2 Aarhus C/DK, 3 Silkeborg/DK<br />

P24 The strong influence of the strength exercise on the cartilage constituents in animal model of osteoarthritis.<br />

S.M. Mattielo, F.A. Vasilceac, M.C. Souza, São Carlos/BR<br />

P25 Intra-articular blood coagulation aggravates joint damage after a bleed in a canine in vivo model<br />

M.E.R. Van Meegeren, G. Roosendaal, A.D. Barten-Van Rijbroek, R.E.G. Schutgens, S.C. Mastbergen,<br />

F.P.J.G. Lafeber, Utrecht/NL<br />

P26 The use of novel aragonite-hyluronate biphasic implant in chondral and osteochondral regeneration<br />

E. Kon 1 , D. Robinson 2 , Y. Chorev 3 , J. Shani 4 , K.R. Zaslav 5 , J.A. Eisman 6 , A.S. Levy 7 , G. Filardo 1 ,<br />

N. Altschuler 8 , 1 Bologna/IT, 2 Petah Tikwa/IL, 3 Netanya/IL, 4 Beit Berl/IL, 5 Richmond/US, 6 Sydney/<br />

AU, 7 Morristown/US, 8 Ariel/IL<br />

83


84<br />

Poster Sessions (Electronic & Traditional)<br />

P27 Novel poly(L/D)lactide scaffold for cartilage repair: a pilot study in a porcine chondral lesion model<br />

V. Muhonen 1 , E. Järvinen 1 , T. Paatela 1 , A.I. Vasara 1 , A. Meller 1 , V. Ellä 2 , M. Kellomäki 2 , I. Kiviranta 1 ,<br />

1 Helsinki/FI, 2 Tampere/FI<br />

Biomarkers<br />

P29 Hitting the Marker for Autologous Chondrocyte Implantation <strong>–</strong> is there a Role for Proteomics?<br />

K. Wright, H. Fuller, E. Humphrey, J.B. Richardson, P. Jones, A. Kerr, S. Roberts, 7ag/UK<br />

P30 A Quantitative Method for Evaluating <strong>Cartilage</strong> Defect <strong>Repair</strong>: Intra- and Inter- Reader Reproducibility<br />

J. Tamez-Pena 1, 2 , S. Trattnig 3 , S.M. Totterman 1 , P. Szomolanyi 3 , M.S. Shive 4 , M.D. Buschmann 5 ,<br />

A. Restrepo 4 , 1 Rochester/US, 2 Monterrey/MX, 3 Vienna/AT, 4 Laval/CA, 5 <strong>Montreal</strong>/CA<br />

P31 Markers of Success for Autologous Chondrocyte Implantation<br />

K. Wright, J.B. Richardson, P. Jones, H. Evans, A. Kerr, S. Roberts, 7ag/UK<br />

P32 Prohibitin a novel osteoarthritis biomarker for early detection and staging of OA patients<br />

A. Moreau 1 , C. Picard 1 , M. Taheri 2 , J.-F. Lavoie 1 , P. Lavigne 1 , 1 <strong>Montreal</strong>/CA, 2 Montréal/CA<br />

P33 The Collagen Type II Degradation Biomarker CTX-II In Athletes From Different Modalities<br />

P. Baches Jorge, G. Runco, A. Duarte, M. Vaz De Lima, P. Kertzman, A. Simões, N. Severino, São Paulo/BR<br />

P34 Inflammatory Cytokine Composition and Metabolic Profile of Post-Traumatic Ankle Joint Arthritis<br />

S. Adams, Jr. 1 , E. Kensicki 1 , L. Jones 2 , A. Haile 2 , S.D. Miller 2 , G. Guyton 2 , L. Schon 2 , 1 Durham/US, 2 Baltimore/US<br />

Biomaterials and Scaffolds<br />

P35 Temporal assessment of lysyl oxidase on tissue engineered articular cartilage.<br />

E.A. Makris, D.J. Responte, K. Athanasiou, Davis/US<br />

P36 Direct Human <strong>Cartilage</strong> <strong>Repair</strong> Using 3D Bioprinting Technology<br />

X. Cui, K. Breitenkamp, M.G. Finn, M.K. Lotz, D.D. D'Lima, La Jolla/US<br />

P37 Innovative approach for osteochondral defect using magnetic scaffold<br />

A. Russo 1 , S. Panseri 1 , T. Shelyakova 1 , M. Sandri 2 , C. Dionigi 1 , A. Strazzari 1 , A. Ortolani 1 , A. Tampieri 2 ,<br />

V. Dediu 1 , M. Marcacci 1 , 1 Bologna/IT, 2 Faenza/IT<br />

P38 Articular <strong>Cartilage</strong> Regeneration: Is there a role for a PVA PCL semi-IPN scaffold? Comparing a novel<br />

monophasic vs a novel biphasic scaffold vs cultured chondrocytes without scaffolds.<br />

V. Dutt 1 , B. Balakumar 1 , R. Karthikeyan 1 , N.M. Walter 1 , S. Chilbule 1 , V. Madhuri 1 , P. Nair 2 , 1 Vellore/<br />

IN, 2 Trivandrum/IN<br />

P39 Alternative macrophage activation and neutrophil chemotaxis induced in vivo by chitosan/blood im<br />

plants correlate with mast cell recruitment<br />

C.-H. Lafantaisie-Favreau 1 , D. Rusu 2 , P.E. Poubelle 2 , J. Sun 3 , C.D. Hoemann 1 , 1 <strong>Montreal</strong>/CA, 2 Quebec/<br />

CA, 3 Laval/CA<br />

P40 Arthroscopic meniscal scaffold implantation: early clinical results at 20 months of follow-up<br />

C. Zorzi, V. Condello, V. Madonna, F. Cortese, R. Giovarruscio, Negrar Vr/IT<br />

P41 Improvement and optimization of mechanical properties hyaluronic acid derivatives based hydrogels<br />

L. Wolfová, M. Pravda, M. Nemcová, M. Foglarová, V. Velebny, Dolní Dobrouc/CZ


Poster Sessions (Electronic & Traditional)<br />

P42 Bio-functional and bio-mimetic injectable hydrogels for cartilage repair<br />

L. Moreira Teixeira, J. Feijen, C. Van Blitterswijk, P. Dijkstra, M. Karperien, Enschede/NL<br />

P43 Employment of hydrogels and Nanomagnetics to direct the Formation of Organized Engineered Tissues<br />

in <strong>Cartilage</strong> Defects.<br />

S.P. Grogan 1 , C. Pauli 1 , H. Hoenecke 1 , C.W. Colwell 1 , M.K. Lotz 1 , C.B. Chung 2 , S. Jin 1 , D.D. D'Lima 1 ,<br />

1 La Jolla/US, 2 San Diego/US<br />

P44 How to treat Osteocondritis Dissecans of the Knee: surgical techniques and new trends<br />

E. Kon, F. Vannini, R. Buda, G. Filardo, M. Marcacci, S. Giannini, Bologna/IT<br />

P45 Nanostructured biomimetic scaffold for the treatment of osteochondral defects: pilot clinical study at 4<br />

years follow-up<br />

S. Patella, E. Kon, A. Di Martino, G. Filardo, F. Perdisa, B. Di Matteo, S. Zaffagnini, M. Marcacci, Bologna/IT<br />

P46 Does 3D micro-tissue assembly eliminate the need for cell-material interactions and improve engineered<br />

cartilage quality?<br />

B.S. Schon, T. Woodfield, Christchurch/NZ<br />

P47 Biocompatibility evaluation of Chitosan-Gelatin hydrogels with embedded PLGA particles as a useful tool<br />

for in vitro culture human chondrocytes<br />

Z.Y. Garcia Carvajal 1 , V. Martinez 2 , D. Garciadiego-Cazeres 3 , F.E. Villalobos Jr 2 , F.S. Arévalo 4 , E.F. Israel 4 ,<br />

M.A.C. Martinez 4 , C. Velasquillo 3 , A. Izaguirre 2 , R. Gomez 3 , F. Pérez 2 , G. Luna-Barcenas 5 ,<br />

R.A.M. Sanchez 5 , L. Solis-Arrieta 2 , C. Ibarra 1 , 1 Del. Tlalpan, Df/MX, 2 Mexico City/MX, 3 México/MX,<br />

4 Mexico, Df/MX, 5 Queretaro/MX<br />

P48 Multilayered Engineered Constructs for <strong>Cartilage</strong> Engineering<br />

C. Millan 1 , R. Mhanna 1 , K. Maniura 2 , Y. Yang 3 , T. Groth 3 , M. Zenobi-Wong 1 , 1 Zürich/CH, 2 St Gallen/CH,<br />

3 Halle/DE<br />

P49 Controlled Release of rhTGF-ß3 from Extracellular Matrix Membrane<br />

S.S. Yang 1 , M.S. Kim 1 , Y.J. Kim 1 , B.H. Choi 2 , S.R. Park 2 , B.-H. Min 1 , 1 Suwon/KR, 2 Incheon/KR<br />

P50 Characterization of Radiation Induced Chitosan Films with Acrylic and Silane Monomers: Fabrication of<br />

Resorbable Composites for Biomedical Application<br />

K. Dey, R.A. Khan, Dhaka/BD<br />

P51 Fabrication and Evaluation of Tissue Engineered Femoral Head Implants for Resurfacing of Osteoarthritic<br />

Joints<br />

F. Pfeiffer 1 , S.P. Franklin 1 , B.S. Bal 2 , J.L. Cook 2 , 1 Boonville/US, 2 Columbia/US<br />

P52 The benefits of a composite design for a Novel Polycarbonate-Urethane Meniscal Implant<br />

J.J. Elsner 1 , G. Zur 1 , E. Hershman 2 , R. Arbel 3 , A. Shterling 1 , F. Guilak 4 , E. Linder-Ganz 1 , 1 Netanya/IL,<br />

2 New York/US, 3 Tel Aviv/IL, 4 Durham/US<br />

P53 <strong>Cartilage</strong> Tissue Engineering with Human Young Chondrocyte and Porcine <strong>Cartilage</strong> ECM powder Scaffold<br />

H.J. Oh 1 , L.H. Jin 1 , Y.J. Kim 1 , J.-H. Cho 1 , B.H. Choi 2 , B.-H. Min 1 , 1 Suwon/KR, 2 Incheon/KR<br />

P54 Characterization of eggshell (membrane) derived biomaterials as potential scaffolds for cartilage tissue<br />

engineering<br />

M. Khanmohammadi, A. Baradar Khoshfetrat, S. Eskandarnezhad, S. Ebrahimi, Tabriz/IR<br />

85


86<br />

Poster Sessions (Electronic & Traditional)<br />

P55 Feasibility study of collagen type I/III membrane fixation in a human cadaver model of the distal<br />

radioulnar joint.<br />

E.A. Van Amerongen, N. Kaoui, J.E.J. Bekkers, A.H. Schuurman, L.B. Creemers, Utrecht/NL<br />

P56 Regeneration of Osteochondral Defects with Novel Multi-Layer Collagen-Based Scaffolds<br />

T.J. Levingstone, E. Thompson, A. Schepens, F.J. O'Brien, J.P. Gleeson, Dublin/IE<br />

P57 <strong>Cartilage</strong> Tissue Engineering with Human Young Chondrocyte and Porcine <strong>Cartilage</strong> ECM Powder Scaffold<br />

H.J. Oh 1 , L.H. Jin 1 , 2 , Y.J. Kim 2 , B.H. Choi 3 , J.-H. Cho 2 , B.-H. Min 1, 4 , 1 Suwon/KP, 2 Suwon/KR, 3 Incheon/<br />

KR, 4 Suwon/KI<br />

P58 A Pilot Study of the Use of an Osteochondral Scaffold Plug for <strong>Cartilage</strong> <strong>Repair</strong> in the Knee and How to<br />

Deal with Early Clinical Failures?<br />

A.A.M. Dhollander 1 , K. Liekens 1 , K.F. Almqvist 2 , R. Verdonk 2 , S. Lambrecht 2 , D. Elewaut 2 ,<br />

G. Verbruggen 2 , P.C. Verdonk 3 , 1 Ghent/BE, 2 Gent/BE, 3 Gent-Zwijnaarde/BE<br />

P59 Biological reconstruction in high grade osteonecrosis of the femoral head: results to a minimum of 2<br />

years of follow-up.<br />

D. Dallari, N. Del Piccolo, N. Rani, R. Fantasia, C. Carubbi, C. Stagni, Bologna/IT<br />

P60 Osteochondral scaffold implant in complex cases management<br />

A. Di Martino, E. Kon, G. Filardo, S. Patella, B. Di Matteo, F. Perdisa, M. Marcacci, Bologna/IT<br />

P61 Chitosan-Polyvinyl alcohol based scaffold for human auricular neocartilage.<br />

C. Velasquillo 1 , E. Abarca 2 , E. Ruvalcaba 1 , Z. Garcia 1 , V. Martinez 3 , L. Solis-Arrieta 1 , M. Perez 1 , C. Ibarra 3 ,<br />

G. Luna-Barcenas 1 , 1 Mexico/MX, 214389/MX, 3 México/MX<br />

P62 Implantation of a thin double-network gel sheet can induce spontaneous articular cartilage regeneration<br />

in vivo in a large osteochondral defect<br />

N. Kitamura 1 , H. Matsuda 1 , T. Kurokawa 1 , K. Arakaki 2 , J.P. Gong 1 , F. Kanaya 2 , K. Yasuda 1 , 1 Sapporo/JP, 2 Naha/JP<br />

P63 A Novel Treatment for Osteonecrotic Femoral Head <strong>–</strong> An Animal Study<br />

C.-J. Liao 1 , C.-C. Jiang 2 , H. Chiang 2 , W.-H. Chang 1 , 1 Hsinchu/TW, 2 Taipei/TW<br />

P64 Reconstruction of Osteochondral Lesions of the Talus with Autologous Spongiosa Graft and Autologous<br />

Matrix Induced Chondrogenesis (AMIC)<br />

M. Wiewiorski, M. Miska, V. Valderrabano, Basel/CH<br />

P65 New biodegradable and biocompatible synthetic scaffold for meniscal regeneration: prospective<br />

evaluation at 2 years follow-up<br />

S. Patella, A. Di Martino, E. Kon, G. Filardo, G.M. Marcheggiani Muccioli, F. Iacono, S. Zaffagnini,<br />

M. Marcacci, Bologna/IT<br />

P66 One step cartilage repair by PVA-H hydrogel implants in the knee: long term results<br />

F.V. Sciarretta, Rome/IT<br />

P68 Biological Reconstruction in Osteo-Chondral Lesion of Talus<br />

D. Dallari, N. Rani, N. Del Piccolo, M. Filanti, G. Sabbioni, C. Stagni, Bologna/IT<br />

P69 Treatment of osteochondritis dissecans of the knee with the nanostructured biomimetic scaffold<br />

combined with platelet-rich plasma gel<br />

W. Widuchowski, P. Lukasik, W. Wawrzynek, R. Kokot, B. Koczy, J. Widuchowski, Piekary Slaskie/PL


Poster Sessions (Electronic & Traditional)<br />

P70 Articulating cartilage versus pHEMA hydrogel. A tribological and histological "in vitro" study<br />

J.L. Sague, B. Andreatta, R. Egli, Y. Loosli, R. Luginbühl, Bettlach/CH<br />

Biomechanics<br />

P71 Frictional response of cartilage after prolonged walking and recovery <strong>–</strong> an in-vitro study<br />

S. Taylor, J. Ingram, E. Ingham, J. Fisher, S. Williams, Leeds/UK<br />

P72 Arthroscopic fixation of matrix associated Autologous Chondrocyte Implantation - Joint compression<br />

forces following biodegradable pin fixation<br />

M. Herbort 1 , S. Zelle 1 , M. Raschke 1 , W. Petersen 2 , T. Zantop 3 , 1 Muenster/DE, 2 Berlin/DE, 3 Straubing/DE<br />

P73 Hypoxia as a possible method of promoting collagen crosslinking in native articular cartilage<br />

E.A. Makris, K. Athanasiou, Davis/US<br />

P74 Flow-perfusion suppresses hypertrophic differentiation of novel MSC-based cartilage constructs<br />

D. Gawlitta, J. Malda, W.J.A. Dhert, Utrecht/NL<br />

P75 Stress relaxation due to pore formation in cartilage under mechanical load and laser heating<br />

A. Shnirelman 1 , E. Sobol 2 , A. Omelchenko 2 , 1 <strong>Montreal</strong>/CA, 2 Troitsk/RU<br />

P76 In-vitro stability testing of a non-fixed meniscal implant: the effect of surgical technique and knee condition<br />

J.J. Elsner 1 , T.F. Bonner 2 , A. Greene 1 , R.R. Gupta 2, R.S. Butler 2, E. Linder-Ganz 1 , E. Hershman3, R.W. Colbrunn2 , 1Netanya/IL, 2Cleveland/US, 3New York/US<br />

P77 Aetiology of experimentally induced osteoarthritis of the knee; biomechanical or biochemical factors?<br />

K. Wiegant, M. Beekhuizen, S.C. Mastbergen, A.D. Barten-Van Rijbroek, D.B.F. Saris, L.B. Creemers,<br />

F.P.J.G. Lafeber, Utrecht/NL<br />

P78 Characterisation of the Regional Osteochondral Properties of Quadruped Tibio-Femoral Joints<br />

S.W.D. McLure, P. Conaghan, J. Fisher, S. Williams, Leeds/UK<br />

<strong>Cartilage</strong><br />

P79 One-stage cartilage defect treatment combining chondrons and mesenchymal stromal cells; in vitro and<br />

in vivo results<br />

J.E.J. Bekkers, L. Vonk, W.J.A. Dhert, L.B. Creemers, D.B.F. Saris, Utrecht/NL<br />

P80 Influence of cryopreservation, cultivation time and patient’s age on the gene expression in cartilage transplants<br />

C. Albrecht, B. Tichy, S. Hosiner, L. Zak, S. Aldrian, S. Nürnberger, S. Marlovits, Vienna/AT<br />

P81 Ex-vivo gene therapy-induced cartilage regeneration: comparison of different subpopulations of primary<br />

muscle-derived cells<br />

H. Li, M. Poddar, A. Usas, J. Huard, Pittsburgh/US<br />

P82 ACI in the management of Bilateral knee chondral defects<br />

A. Von Keudell, M. Berninger, T. Bryant, T. Minas, Boston/US<br />

P83 S100 - a biomarker for human articular chondrocyte potency<br />

C. Czücs, J. Diaz-Romero, A. Quintin, D. Nesic, Bern/CH<br />

P84 Expression of Collagen I in macroscopically healthy cartilage. Useful for tissue engineering?<br />

C. Velasquillo, A. Lopez-Reyes, I. Alba-Sanchez, C. Ortega-Sanchez, M. Santamaria-Olmedo,<br />

V. Martinez-Lopez, L. Tamay De Dios, A. Izaguirre, C. Ibarra, Mexico/MX<br />

87


88<br />

Poster Sessions (Electronic & Traditional)<br />

P85 An Autologous <strong>Cartilage</strong> Tissue Implant (ACTI) NeoCart® for Treatment Grade III Chondral Injury to the<br />

Femur. Intermediate Term Results from Initial FDA Trials.<br />

D.C. Crawford 1 , T.M. Deberardino 2 , C.T. Moorman 3 , D. Taylor 3 , B. Nelson 4 , J. Chesnutt 1 , C.B. Ma 5 ,<br />

R.J. Williams, Iii 6 , 1 Portland/US, 2 Farmington/US, 3 Durham/US, 4 Minneapolis/US, 5 San Francisco/<br />

US, 6 New York/US<br />

P86 Treatment of osteochondritis dissecans of the knee with arthroscopic Bone Marrow-Derived Cells<br />

transplantation (“one step” technique): Results and T2 mapping evaluation<br />

R. Buda, F. Vannini, M. Battaglia, M. Cavallo, G. Pagliazzi, A. Ruffilli, S. Giannini, Bologna/IT<br />

P87 Autologous chondrocyte Implantation (ACI) with a bilayer collagen membrane and bone graft in the<br />

knee: 2-8 year follow-up study<br />

S. Vijayan, W. Bartlett, G. Bentley, R. Carrington, J. Skinner, R. Pollock, M. Alorjani, T. Briggs, Stanmore/UK<br />

P88 Two-year follow up from arthroscopic implantation of matrix-seeded autologous chondrocytes at the knee<br />

J.C. Ibarra, F.E. Villalobos Córdova, A. Izaguirre, V. Guevara, C. Velasquillo, S. Cortes, D. Chavez, F. Pérez,<br />

V. Martinez, L.G. Ibarra, Mexico City/MX<br />

P89 In vitro preculture enhances the in vivo cartilage forming capacity of constructs generated by combining<br />

human bone marrow stromal cells with a low fraction of human articular chondrocytes in collagen scaffold<br />

M.-A. Sabatino 1 , R. Santoro 2 , S. Gueven 2 , D. Wendt 2 , I. Martin 2 , M. Moretti 1 , A. Barbero 2 , 1 Miland/IT, 2 Basel/CH<br />

P90 Comparison between articular chondrocytes from osteoarthritis knees and mesenchymal stem cells from<br />

young healthy donors in an alginate scaffold model of in vitro chondrogenesis<br />

A.M. Fernandes, S.R. Herlofsen, T.A. Karlsen, A.M. Küchler, J.E. Brinchmann, Oslo/NO<br />

P91 Matrix assisted chondrocyte implantation (MACI ® ) improves healing of full thickness cartilage defects in<br />

an equine model<br />

A.J. Nixon 1 , H. Sparks 1 , W. Linnenkohl 1 , L. Begum 1 , J. Hart 1 , N. Moran 2 , G. Matthews 2 , 1 Ithaca/US, 2 Farmingham/US<br />

P92 Autologous Chondrocyte Implantation Augments the Production of Clusterin in <strong>Repair</strong> <strong>Cartilage</strong><br />

H. McCarthy 1 , J. Malda 2 , J.B. Richardson 1 , S. Roberts 1 , 1 Oswestry/UK, 2 Utrecht/NL<br />

P93 One Step <strong>Repair</strong> In Talar Osteochondral Lesions: 4 Years Results And T2-mapping Capability In<br />

Outcome Prediction<br />

F. Vannini, R. Buda, M. Battaglia, M. Cavallo, L. Ramponi, A. Ruffilli, A. Timoncini, S. Giannini, Bologna/IT<br />

P94 Arthroscopic matrix-induced autologous chondrocyte implantation: 24 month clinical and radiological outcomes<br />

J. Ebert, K. Hambly, M. Fallon, D.J. Wood, G. Janes, Perth/AU<br />

P95 Five-year clinical and radiological outcome following matrix-induced autologous chondrocyte<br />

implantation (MACI) in the patellofemoral joint<br />

J. Ebert, K. Hambly, D. Meyerkort, M. Fallon, T.R. Ackland, M.H. Zheng, D.J. Wood, Perth/AU<br />

P96 Using Partial Enzyme Digestion to Facilitate the Fusion of <strong>Cartilage</strong> Interface<br />

C.-J. Liao 1 , C.-C. Jiang 2 , H. Chiang 2 , W.-H. Chang 1 , W.-J. Liao 1 , 1 Hsinchu/TW, 2 Taipei/TW<br />

P97 Getting closer to hyaline repair - Xenograft chondrocytes<br />

G. Maor, G. Nierenberg, Haifa/IL


Poster Sessions (Electronic & Traditional)<br />

P98 Behaviour of adult human articular chondrocytes during in vivo culture in semipermeable cell chambers.<br />

M. Polacek 1 , T. Annala 2 , I. Martinez 1 , 1 Tromsø/NO, 2 Tampere/FI<br />

P99 Biologic Arthroplasty In Patello-Femoral <strong>Cartilage</strong> Lesions<br />

A. Gobbi, A. Kumar, G. Karnatzikos, Milan/IT<br />

P100 CARTIPATCH ® phase III clinical trial: Comparison of autologous chondrocytes implantation versus<br />

Mosaicplasty, 1 year follow-up<br />

P. Neyret 1 , J.-F. Potel 2 , H. Robert 3 , C. Bussiere 4 , E. Servien 1 , F. Dubrana 5 , 1 Lyon/FR, 2 Toulouse/<br />

FR, 3 Mayenne/FR, 4 Dracy Le Fort/FR, 5 Brest/FR<br />

P101 Articular cartilage repair with magnetically labeled mesenchymal stem cells and external magnetic device<br />

G. Kamei, N. Adachi, M. Deie, T. Kobayashi, H. Shibuya, S. Ohkawa, K. Takazawa, W. Kongcharoensombat,<br />

M. Ochi, Hiroshima/JP<br />

P102 Evaluation of a mesenchymal stem cell seeded PEOT/PBT scaffold in an osteochondral defect in vivo.<br />

K.M. Salih Mohamed 1 , V. Barron 1 , A. Manian 1 , A. Nandakumar 2 , L. Moroni 2 , P. Habibovic 2 , F. Shannon 1 ,<br />

M. Murphy 1 , F. Barry 1, 1 Galway/IE, 2 Twente/NL<br />

P103 Autologous nasal chondrocytes and a cellulose-based self-setting hydrogel for the repair of articular<br />

cartilage in horses<br />

C. Vinatier, E. Lallemand, O. Geoffroy, C. Merceron, O. Gauthier, B.H. Fellah, S. Portron, M. Masson,<br />

J. Lesoeur, P. Weiss, J. Guicheux, Nantes/FR<br />

P104 <strong>Cartilage</strong> of different mammalian species demonstrate variable tissue thickness, cell density,<br />

proteoglycan content and cartilage specific genes<br />

T. Zaman, N.B. Kamisan, S. Naveen, R.E. Ahmad, Kuala Lumpur/MY<br />

P105 Effect of intra-articular Hyaluronic acid, mesenchymal stem cells and microfracture in the treatment of<br />

cartilage defects <strong>–</strong> In vivo study<br />

T. Zaman, P.N. Shilpa, P. Wagner, N. Huiyin, K. Puvanan, Kuala Lumpur/MY<br />

P106 Chondrogenic differentiation of bone marrow-derived MSCs with the high affine GDF-5 variant (R57A) in vitro<br />

F. Gilbert 1 , M. Kunz 1 , J. Nickel 2 , M. Rudert 1 , A.F. Steinert 1 , 1 Wuerzburg/DE, 2 Würzburg/DE<br />

P107 Instant CEMTROCELL ® , a new approach based on autologous chondrocyte implantation for the<br />

treatment of cartilage lesions.<br />

P. Guillen-Garcia 1 , E. Rodriguez Iñigo 1, 2 , M. Guillen-Vicente 1 , I. Guillen-Vicente 1 , E. Santos 1 ,<br />

R. Caballero-Santos 1 , 2, J.M. Lopez-Alcorocho 1, 2 , 128045/ES, 2 Madrid/ES<br />

P108 One-Year Clinical and Radiological Results of a Prospective, Investigator-Initiated Trial Examining a<br />

Novel, Purely Autologous 3-Dimensional Autologous Chondrocyte Transplantation Product in the Knee<br />

S. Fickert 1 , P. Gerwien 1 , B. Helmert 1 , T. Schattenberg 1 , S. Weckbach 2 , M. Kaszkin-Bettag 1 , L. Lehmann 1 ,<br />

1 Mannheim/DE, 2 Munich/DE<br />

P109 Mechanical loading reduces chondrocyte death after impact injury in porcine model<br />

L. Vernon 1 , D. Wilensky 2 , D. Ajibade 2 , L.D. Kaplan 1, 2, C.-Y.C. Huang 1 , 1 Coral Gables/US, 2 Miami/US<br />

P110 Feasibility of an arthroscopic 3-dimensional, purely autologous chondrocyte transplantion for the<br />

treatment for full thickness chondral lesions in the hip caused by femoroacetabular impingement<br />

S. Fickert, S. Thier, Mannheim/DE<br />

89


90<br />

Poster Sessions (Electronic & Traditional)<br />

P111 Biological resurfacing techniques, fully arthroscopically performed, for degenerative multifocal chondral<br />

defects, at the knee. Mid term results<br />

S. Alevrogiannis, G. Skarpas, Athens/GR<br />

P112 Coefficient of Friction of Poly(vinyl alcohol) Hydrogels Against Swine Articular <strong>Cartilage</strong><br />

H. Bodugoz Senturk, D. Ling, S. Nanda, H.L. Kluk, G. Braithwaite, O.K. Muratoglu, Boston/US<br />

P113 Protection of chondrocyte phenotype by heat inactivation of serum in monolayer expansion cultures.<br />

M. Matmati, T.F. Ng, T.M. Quinn, <strong>Montreal</strong>/CA<br />

P114 Evaluation of the clinical results and MRI of a novel II generation autologous chondrocytes implant, for<br />

cartilage repair in Knees and Ankles.<br />

R. Arbel, Tel Aviv/IL<br />

P115 Nonclinical Efficacy Tests of Pellet-Type Autologous Chondrocytes for Regeneration of Articular <strong>Cartilage</strong><br />

J.-Y. Lee 1 , J. Lee 1 , B.C. Chae 1 , Y. Son 2 , 1 Seoul/KR, 2 Yongin/KR<br />

P116 Ten year follow up of matrix associated autologous chondrocyte transplantation in the knee<br />

M. Brix 1 , C. Chiari 1 , S. Nehrer 2 , R. Windhager 1 , S. Domayer 1 , 1 Vienna/AT, 2 Krems/AT<br />

Chondrocytes<br />

P117 Enhanced cartilage repair in defects implanted with autologous chondrocytes transduced with AAV5-<br />

IGF-I in the equine model<br />

K. Ortved, L. Begum, M. Scimeca, A.J. Nixon, Ithaca/US<br />

P118 Differential Effects of Cyclooxygenase-1 and -2 specific NSAIDs on Chondrogenic Differentiation<br />

M.M.J. Caron, P. Emans, D.A.M. Surtel, A. Cremers, D. Ophelders, K. Sanen, L.W. Van Rhijn, T.J.M. Welting,<br />

Maastricht/NL<br />

P119 c-Maf Regulates ADAMTS-12 Expression in Human Chondrocytes<br />

E. Hong, J. Yik, D. Amanatullah, P. Di Cesare, D.R. Haudenschild, Sacramento/US<br />

P120 Inhibition of Cyclooxygenase-2 Impacts Chondrocyte Hypertrophy<br />

M.M.J. Caron, P. Emans, M.P.F. Janssen, K. Sanen, M.M.E. Coolsen, L. Voss, D.A.M. Surtel, A. Cremers,<br />

J.-W. Voncken, T.J.M. Welting, L.W. Van Rhijn, Maastricht/NL<br />

P121 BMP-2 and BMP-7: Differential Regulation of Chondrogenic Differentiation<br />

M.M.J. Caron, T.J.M. Welting, M.M.E. Coolsen, D.A.M. Surtel, A. Cremers, L.W. Van Rhijn, P. Emans,<br />

Maastricht/NL<br />

P122 Extendable Surfaces for Dynamic Culture of Chondrocytes: Surface Functionalization with a<br />

Decellularized <strong>Cartilage</strong> ECM Extract to Enhance Chondrogenic Phenotype.<br />

D.H. Rosenzweig, S. Solar-Cafaggi, T.M. Quinn, <strong>Montreal</strong>/CA<br />

P123 Spatial and Temporal Apoptotic Activity in Mechanically Injured <strong>Cartilage</strong> Explants<br />

D.H. Rosenzweig, T.M. Quinn, <strong>Montreal</strong>/CA<br />

P124 Differential Responses of Normal and OA Human Chondrocytes to Local Anesthetics: Role of Oxidative Stress<br />

A. Pearsall, A. Mates, G. Wilson, V. Grishko, Mobile/US<br />

P125 Long-term patient satisfaction and clinical outcome of autologous chondrocyte implantation in the knee joint<br />

M.N. Dugard, J.C. Parker, E. Robinson, J.H. Kuiper, S. Roberts, J.B. Richardson, Oswestry/UK


Poster Sessions (Electronic & Traditional)<br />

P126 Hyaluronic acid decreases proliferation time and improves redifferentation of isolated chondrocytes<br />

K. Wijnands 1 , E.J.P. Jansen 2 , T.J.M. Welting 1 , D.A.M. Surtel 1 , L.W. Van Rhijn 1 , P. Emans 1 , 1 Maastricht/<br />

NL, 2 Maastricht And Sittard/NL<br />

P127 Inhibition of Catabolic Responses in Rat Chondrocytes via Delivery of Interleukin-1 Receptor Antagonist<br />

Protein (IRAP) Using Adeno-Associated Virus (AAV)<br />

H.H. Lee 1 , X. Xiao 2 , C. Chu 1 , 1 Pittsburgh/US, 2 Chapel Hill/US<br />

P128 Enhanced Tissue Regeneration Potential of Juvenile Articular <strong>Cartilage</strong><br />

H. Liu, Z. Zhao, R. Clarke, J. Gao, R. Garrett, Austin/US<br />

P130 Expression of PPARa, b, and g in the Hartley guinea pig model of primary osteoarthritis<br />

F.E. El Mansouri, S.S. Nebbaki, N. Zayed, M. Benderdour, J. Martel-Pelletier, J.-P. Pelletier, H. Fahmi, <strong>Montreal</strong>/CA<br />

P131 Influence of Growth Factors for Chondrogenic Re-differentiation under Normoxic and Hypoxic<br />

Culture Conditions<br />

A. Jonitz, K. Lochner, D. Hansmann, T. Tischer, R. Bader, Rostock/DE<br />

P132 10- years experience in Autologous Disc Chondrocyte Tranplantation (ADCT) A critical clinical review<br />

of 115 cases<br />

C. Hohaus 1 , H.J. Meisel 1 , T. Ganey 2 , U. Mansmann 3 , 1 Halle/DE, 2 Atlanta/US, 3 Muenchen/DE<br />

P133 Global gene array comparison between successful and failed outcome after autologous chondrocyte<br />

implantation<br />

J. Stenberg 1 , T.S.S. De Windt 2 , J. Van Der Lee 1 , M. Brittberg 3 , D.B.F. Saris 2 , A. Lindahl 1 , 1 Gothenburg/SE,<br />

2 Utrecht/NL, 3 Kungsbacka/SE<br />

P134 The Use of Talus Osteochondral Defect <strong>Cartilage</strong> for Chondrocyte Harvesting: Results of 151<br />

Consecutive Patients<br />

C. Kreulen 1, 2 , E. Giza 2 , J. Kim 2 , M. Sullivan 1 , 1 Sydney/AU, 2 Sacramento/US<br />

P135 Effect of SOX9 and IGF-I overexpression over some extracellular matrix components in cultivated human<br />

articular chondrocytes.<br />

M. Simental, J. Lara, A. Soto, E. Álvarez, J. Sepúlveda, H.G. Martínez, Monterrey/MX<br />

Clinical Outcome<br />

P138 Chondropenia Severity Score: An Arthroscopic Stratification tool of Structural <strong>Cartilage</strong> Changes in the<br />

Knee as Correlated to Patient Reported Outcomes.<br />

S.L. Pro 1 , B.W. Blatz 2 , T.R. McAdams 3 , B. Mandelbaum 2 , 1 Tigard/US, 2 Santa Monica/US, 3 Palo Alto/US<br />

P139 Single V/S Double Bundle ACL Reconstruction: Functional Outcome At 3 Years In Athletes<br />

A. Gobbi, A. Kumar, G. Karnatzikos, Milan/IT<br />

P140 AMIC or ACI for arthroscopic repair of delaminated acetabular cartilage in femoroacetabular impingement.<br />

A. Fontana, Milano/IT<br />

P141 Arthroscopic treatment of ankle anterior bony impingement: the long term clinical outcome<br />

F. Vannini, R. Buda, A. Parma, G. Pagliazzi, M. Cavallo, A. Ruffilli, S. Giannini, Bologna/IT<br />

P142 A Phase I/II study of a tissue engineered cartilage implant derived from allogeneic juvenile chondrocytes:<br />

3 year results<br />

B.J. Cole 1 , J. Farr 2 , K. Bonner 3 , G. Gold 4 , H.D. Adkisson 5 , P. Reischling 6 , 1 Chicago/US, 2 Greenwood/<br />

US, 3 Virginia Beach/US, 4 Stanford/US, 5 St. Louis/US, 6 Austin/US<br />

91


92<br />

Poster Sessions (Electronic & Traditional)<br />

P143 Strategies for Patient Profiling in Articular <strong>Cartilage</strong> <strong>Repair</strong> of the Knee: A Prospective Cohort of Patients<br />

Treated by One Experienced <strong>Cartilage</strong> Surgeon<br />

T.S.S. De Windt 1 , S. Concaro 2 , A. Lindahl 3 , D.B.F. Saris 1 , M. Brittberg 4 , 1 Utrecht/NL, 2 Kungälv/SE,<br />

3 Göteborg/SE, 4 Kungsbacka/SE<br />

Extracellular Matrix<br />

P145 Evaluation of the type II collagen expression in the extracellular matrix of chondrocyte by atomic force<br />

microscopy<br />

C.-H. Hsieh, C.-H. Liu, S. Lin, J.-J. Tsai-Wu, H. Chiang, S. Chen, C.-C. Jiang, Taipei/TW<br />

P146 Effects of Different Three-dimensional Conditions on Chondrocyte and MSC Capacity to Regenerate<br />

Cartilaginous Tissue<br />

S.P. Grogan, X. Chen, S. Sovani, T. Olee, M.K. Lotz, D.D. D'Lima, La Jolla/US<br />

Growth Factors and Cytokines<br />

P147 The effect of retroviral-mediated overexpression of BMP-2 on hMSCs during monolayer proliferation<br />

A. Neumann 1 , M. Alini 1 , M. Anton 2 , C. Archer 3 , M. Stoddart 1 , 1 Davos Platz/CH, 2 Muenchen/DE, 3 Cardiff/UK<br />

P149 Intermittent but not continuous exposure to recombinant human fibroblast growth factor-18 (rhFGF18)<br />

promotes chondrocyte anabolism and phenotype in 3D culture.<br />

A. Gigout, S. Lindemann, H. Guehring, Darmstadt/DE<br />

P150 TGFß during expansion phase negatively affects the chondrogenic redifferentiation of human chondrocytes<br />

R. Narcisi 1, 2 , L. Signorile 1 , P. Giannoni 2 , G.J.V.M. Van Osch 1 , 1 Rotterdam/NL, 2 Genova/IT<br />

P151 The Role of IL-6 in Osteoarthritis and <strong>Cartilage</strong> Regeneration<br />

A.I. Tsuchida 1 , M. Beekhuizen 1 , A.G.J. Bot 1 , B. Geurts 1 , J.E.J. Bekkers 1 , W.J.A. Dhert 1 , L.B. Creemers 1 ,<br />

D.B.F. Saris 1, 2, 1 Utrecht/NL, 2 Enschede/NL<br />

P152 <strong>Repair</strong> of Rabbit Osteochondral Defects by an Acellular Technique Using an Ultrapurified Alginate Gel<br />

Containing Stromal Cell-Derived Factor-1<br />

A. Sukegawa, N. Iwasaki, Y. Kasahara, T. Onodera, T. Igarashi, A. Minami, Sapporo/JP<br />

P153 A short time window to profit from IL-4 plus IL-10 addition to protect cartilage from blood-induced<br />

damage in vitro<br />

M.E.R. Van Meegeren, G. Roosendaal, J.A.G. Van Roon, S.C. Mastbergen, F.P.J.G. Lafeber, Utrecht/NL<br />

P154 Direct BMP-2 Gene Delivery to Osteochondral Defects using Coagulated Bone Marrow Aspirate - Potent<br />

<strong>Cartilage</strong> Regeneation with the Risk of Osteophyte Formation in a Rabbit Model<br />

J. Sieker 1 , M. Kunz 2 , M. Weissenberger 2 , F. Gilbert 2 , C.H. Evans 3 , M. Rudert 2 , A.F. Steinert 2 , 1 Würzburg/DE,<br />

2 Wuerzburg/DE, 3 Boston/US<br />

P156 CXCL8 in osteoarthritis, symptomatic cartilage defects and cartilage regeneration<br />

A.I. Tsuchida 1 , D.B.F. Saris 1, 2 , B. Geurts 1 , A.H.M. Kragten 1 , M. Beekhuizen 1 , J.E.J. Bekkers 1 , W.J.A.<br />

Dhert 1 , L.B. Creemers 1 , 1 Utrecht/NL, 2 Enschede/NL<br />

Histology<br />

P157 The Effect of Recombinant Human Fibroblast Growth Factor-18 on Articular <strong>Cartilage</strong> Following Single<br />

Impact Load<br />

L. Barr, A. Getgood, N. Rushton, F.M. Henson, Cambridge/UK


Poster Sessions (Electronic & Traditional)<br />

P158 A Stereological Method for the Quantitative Evaluation of <strong>Cartilage</strong> <strong>Repair</strong> Tissue<br />

C.B. Foldager 1, 2 , J.R. Nyengaard 1 , M. Lind 1 , M. Spector 2 , 1 Aarhus/DK, 2 Boston/US<br />

P159 Healing of cartilage lesions in sheep after microfracturing technique and transcutaneous application of<br />

non-steroidal anti-inflammatory or chondroprotective medication - histological evaluation<br />

M. Sidler, N. Fouche, I. Meth, B. Von Rechenberg, P. Kronen, Zurich/CH<br />

Imaging<br />

P160 Comparison of Three Methods to Quantify <strong>Repair</strong> <strong>Cartilage</strong> Collagen Orientation<br />

K. Ross 1 , H.G. Potter 2 , R. Williams 1 , L.V. Schnabel 1 , G. Bradica 3 , E. Castiglione 3 , S.L. Pownder 1, 2 ,<br />

P.W. Satchell 1 , L.A. Fortier 1 , 1 Ithaca/US, 2 New York/US, 3 Exton/US<br />

P161 High resolution diagnosis of cartilage and meniscus damage with a novel extremity-CBCT.<br />

J. Salo, H. Kokkonen, H. Kröger, J.S. Jurvelin, J. Töyräs, Kuopio/FI<br />

P162 <strong>Cartilage</strong> assessment after mosaicplasty with delayed gadolinium-enhanced MRI of the cartilage (dGEMRIC)<br />

M. Kobayashi, T. Shirai, S. Nakamura, R. Arai, K. Nishitani, T. Satake, H. Kuroki, Y. Nakagawa,<br />

T. Nakamura, Kyoto/JP<br />

P163 Normative Articular <strong>Cartilage</strong> T2 Values in Clinically Relevant Subregions of the Knee<br />

E. Lucas, R. Surowiec, J..E. Giphart, E. Fitzcharles, B. Petre, C. Ho, Vail/US<br />

P164 Compositional Observation of <strong>Cartilage</strong> <strong>Repair</strong> Tissue by Magnetic Resonance Imaging <strong>–</strong> a Clinically<br />

Oriented Approach<br />

G.H. Welsch 1, 2 , S. Apprich 2 , D. Stelzeneder 2 , M. Blanke 1 , S. Marlovits 2 , S. Trattnig 2 , 1 Erlangen/<br />

DE, 2 Vienna/AT<br />

P166 <strong>Cartilage</strong> <strong>Repair</strong> of the Ankle: First Results of T2 mapping at 7.0 Tesla after Microfracture and Matrix<br />

associated autologous <strong>Cartilage</strong> Transplantation<br />

S. Apprich, M. Brix, M. Sokolowski, R. Windhager, S. Trattnig, S. Domayer, Vienna/AT<br />

P167 Can x-ray morphometric parameters for the hip be assessed on magnetic resonance imaging?<br />

A. Hingsammer, D. Stelzeneder, Y.-J. Kim, Boston/US<br />

P168 Development of central osteophytes in native cartilage: an observational MR imaging study<br />

F.A. Sakamoto, C.S. Winalski, E. Schneider, J.P. Schils, Cleveland/US<br />

P169 Application of micro-CT for non-destructive quality control in engineered cartilage<br />

L.H. Jin 1 , Y.J. Kim 1 , B.H. Choi 2 , H.J. Oh 1 , T.Z. Li 1 , B.-H. Min 1 , 1 Suwon/KR, 2 Incheon/KR<br />

P170 Utilizing <strong>Cartilage</strong> T2 Relaxation Time in Clinical Trials<br />

J. Riek, Rochester/US<br />

Intervertebral Disc<br />

P171 Intervertebral disc regeneration after implantation of a cell-free bioresorbable implant in an ovine disc<br />

degeneration model.<br />

M. Endres, U. Freymann, A. Abbushi, M.L. Zenclussen, P. Casalis, C. Woiciechowsky, C. Kaps, Berlin/DE<br />

P172 Viscoelastic moduli of the nucleus pulposus decrease with mild degeneration in a goat lumbar<br />

intervertebral disc model<br />

S.E.L. Detiger, R.J.W. Hoogendoorn, A.J. Van Der Veen, B.J. Van Royen, M.N. Helder, G.H. Koenderink,<br />

T.H. Smit, Amsterdam/NL<br />

93


94<br />

Poster Sessions (Electronic & Traditional)<br />

P173 An immunohistochemical investigation of progenitor cell markers in the human intervertebral disc.<br />

S. Turner, C. Morgan, J. Trivedi, S. Roberts, Oswestry/UK<br />

P174 The Microscopic Pictures of Degenerative Intervertebral Disc with Histochemical Methods<br />

C.-M. Shih 1 , K.-C. Liu 2 , J.-T. Chien 2 , S.-F. Lai 2 , T.-Y. Tsai 2 , 1 Taichung City/TW, 2 Chiayi County/TW<br />

Meniscus<br />

P175 Marrow Stimulation Improves Healing following Avascular Zone Meniscal Injury in a Rabbit Model<br />

M.D. Driscoll 1 , B.N. Robin 1 , M. Horie 2 , H..W. Sampson 1 , Z. Hubert 1 , B. Tharakan 1 , R.E. Reeve1,<br />

W.P. Hamilton 1 , 1 Temple/US, 2 Tokyo/JP<br />

P176 Differentiation of mesenchymal stem cells by soluble factors mediated by direct co-culture of meniscus<br />

cells and mesenchymal stem cells<br />

M. Sommerfeldt, L. Laouar, A. Sierra, N. Jomha, A.B. Adesida, Edmonton/CA<br />

P177 A Novel Polycarbonate-Urethane Meniscal Implant: A Functional Evaluation of Sizing<br />

J.J. Elsner 1 , V. Condello 2 , E. Hershman 3 , R. Arbel 4 , A. Shterling 1 , C. Zorzi 5 , F. Guilak 6 , E. Linder-Ganz 1 ,<br />

1 Netanya/IL, 2 Negrar, Verona/IT, 3 New York/US, 4 Tel Aviv/IL, 5 Negrar <strong>–</strong> Verona/IT, 6 Durham/US<br />

P178 Poly-urethane scaffold for the treatment of medial and lateral partial meniscus defects: A single center<br />

experience with focus on scaffold integrity.<br />

P.C. Verdonk, L. Willemot, P. Beekman, R. Verdonk, Gent/BE<br />

P179 Transplantation of Achilles tendon treated with BMP-7 promoted meniscus regeneration in a rat massive<br />

meniscus defect model.<br />

N. Ozeki 1 , 2 , I. Sekiya 1 , T. Saito 2 , T. Muneta 1 , 1 Tokyo/JP, 2 Yokohama/JP<br />

P180 Co-culture of meniscus cells and bone marrow mesenchymal stromal cells on a collagen scaffold for<br />

inner meniscus reconstruction<br />

A. Adesida, S. Lim, N.-F. Matthies, A. Sierra-Mulet, N.M. Jomha, Edmonton/CA<br />

P181 All-arthroscopic technique of biological meniscal tear therapy with collagen membrane- early results<br />

T. Piontek 1 , K. Ciemniewska-Gorzela 1 , J. Naczk 1 , M. Slomczykowski 2 , R. Jakob 3 , 1 Poznan/PL, 2 Scholles/<br />

UK, 3 Tafers/CH<br />

P182 Oxygen tension is a determinant of cultured human meniscus cells matrix-forming phenotype<br />

A. Adesida, A. Sierra-Mulet, N.M. Jomha, Edmonton/CA<br />

P183 Comparison of the displacement pre- and post-surgery of 24 polyurethane meniscal implants; is there a<br />

difference in displacement and is there any correlation with the thickness of the residual meniscal rim?<br />

P.C. Verdonk 1 , T. De Coninck 1 , W.C.J. Huysse 1 , R. Verdonk 2 , 1 Gent/BE, 2 Ghent/BE<br />

P184 Tissue Engineered Whole Menisci Generated from High Density Type I Collagen Gels<br />

J. Puetzer, L. Bonassar, Ithaca/US<br />

P185 A pilot study of the use of a polycarbonate-urethane implant (NUsurface) for the treatment of<br />

postmeniscectomy medial knee pain.<br />

P.C. Verdonk 1 , A.A.M. Dhollander 2 , R. Verdonk 2 , 1 Gent-Zwijnaarde/BE, 2 Ghent/BE<br />

P186 Biological reconstruction of the failing knee: experience of meniscus allograft transplantation with<br />

osteotomy, ligament reconstruction and articular cartilage repair.<br />

A. Getgood, S. Spencer, J. Bird, P. Thompson, T. Spalding, Coventry/UK


Poster Sessions (Electronic & Traditional)<br />

P187 Degradation of articular cartilage after partial meniscectomy <strong>–</strong> is there a need for meniscal transplantation?<br />

S. Quirbach 1 , M. Eichinger 1 , S. Apprich 2 , R. Rosenberger 1 , 1 Innsbruck/AT, 2 Vienna/AT<br />

P188 Too old for regenerative treatment? Too young for joint replacement? Clinical indication for a novel<br />

meniscus implant<br />

C. Zorzi 1 , V. Condello 2 , P.C. Verdonk 3 , R. Arbel 4 , S. Israeli 5 , Y. Beer 6 , N. Blumberg 4 , N. Shabshin 4 ,<br />

J.J. Elsner 7 , E. Nocco 7 , E. Linder-Ganz 7 , G. Agar 6 , N. Rozen 5 , R. Verdonk 8 , E. Hershman9, 1 Verona/IT,<br />

2 Negrar, Verona/IT, 3 Gent-Zwijnaarde/BE, 4 Tel Aviv/IL, 5 Afula/IL, 6 Zeriffin/IL, 7 Netanya/IL, 8 Gent/<br />

BE, 9 New York/US<br />

Microfracture<br />

P189 Bone marrow stimulation produces superior cartilage repair in trochlea versus medial femoral condyle in<br />

a rabbit model<br />

H. Chen 1 , A. Chevrier 1 , C.D. Hoemann 1 , J. Sun 2 , G. Picard 1 , M.D. Buschmann 1 , 1 <strong>Montreal</strong>/CA, 2 Laval/CA<br />

P190 Autologous matrix-induced chondrogenesis (AMIC) combined with platelet-rich plasma gel compared to<br />

AMIC enhanced by concentrated bone marrow for the treatment of severe patellar cartilage defects in<br />

the knee.<br />

W. Widuchowski, P. Lukasik, W. Wawrzynek, R. Kokot, J. Widuchowski, Piekary Slaskie/PL<br />

P192 Surgical Treatment of Chondral Defects of Knee Using Microdrilling and Atelocollagen Gel as One Stage<br />

Arthroscopic Procedure<br />

A.A. Shetty 1 , S.-J. Kim 2 , D. Stelzeneder 1 , P. Bilagi 1 , 1 Kent/UK, 2 Uijeongbu City/KR<br />

P193 T2* Mapping After Arthroscopic Treatment of <strong>Cartilage</strong> Defects of the Knee using Microdrilling and an<br />

Atelocollagen Gel as a One Stage Procedure<br />

P. Bilagi 1 , D. Stelzeneder 1 , S.-J. Kim 2 , A.A. Shetty 1 , 1 Kent/UK, 2 Uijeongbu City/KR<br />

P194 Microfracture in the Treatment of Osteochondral Defects of the Talus: Clinical results at an average<br />

follow-up of 9 years with T2-Mapping at 3 Tesla<br />

C. Becher 1 , D. Zuehlke 1 , C. Plaas 1 , M. Ewig 1 , C. Stukenborg-Colsman 1 , H. Thermann 2 , 1 Hannover/DE,<br />

2 Heidelberg/DE<br />

P195 All arthroscopic AMIC cartilage reconstruction in the knee <strong>–</strong> 1 year results<br />

T. Piontek, K. Ciemniewska-Gorzela, A. Szulc, J. Naczk, Poznan/PL<br />

P197 The AMIC technique in the cartilage lesions of the knee: 97 cases<br />

A. Siclari 1 , E. Boux 1 , C. Gentili 2 , C. Kaps 3 , U. Freymann 3 , 1 Biella/IT, 2 Genova/IT, 3 Berlin/DE<br />

Osteoarthritis<br />

P198 Blocking oncostatin M in synovial fluid of osteoarthritic patients increases matrix turnover.<br />

M. Beekhuizen 1 , A.G.J. Bot 1 , D.B.F. Saris 1 , 2 , W.J.A. Dhert 1 , G.J.V.M. Van Osch 3 , L.B. Creemers 1 , 1 Utrecht/NL,<br />

2 Enschede/NL, 3 Rotterdam/NL<br />

P199 Comprehensive Arthroscopic Management (CAM) Survivorship: Alternative to Arthroplasty in Active<br />

Patients with Advanced Shoulder Osteoarthritis<br />

P.J. Millett, M. Horan, A. Pennock, D. Rios, Vail, Co/US<br />

P200 Does Kellgren-Lawrence Grade Correlate with Arthroscopic Findings in the Knee?<br />

K.K. Briggs, L. Matheny, J..R. Steadman, W.G. Rodkey, Vail/US<br />

95


96<br />

Poster Sessions (Electronic & Traditional)<br />

P201 Superficial chondrocytes from osteoarthritis patients have reduced matrix metalloproteinase expression<br />

and increased matrix synthesis following dynamic compression<br />

J.E. Jeon, K. Schrobback, D. Hutmacher, T.J. Klein, Kelvin Grove/AU<br />

P202 Quantitative CT and MRI changes in arthritic and prearthritic hips<br />

A.D. Speirs, A. Cárdenas-Blanco, K. Rakhra, M. Schweitzer, P. Beaule, Ottawa/CA<br />

P203 Hyaluronan Derivative Hydrogel treatment for Preserving <strong>Cartilage</strong> Integrity in Rabbit Model of<br />

Osteoarthritis (ACLT)<br />

A. Schiavinato 1 , M. Ganster 2 , P. Mainil-Varlet 3 , 1 Abano Terme (pd)/IT, 2 Bern/CH, 3 Marly/CH<br />

P204 >Topographical Distribution of Glycosaminoglycans in Developmental Dysplasia of the Hip as Measured<br />

by Delayed Gadolinium-Enhanced MRI of <strong>Cartilage</strong> (dGEMRIC)<br />

A. Hingsammer, J. Chan, L.A. Kalish, Y.-J. Kim, Boston/US<br />

P205 Pattern of cartilage erosion in end-stage osteoarthritis of the knee<br />

O.R. Kwon, S.K. Kwon, Seoul/KP<br />

P206 Intra-articular lubricin injection as a means of preventing early articular surface damage<br />

N. Galley 1 , M. Rivera-Bermudez 2 , T. Blanchet 2 , C.R. Flannery 2 , L. Bonassar 1 , 1Ithaca/US, 2 Cambridge/US<br />

P207 Prostaglandin E2 upregulation by cyclic compressive loading on 3-D tissue of human synovial fibroblasts<br />

via COX-2 and IL-1 receptor signal pathway<br />

K. Shimomura, K. Kita, T. Kanamoto, N. Nakamura, S. Miyamoto, T. Mae, T. Matsuo, H. Yoshikawa,<br />

K. Nakata, Osaka/JP<br />

P208 The effects of Hyaluronic Acid and Allogenic Bone Marrow derived-Mesenchymal stem cells in delaying<br />

osteoarthritic progression of the knee: A preliminary study<br />

T. Zaman, A. Suhaeb, S. Naveen, Kuala Lumpur/MY<br />

P209 Comparison of Factors Associated with Early Osteoarthritis vs. Moderate to Severe Osteoarthritis of the Knee<br />

K.K. Briggs, L. Matheny, J..R. Steadman, W.G. Rodkey, Vail/US<br />

P210 Pattern of cartilage erosion in end-stage osteoarthritis of the knee; its association with the condition of<br />

anterior cruciate ligament and medial meniscus posterior horn<br />

O.R. Kwon, S.K. Kwon, Seoul/KP<br />

P211 Sequentially Programmed Magnetic Fields (SPMF) therapy as an effective treatment for osteoarthritis:<br />

follow up of phase II study.<br />

V.G. Vasishta, Bangalore/IN<br />

P212 Inhibition of Inflammatory Response in Human, Camel, Dog, and Cat Chondrocyte Microcarrier Spinner<br />

Cultures by NSAIDs and the Combination of Avocado Soybean Unsaponifiables, Glucosamine and<br />

Chondroitin Sulfate<br />

M.W. Grzanna, L.F. Heinecke, A.Y. Au, S.L. Ownby, A.C. Mrozinski, C.G. Frondoza, Edgewood/US<br />

P213 The effect of hyaluronic acid on the outcome of a total hip arthroplasty<br />

S. Colen 1 , L. Maeckelbergh 2 , M. Mulier 2 , 1 Maastricht/NL, 2 Leuven/BE<br />

P214 Validation of Novel Indentation Device for Measuring Indentation Forces of <strong>Cartilage</strong> in Osteoarthritic Knees<br />

C. Bell, H. Khan, S. Arno, P. Walker, P. Desai, New York/US


Poster Sessions (Electronic & Traditional)<br />

P215 Chitosan-hyaluronate hybrid is superior to hyaluronate or saline in delaying osteoarthrtis in a rat model<br />

D. Robinson 1 , Z. Nevo 2 , S. Patchornik 1 , N. Ben Shalom 1 , 1 PetahTikwa/IL, 2 Tel Aviv/IL<br />

P216 Hemicap- and Unicap <strong>–</strong> miniprosthesis and Hemicap patellofemoral (PF) - and PF-xl ( Wave ) <strong>–</strong> miniprosthesis.<br />

J.O. Laursen, Sønderborg/DK<br />

P217 The Load Absorbing KineSpring System as an Early Surgical Option for Patients with Knee Osteoarthritis<br />

R. Williams 1 , N.J. London 2 , K.F. Almqvist 3 , R. Verdonk 3 , T. Wilton 4 , J.B. Richardson 5 , C.S. Waller 6 ,<br />

D.A. Hayes 7 , 1 Cardiff/UK, 2 Harrogate/UK, 3 Gent/BE, 4 Derby/UK, 5 Oswestry/UK, 6 Sydney/AU, 7 Brisbane/AU<br />

Osteochondral Grafts<br />

P218 Autologous Osteochondral Transplantation of the Talus Partially Restores Contact Mechanics of the Ankle Joint<br />

A. Fansa, C.D. Murawski, C.W. Imhauser, J.T. Nguyen, J.G. Kennedy, New York/US<br />

P219 Vitrification of intact human articular cartilage<br />

N.M. Jomha, J.A.W. Elliott, G.K. Law, B. Maghdoori, J.F. Forbes, A.B. Adesida, A. Abazari, L. Laouar, X.<br />

Zhou, L.E. McGann, Edmonton/CA<br />

P220 Functional and T2-Mapping MRI Results of Autologous Osteochondral Transplantation of the Talus in 72 Patients<br />

C.D. Murawski, J.G. Kennedy, New York/US<br />

P221 Osteochondral repair using a novel biphasic implant made of scaffold-free tissue engineered construct<br />

derived from synovial mesenchymal stem cells and hydroxyapatite-based artificial bone<br />

K. Shimomura 1 , Y. Moriguchi 1 , W. Ando 2 , R. Nansai 3 , T. Susa 3 , K. Imade 3 , S. Mochizuki 3 , H. Fujie 3 , K.<br />

Kita 1 , T. Mae 1 , K. Nakata 1 , K. Shino 1 , H. Yoshikawa 1 , N. Nakamura 1 , 1 Osaka/JP, 2 Hyogo/JP, 3 Tokyo/JP<br />

P222 Viral Inactivation of Human Osteochondral Grafts with Methylene Blue and Light<br />

D.M. Squillace, Z. Zhao, G. Ofek, T. Jiang, R. Garrett, J. Gao, Austin/US<br />

P223 Second-Look Arthroscopic and Clinical Evaluation of Osteochondral Autograft Transplantation in Patients<br />

with Early Osteoarthritis Aged 45 or More<br />

H.-S. Seo, S.C. Lee, Seoul/KR<br />

P224 Mosaicpalsty (osteochondral autografting) in osteochondral lesions of the knee: a prospective study<br />

T.Y. Emre, M. Uzun, B. Seyhan, H.T. Cift, Ankara/TR<br />

P225 Open mosaicplasty in osteochondral lesions of the talus : A prospective study<br />

T.Y. Emre, M. Uzun, B. Seyhan, H.T. Cift, Ankara/TR<br />

P226 Cryoprotective agent efflux from intact articular cartilage<br />

H. Yu, K. Al-Abbasi, X. Zhou, A. Abazari, J.A.W. Elliott, L.E. McGann, N.M. Jomha, Edmonton/CA<br />

P227 Polyvinyl alcohol-polyacrylic acid (PVA-PAA) hydrogels for osteochondral defect repair<br />

D.A. Bichara, H. Bodugoz-Senturk, C.R. Bragdon, D. Ling, O.K. Muratoglu, Boston/US<br />

Others<br />

P229 Prevalence of Chondral Defects of the Hip in Professional Hockey Players vs. Non-Contact Professional Athletes<br />

M. Philippon, M.M. Herzog, K.K. Briggs, Vail/US<br />

P230 Intra- and Inter-rater Reliability of Arthroscopic Measurements of <strong>Cartilage</strong> Defects<br />

D. Flanigan 1 , R.H. Brophy 2 , J. Carey 3 , J. Mitchell 1 , W. Graham 1 , D. Hamilton 4 , R. Siston 1 , H. Nagaraja 1 ,<br />

C. Lattermann 4 , 1 Columbus/US, 2 Chesterfield/US, 3 Philadelphia/US, 4 Lexington/US<br />

97


98<br />

Poster Sessions (Electronic & Traditional)<br />

P231 The gene-expression profiles of immature bovine articular cartlage are joint-specific and differentially<br />

influenced by BMP-2<br />

N. Shintani, F. Bourquin, E. Hunziker, Bern/CH<br />

P232 Partial Anterior Cruciate Ligament Tears: Anatomic Reconstruction Versus Non Anatomic Augmentation Surgery<br />

R. Buda, F. Vannini, A. Ruffilli, M. Cavallo, A. Parma, L. Ramponi, S. Giannini, Bologna/IT<br />

P233 A Moderate-Intensity Static Magnetic Field Enhances <strong>Repair</strong> of <strong>Cartilage</strong> Damage in Rabbits<br />

F. Mojtahed Jaberi 1 , S. Keshtgar 2 , A. Tavakoli 1 , E. Pishva 1 , B. Gramizadeh 3 , N. Tanideh 3 , M. Mojtahed Jaberi 4 ,<br />

3 Shiraz/IR, 4 <strong>Montreal</strong>/CA<br />

P234 Using diffuse reflectance spectroscopy for optical detection of cartilage degeneration arthroscopically<br />

M. Junker 1 , N. Trutiak 2 , J..Q. Brown 1 , J. Von Windheim 1 , N. Ramanujam 1 , M.B. Hurtig 2 , 1 Durham/US, 2 Guelph/CA<br />

P236 Single Versus Double Strand Anterior Cruciate Ligament Reconstruction With Hamstrings: A 12 years<br />

follow-up Study<br />

R. Buda, A. Ruffilli, F. Vannini, A. Parma, L. Ramponi, G. Pagliazzi, S. Giannini, Bologna/IT<br />

P237 Treatment of osteochondral injuries of the ankle: Our Experience<br />

F. Cortese 1 , V. Iacono 1 , A. Russo 2 , G. Piovan 3 , C. Zorzi 1 , 1 Verona/IT, 2 Napoli/IT, 3 Trieste/IT<br />

P238 All-arthroscopic labral reconstruction in the hip by use of collagen graft - operative technique and early results<br />

T. Piontek, K. Ciemniewska-Gorzela, A. Szulc, J. Naczk, Poznan/PL<br />

P239 Gene expression profiles of the regenerated cartilage tissue induced by implantation of a novel double-network<br />

hydrogel : comparisons with the immature articular cartilage<br />

R. Imabuchi 1 , Y. Ohmiya 1 , H.J. Kwon 1 , S. Onodera 1 , N. Kitamura 1 , T. Kurokawa 2 , J.P. Gong 2 , K. Yasuda 1 ,<br />

1 Sappro/JP, 2 Sapporo/JP<br />

Platelet Rich Plasma<br />

P241 The Anti-inflammatory and Matrix Restorative Mechanisms of Platelet Rich Plasma in Osteoarthritis.<br />

E. Sundman 1 , B.J. Cole 2 , V. Karas 2 , C. Della Valle 2 , L.A. Fortier 1 , 1 Ithaca/US, 2 Chicago/US<br />

P243 In vivo and in vitro effects of PRP in chondrogenic differentiation<br />

A. Siclari 1 , E. Boux 1 , C. Gentili 2 , C. Kaps 3 , J.P. Krueger 3 , 1 Biella/IT, 2 Genova/IT, 3 Berlin/DE<br />

P244 Effect of platelet gel on healing response after anterior cruciate ligament reconstruction<br />

J. Naranda, M. Vogrin, Maribor/SI<br />

P246 Platelet rich plasma enhanced collagen II production in Transwell cultured mesenchymal stem cells<br />

A. Kabiri, M. Mardani, E. Esfandiary, B. Hashemibeni, A. Esmaeili, A. Pourazar, 8174673548/IR<br />

Rehabilitation<br />

P247 Five year outcomes of a randomized comparison of traditional and accelerated approaches to post-operative<br />

rehabilitation following matrix-induced autologous chondrocyte implantation (MACI)<br />

J. Ebert, K. Hambly, M. Fallon, T.R. Ackland, M.H. Zheng, D.J. Wood, Perth/AU<br />

P248 A web-based clinical decision support system for individualising rehabilitation after articular cartilage<br />

repair of the knee.<br />

K. Hambly, Chatham/UK<br />

P249 Physiotherapeutic treatment after AMIC knee all arthroscopic reconstruction of cartilage procedure <strong>–</strong><br />

cases study<br />

A. Prusinska, T. Piontek, K. Ciemniewska-Gorzela, J. Naczk, M. Grygorowicz, W. Dudzinski, Poznan/PL<br />

P250 Flexibility disorder of lower extremity muscles affects the location of pain in the patellofemoral pain<br />

syndrome (PFPS) patients.<br />

N. Reke, A. Madaj, T. Piontek, K. Ciemniewska-Gorzela, A. Pyda, M. Grygorowicz, W. Dudzinski, Poznan/PL<br />

P253 Relationship Between Leg Strength and Patient Report Outcomes in Autologous Chondrocyte<br />

Implantation (ACI) Patients C.G. Mattacola, J.S. Howard, C. Lattermann, Lexington/US


Poster Sessions (Electronic & Traditional)<br />

Stem Cells<br />

P254 A checkerboard analysis of the effect of commonly used growth factors on in vitro chondrogenesis using<br />

a custom-made probeset and the Nanostring nCounter platform<br />

R.B. Jakobsen 1 , T.S. Mikkelsen 2 , A.M. Küchler 1 , J.E. Brinchmann 1 , 1 Oslo/NO, 2 Cambridge Massachusetts/US<br />

P256 Pharmacological modulation of human mesenchymal stem cells chondrogenesis by a chemically<br />

over-sulphated polysaccharide of marine origin: potential application to cartilage regenerative medicine.<br />

C. Merceron, S. Portron, C. Vignes-Colombeix, E. Rederstorff, M. Masson, J. Lesoeur, S. Sourice,<br />

C. Sinquin, S. Colliec-Jouault, P. Weiss, C. Vinatier, J. Guicheux, Nantes/FR<br />

P257 Low oxygen tension prevents the terminal hypertrophic differentiation of chondrogenic cells<br />

S. Portron, C. Merceron, O. Gauthier, M. Masson, J. Lesoeur, S. Sourice, P. Weiss, J. Guicheux, C. Vinatier,<br />

Nantes/FR<br />

P258 Demonstration of the Chondrogenic Capacity of Mesenchymal Stem Cells Derived from Human Foetal<br />

Bone Marrow<br />

K. Brady 1 , S.C. Dickinson 1 , W. Kafienah 1 , P.V. Guillot 2 , J. Polak 2 , A.P. Hollander 1 , 1 Bristol/UK, 2 London/UK<br />

P261 Intra-individual comparison of the chondrogenic differentiaton capacity of ASCs and MSCs in type I<br />

collagen-hydrogel<br />

A. Nedopil 1 , R. Hallinger 1 , T. Schuster 2 , M. Rudert 1 , U. Nöth 1 , L. Rackwitz 1 , 1 Wuerzburg/DE, 2 Munich/DE<br />

P262 Isolation and characterisation of mesenchymal stem cells from different regions of the human umbilical cord<br />

C. Mennan, A. Bhattacharjee, K. Wright, S. Roberts, J.B. Richardson, Oswestry/UK<br />

P264 Multilineage differentiation potential of the cells derived from degenerative torn human rotator cuff<br />

I. Nagura, T. Kokubu, Y. Mifune, R. Sakata, H. Nishimoto, T. Muto, M. Kurosaka, Kobe/JP<br />

P265 Chondrogenesis by Bone Marrow-Derived Stem Cells Grown in Chondrocyte Conditioned Media for<br />

Possible Auricular Reconstruction<br />

X. Zhao 1 , N.S. Hwang 2 , D.A. Bichara 1 , I. Pomerantseva 1 , C.A. Sundback 1 , J. Vacanti 1 , D.G. Anderson 2 ,<br />

M.A. Randolph 1 , 1 Boston/US, 2 Cambridge/US<br />

P267 Adult adipose mesenchymal stem cell implantation for one step knee chondral defects repair<br />

F.V. Sciarretta, C. Ascani, Rome/IT<br />

P268 Infrapatellar Fat Pad-Derived Mesenchymal Stem Cell Therapy for Knee Osteoarthritis<br />

Y.J. Choi, Y.G. Koh, Y.-C. Kim, Y.-S. Park, 137-820/KR<br />

P270 Effect of intraarticular injection of stem cells on joint pain and structural changes associated with<br />

monoiodoacetate injection in rats<br />

L. Song 1 , A. Bendele 2 , S. Coyle 1 , R. Zhang 1 , T. Davisson 1 , 1 Mahwah/US, 2 Boulder, Co/US<br />

P271 Combined effect of mechanical load and BMP-2 overexpression on the chondrogenesis of human bone<br />

marrow derived stem cells<br />

A. Neumann 1 , C. Archer 2 , M. Alini 1 , M. Stoddart 1 , 1 Davos Platz/CH, 2 Cardiff/UK<br />

P272 Tet-regulated, lentivirally mediated BMP2 expression in rabbit mesenchymal stem cells for treatment of<br />

osteochondral defects<br />

S. Vogt 1 , D. Wuebbenhorst 1 , G. Wexel 1 , B. Gansbacher 1 , A.B. Imhoff 2 , M. Anton 1 , 1 Muenchen/DE, 2 Munich/DE<br />

P273 Manipulating the rate of biosynthesis and patterns of distributions of GAG macromolecules in murine<br />

chondrocyte cultures, by employing para-nitrophenyl ß-D-xyloside<br />

Z. Nevo, T. Weinstein, Z. Evron, M. Aviv, D. Robinson, Tel Aviv/IL<br />

99


100<br />

Notes


Extended Abstracts & Index<br />

1. Abstract Book<br />

The abstracts of all Free Papers and posters can be found in a searchable<br />

database as well as in a pdf-format for your print-out or download<br />

on our website www.cartilage.org<br />

(ICRS 2012 <strong>–</strong> Final Programme & Abstracts)<br />

2. Online Itinerary Planner<br />

Create and export your personal ICRS 2012 congress programme and<br />

abstracts to your own calendar on your computer or mobile device.<br />

The ICRS online itinerary planner is an interactive tool for<br />

ICRS conference attendees to plan their individual meetings and<br />

activities during the ICRS 2012 conference.<br />

Various search options and filters empower the user to find the<br />

presentations and sessions of his interest. Filters can be easily applied to<br />

narrow the search results to only specific days, rooms, key words, topics,<br />

authors. After browsing the program, all sessions or only individual<br />

presentations of interest can be added to a personal itinerary. The individualized<br />

PDF document and the information downloaded to the calendar can<br />

include abstracts or any available media.


102<br />

Extended Abstracts<br />

1.1<br />

Articular <strong>Cartilage</strong> Mechanisms, Management and Outcome<br />

B. Mandelbaum<br />

Santa Monica/United States of America<br />

Introduction: The game of Football as it is called worldwide is the<br />

played by more than 300 million people globally. It is the most<br />

popular sport in the world and participation in this dynamic sport<br />

continues to grow. We will from this point forward refer to the<br />

Football athlete as a Football (Soccer) Player for the importance<br />

of convention. The inherent nature of the game is played at the<br />

extremes of intense performance and at times is a high-impact<br />

contact sport associated with significant acute and chronic joint<br />

contact forces with potential detrimental effects to the joint surface.<br />

Articular cartilage injury is observed with increasing frequency in<br />

football players and is commensurate with the competitive level.<br />

Due to the limited spontaneous regeneration of articular cartilage<br />

injuries often lead to significant symptoms under the continued<br />

high demands of football ultimately resulting in the decrease in<br />

performance or the inability to play. The FIFA Medical Assessment<br />

and Research Center (F-MARC) and <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong><br />

<strong>Society</strong> (ICRS) recognize the enormous impact of articular cartilage<br />

injury for the football (Soccer) player. This session amplifiies the<br />

relationship between FIFA and ICRS in an effort to help advance<br />

the science and the understanding of articular cartilage injury and<br />

degeneration in the football (soccer) player as well as the options<br />

for its treatment and prevention The approach to the athlete always<br />

uses the “Spectrumåof Care” Paradigm . Prevention, Performance<br />

optimization, Injury care, rehabilitation and restoration and return<br />

to sport, prevention of re-injury and osteoarthritis and keeping<br />

the aging athlete healthy through exercise, These are the hallmark<br />

principles that illuminate the importance of these collaborative<br />

relationships.<br />

Content: During this session we will demonstrate how evidence based<br />

medicine (EBM) and innovation drives clinical decision making and<br />

practice and vice versa. The Scientific Evidence Base for <strong>Cartilage</strong><br />

Injury and <strong>Repair</strong> in the Athlete in Football (Soccer) players frequently<br />

injure the Anterior Cruciate Ligament (ACL) and menisci. These injuries<br />

and the inherent risk of the game create an overall prevalence of knee<br />

articular cartilage lesions of 36% to 38%. The resultant for athletes<br />

with articular cartilage lesions is often challenging because of the<br />

high demands placed on the normal, repaired and regenerated tissue<br />

by the game over time. These cartilage defects in athletes can be<br />

treated with microfracture, osteochondral auto or allogeniec tissues<br />

and autologous chondrocyte implantation. There is increasing and<br />

robust scientific evidence for cartilage repair in athletes, with more<br />

extensive information available for microfracture and autologous<br />

chondrocyte implantation than for osteochondral grafting. The<br />

ultimate challenge is the quality of the repair tissue; <strong>Repair</strong>ed and or<br />

regenerated cartilage must closely must resemble and function like<br />

hyaline cartilage may be the most significant factor for the return to<br />

sport. So what do we do for the specific athlete with a spectrum of<br />

cartilage deficits? How do we select the best specific option and at<br />

what timing? We will present the best available evidence for cartilage<br />

surgery and treatment selection, evaluate specific patient profiles<br />

for professional and recreational athletes, and propose a treatment<br />

algorithm for the treatment of focal cartilage lesions in football<br />

(soccer) players. Once the intervention is completed attention to<br />

detail of post operative protocols, rehabilitation, restoration of<br />

function, return to sport participation and competition and prevention<br />

of future injury and or progression towards osteoarthritis is essential.<br />

This necessitates a multidisciplinary approach to rehabilitation,<br />

especially in the transition from therapy to performance, training<br />

and playing in games. Return to football is always a “significant<br />

challenge” as presented by Drs. Mithoefer and Della Villa. . It should<br />

be recognized that not all players will return to football after articular<br />

cartilage repair. Factors that must be considered are return rate, time<br />

to return and durability after return. The .average return rate to sport<br />

after Articluar <strong>Cartilage</strong> repair is 79% without a significant difference<br />

in return rate or postoperative level of play between cartilage<br />

repair techniques. The range of time to return varied between 7<br />

to 17 months, with the longest time is for autologous chondrocyte<br />

transplantation (ACI).These parameters and expectations by the<br />

patient, athlete and coach must considered in all clinical decisions.<br />

Advanced sport-specific and a multidisciplinary phasic rehabilitation<br />

were able to reduce recovery time. The durability of results was best<br />

after ACI, with up to 96% continued sport participation after more<br />

than 3 years. There are several factors that determine favorable<br />

and successful return to football (soccer). These include player age<br />

(younger), time between injury symptoms and treatment (less time),<br />

competitive level of participation, (higher) defect size (smaller),<br />

and repair tissue morphology (hyaline or hyaline like). Sports and<br />

exercise participation after cartilage repair can and will facilitate<br />

joint restoration, functional recovery and fitness levels. Although the<br />

spectrum of care approaches listed above are hopeful, the probability<br />

of developing Osteoarthritis in soccer players is 5 <strong>–</strong> 12 times more<br />

frequent than in the general population. It is also highest for the most<br />

elite athletes. It is also responsible for retirement in about a third of<br />

professional players and diagnosed 4-5 years earlier. This process is<br />

a spectrum of degeneration and loss of volume or chondropenia that<br />

ultimately leads to frank osteoarthritis The etiology is multifactorial<br />

and involves the demands of the game, biochemical, biomechanical,<br />

inflammatory, nutritional and aging factors. As a result, athletes,<br />

particularly those with a history of knee injury, have an earlier onset<br />

and higher prevalence of osteoarthritis that would be expected based<br />

on their age. In essence these are “old knees in young people”. It<br />

remains a major cause of disability from the sport of football (soccer).<br />

This is our call for concern! Therefore the ultimate goal for FIFA and<br />

the ICRS is a prevention paradigm. Prevention of the injury, whether<br />

acute or overuse, prevention of chondropenia and osteoarthritis and<br />

the progression over time. .Injury prevention has been a major focus<br />

of FIFA and F-MARC through development of the PEP Program (Prevent<br />

Injury and Enhance Performance program) and then the evolution to<br />

the FIFA 11+ program. The PEP program was designed to reduce ACL<br />

injuries and several studies including a level I randomized control<br />

trial have documented significant reductions when this program<br />

is successfully utilized as a warm up. The evolution to the FIFA 11+<br />

program now includes focus on not only knee, but also muscle,<br />

groin and ankle injury. Significant reductions in severe, overuse and<br />

knee injuries have been demonstrated in controlled trials Therefore<br />

injury reduction is not only possible, it can and should be a reality<br />

and part of any football program. These programs are time efficient,<br />

easy to do, and are free of expense! The natural course progression<br />

is that articular cartilage defects are to become osteoarthritis over<br />

time. The next step in the FIFA/ ICRS initiative is how to prevent this<br />

progression? Although recent treatments for damage to articular<br />

cartilage have been successful in alleviating symptoms, more durable<br />

and complete, long-term articular surface restoration remains the<br />

unattained. This hopeful and futuristic approach looks an at both<br />

new ways to prevent damage to articular surfaces as well as new<br />

techniques to recreate biomechanically sound and biochemically<br />

true articular surfaces once an athlete injures this surface. This<br />

“holy grail “objective should be to produce hyaline cartilage with<br />

a well-integrated and flexible subchondral base and the normal<br />

zonal variability. Newer surgical techniques, some already in clinical<br />

study, and others on the horizon offer opportunities to improve the<br />

surgical restoration of the hyaline matrix often disrupted in athletic<br />

injury. These include new scaffolds, single-stage cell techniques,<br />

engineered allogeneic tissues, the use of mesenchymal stem cells,<br />

and gene therapies. The last step of the prevention paradigm is to<br />

prevent the progression of Osteoarthitis severity over time. There are<br />

a number of non-operative interventions have shown early promise in<br />

mitigating cartilage symptoms and in preclinical studies have shown<br />

evidence for the potential disease modification, chondrofacilitation<br />

and chondroprotection. These include the use of glucosamine,<br />

chondroitin, and other neutraceuticals, viscosupplementation with<br />

hyaluronic acid, platelet-rich plasma, and pulsed electromagnetic<br />

fields In conclusion, the approach to the athlete always uses the<br />

“Spectrum of Care” systematic paradigm. Prevention, Performance,<br />

Injury care, rehabilitation and restoration and return to sport,<br />

prevention of re-injury and osteoarthritis and keeping the aging<br />

athlete healthy through exercise. The main objective of the FIFA and<br />

the ICRS collaboration is the identify the scope of these problems;<br />

develop multidisciplinary solutions with the major goal of prevention<br />

for now and the future. It was Einstein that said, “It is the intelligent<br />

that can solve problems but the genius will prevent them.” It is with<br />

this spirit that we thank FIFA and its President Mr.Sepp Blatter, the<br />

ICRS and its president Professor Daniel Saris and all the authors<br />

for their timely and impactful contributions all of which making the<br />

game of football… a better game! Play on!<br />

References:<br />

1. Peterson L, Junge A, Chomiak J, Graf-Baumann T, Dvorak J. Incidence<br />

of football injuries and complaints in different age groups and skilllevel<br />

groups. Am J Sports Med. 2000;28(5 Suppl):S51-7<br />

2. Levy AS, Lohnes J, Sculley S, et al: Chondral delamination of the knee<br />

in soccer players. Am J Sports Med 1996; 24:634-39.<br />

3. Dvorak J.Br J Sports Med. 2011 Jun;45:673-6.<br />

4. Steinwachs, Engebretsen, Brophy, <strong>Cartilage</strong> 2012, FIFA/ICRS<br />

Supplement


5. McAdams T, Mithoefer K, Scopp J, Mandelbaum B. Articular cartilage<br />

injury in athletes. <strong>Cartilage</strong>, 2010;1, 3:165-179.<br />

6. Roos H, Dahlberg L, Hoerrner LA, Lark MW, Thonar EJ, Shinmei M,<br />

Lindqvist U, Lohmander LS. Osteoarthritis <strong>Cartilage</strong>. 1995 Mar;3(1):7-<br />

14.<br />

7. Mithoefer and Steadman, <strong>Cartilage</strong> 2012, FIFA/ICRS Supplement<br />

8. Panics et al, <strong>Cartilage</strong> 2012, FIFA/ICRS Supplement<br />

9. Mithoefer, Peterson, Minas, Mandelbaum, <strong>Cartilage</strong> 2012, FIFA/ICRS<br />

Supplement<br />

10. Mithöfer K, Peterson L, Mandelbaum B, Minas T. Articular cartilage<br />

repair in soccer players with autologous chondrocyte transplantation:<br />

functional outcome and return to competition. Am J Sports Med.<br />

2005;33:1639-46.<br />

11. Gortz et al, <strong>Cartilage</strong> 2012, FIFA/ICRS Supplement<br />

12. Robertson CM, Williams R. Return to sport after fresh osteochondral<br />

allograft transplantation. Annula Meeting of the American Orthopedic<br />

<strong>Society</strong> for Sports Medicine (AOSSM), Providence, Rhode Island, July<br />

17, 2010.<br />

13. Bekkers, de Windt, Brittberg. <strong>Cartilage</strong> repair in football athletes:<br />

What evidence leads to which treatment. <strong>Cartilage</strong> 2012 , FIFA/ICRS<br />

Supplement<br />

14. Hambly, Silvers, Steinwachs. Rehabilitation and Injury Prevention<br />

of <strong>Cartilage</strong> Injury in the Football Athlete, <strong>Cartilage</strong> 2012, FIFA/ICRS<br />

Supplement<br />

15. Mithoefer, Della Villa , <strong>Cartilage</strong> 2012, FIFA/ICRS Supplement<br />

16. Della Villa S, Kon E, Filardo G, Ricci M, Vincentelli F, Delcogliano<br />

M, Marcacci M. Does intensive rehabilitation permit early return to<br />

sport without compromising the clinical outcome after arthroscopic<br />

autologous chondrocyte implantation in highly competitive athletes.<br />

Am J Sports Med. 2010 ;38(1):68-77.<br />

17. Kreuz PC, Steinwachs M, Erggelet C, Lahm A, Krause S, Ossendorf<br />

C, et al. Importance of sports in cartilage regeneration after autologous<br />

chondrocyte implantation. Am J Sports Med 2007; 35: 1261-68.<br />

18. Lee and Chu, <strong>Cartilage</strong> 2012, FIFA/ICRS Supplement<br />

19. Drawer S, Fuller CW: Propensity for osteoarthritis and lower limb<br />

joint pain in retired professional soccer players. Br J Sports Med 2001;<br />

35:402-408.<br />

20. Kujala UM, Kettunen J, Paananen H, Aalto T, Battie MC, Impivaara<br />

O, Videman T, Sarna S. Knee Osteoarthritis in former runners, soccer<br />

players, weight lifters, and shooters. Arth Rheum 1995; 38:539-546.<br />

21. Kirkendall and Garrett. <strong>Cartilage</strong> 2012, FIFA/ICRS Supplement<br />

22. Mandelbaum BR, Silvers HJ, Watanabe DS, Knarr JF, Thomas SD,<br />

Griffin LY, Kirkendall DT, Garrett W Jr. Effectiveness of a neuromuscular<br />

and proprioceptive training program in preventing anterior cruciate<br />

ligament injuries in female athletes: 2-year follow-up. Am J Sports Med.<br />

2005 Jul;33(7):1003-10.<br />

23. Soligard T, Nilstad A, Steffen K, Myklebust G, Holme I, Dvorak J,<br />

Bahr R, Andersen TE. Comprehensive warm-up programme to prevent<br />

injuries in young female footballers: cluster randomised controlled<br />

trial. Br J Sports Med. 2010 Sep;44(11):787-93<br />

24. Zaslav et al. <strong>Cartilage</strong> 2012, FIFA/ICRS Supplement<br />

Extended Abstracts 103<br />

1.2<br />

Basic science mechanisms & animal models of articular cartilage<br />

injury<br />

M.B. Hurtig<br />

Guelph/<strong>Canada</strong><br />

Introduction: Osteoarthritis and osteochondral injury are possibly<br />

the most pressing medical issues of our time as population dynamics,<br />

psychosocial factors, and increasing life expectancy drive aging adults<br />

to maintain their activity level in the hope of optimizing their BMI,<br />

muscle tone, neuromuscular coordination, mental acuity and mental<br />

health. The current generation of children faces a less optimistic<br />

forecast because inactivity, diabetes and obesity contribute to multisystem<br />

metabolic disorders that are only partially understood. These<br />

children will miss the window of opportunity for conditioning their<br />

developing tissues during an active childhood (Helminen, 2000),<br />

and their less optimized neuromuscular conditioning and larger BMI<br />

may make them more susceptible to osteoarticular disease. Our<br />

attempts to control the incidence of joint injury and OA by modifying<br />

contributing factors in young people have only met with partial<br />

success; Knee injury prevention by neuromuscular conditioning in<br />

young female athletes may be the notable exception. Unlike mice,<br />

we have not identified genotypes among our population who display<br />

down-regulated inflammation and osteochondral repair after injury,<br />

though virtually all surgeons seem to believe that tissue quality and<br />

response to injury (surgical or otherwise) is a major determinant of<br />

postoperative success. In deference to our heterogeneous society, it<br />

seems that our scientific community should focus on the complexities<br />

of contributing factors in joint injury and repair. The reductionist<br />

approach we have needed in past was necessary to provide a proof<br />

of principle in genetically similar laboratory and other animals.<br />

Now that OA can be cured in mice and rats, and chondral defects<br />

repaired in rabbits, sheep, goats, pigs and horses; however noble<br />

these targets may have been, what are our next challenges? Clearly,<br />

notwithstanding the financial and regulatory barriers facing us, our<br />

scientific mandate must be to understand more complex model<br />

systems that have better predictive value for our patients.<br />

Content: Acute versus chronic chondral injuries<br />

As outlined in the ICRS consensus report on animal models (Hurtig<br />

2011), delayed repair of chronic chondral defects is a logical target.<br />

No patient arrives at the operating room within minutes of a chondral<br />

injury so it is no surprise that surgically induced defects treated at<br />

time=0 have much predictive value in development of new therapies.<br />

<strong>Cartilage</strong> resurfacing might need to be combined with therapies that<br />

address the dysregulation of matrix metabolism because many of<br />

our patients have long-standing synovitis, cartilage thinning and<br />

other evidence of a catabolic synovial environment environment.<br />

The anti-catabolic effects of antibodies against chemokines such at<br />

TNFα needed to be investigated better, and other targets developed.<br />

The anabolic effect of growth factors has been exploited in tissue<br />

engineering and to some extent in the selection of chondrocytes<br />

expressing genes that would support anabolism. One technical issue<br />

is cost, delivery and avoiding adverse systemic effects. A significant<br />

regulatory barrier exists for combination therapies that might include<br />

some combination of drugs, biologics, scaffolds or cells so a strong<br />

case for efficacy and safety needs to be constructed.<br />

<strong>Cartilage</strong> repair in suboptimal biomechanical, metabolic and other<br />

conditions<br />

The rapidly growing literature around diagnosis and management<br />

of FAI (femoro-acetabular impingement) is possibly the best<br />

example of how controversial clinical practices can develop before<br />

evidence based medicine is available. Reshaping the femoral<br />

neck, trimming the acetabular rim, repairing and reattaching the<br />

acetabular labrum all have uncertain long term outcomes, and<br />

the evidence that these change the progression of OA is lacking.<br />

It seems unlikely that the geometrical and shape changes in this<br />

joint can be completely addressed with surgery since both femoral<br />

and acetabular components can have changes in their orientation<br />

that would require invasive osteotomies and internal fixation. The<br />

evolution of hip arthroscopy is gaining considerable momentum and<br />

this will create a need for biologic hip resurfacing that can withstand<br />

some incongruity. It is interesting to note that a canine model of OA<br />

induced by reduced acetabular coverage was described by Inerot<br />

et al (1991) but few investigators have risen to this challenge. The<br />

considerable incidence of canine hip dysplasia, a polygenetic<br />

trait in large breed dogs, could provide a proving ground for new<br />

technologies in the face of naturally occurring disease with variable<br />

expression. National and international case canine surgical registries<br />

would be invaluable.


104<br />

Extended Abstracts<br />

Intercurrent disease and complex model systems<br />

Diabetes, other metabolic syndromes as well as environmental toxins<br />

share a common pathway in creating dysvascular tissues that response<br />

poorly to injury and interventions. There is clear clinical evidence that<br />

bone regeneration and cell-mediated cartilage repair are both affected<br />

by smoking. Diabetes and other peripheral vasculopathies have not<br />

been modeled in conjunction with animal models of cartilage repair, so<br />

little is known about how to promote vascularity, for what duration and<br />

during which phase of the repair.<br />

Logistics of delivery to the synovial environment<br />

Delivery of ancillary treatments for cartilage repair and prevention of<br />

chondral injury after injury<br />

The contraction of the pharmaceutical industry’s interest in OA is partly<br />

due to the need for patients to be exposed to a drug or therapy for a<br />

very long period-perhaps 10 or more years. This triggers safety concerns<br />

and the probability of registering a drug without long, expensive and<br />

possibly inconclusive results seems remote. To date, intra-articular<br />

therapies have been limited to long acting corticosteroids and (shortacting)<br />

hyaluronates but the development of sustained release intraarticular<br />

formulation of existing parental therapies seems logical. This<br />

could include growth factors, apoptosis inhibitors, surfactants, antiinflammatory<br />

drugs and other small molecules. Anderson et al (2011)<br />

describes the range of known therapies that could no doubt be expanded<br />

upon. The evolution of microspheres, encapsulated proteins, and other<br />

vehicles in sustained release formulations is well described in the tissue<br />

engineering literature and has potential to solve some of the problems<br />

inherent in managing patients with large, chronic chondral defects.<br />

References:<br />

Selected References<br />

1. Functional adaptation of articular cartilage from birth to maturity under<br />

the influence of loading: a biomechanical analysis. Brommer H., Brama<br />

P. A., Laasanen M. S., Helminen H. J., van Weeren P. R. and Jurvelin J. S.<br />

Equine Vet J 37(2):148-54 (2005)<br />

2. Early exercise advances the maturation of glycosaminoglycans and<br />

collagen in the extracellular matrix of articular cartilage in the horse.<br />

van Weeren P. R., Firth E. C., Brommer B., Hyttinen M. M., Helminen A.<br />

E., Rogers C. W., Degroot J. and Brama P. A. Equine Vet J 40(2):128-35<br />

(2008).<br />

3. Regular joint loading in youth assists in the establishment and<br />

strengthening of the collagen network of articular cartilage and<br />

contributes to the prevention of osteoarthrosis later in life: a hypothesis.<br />

Helminen H. J., Hyttinen M. M., Lammi M. J., Arokoski J. P., Lapveteläinen<br />

T., Jurvelin J., Kiviranta I. and Tammi M. I. J Bone Miner Metab 18(5):245-<br />

57 (2000).<br />

4. Anterior cruciate ligament injuries: etiology and prevention.. Brophy<br />

RH, Silvers HJ, Mandelbaum BR. Sports Med Arthrosc. 2010 Mar;18(1):2-<br />

11. Review.<br />

5. The association between knee joint biomechanics and neuromuscular<br />

control and moderate knee osteoarthritis radiographic and pain severity.<br />

Astephen Wilson JL, Deluzio KJ, Dunbar MJ, Caldwell GE, Hubley-Kozey<br />

CL. Osteoarthritis <strong>Cartilage</strong>. 2011 Feb;19(2):186-93.<br />

6. Proteoglycan alterations during developing experimental osteoarthritis<br />

in a novel hip joint model. Inerot S., Heinegård D., Olsson S. E., Telhag<br />

H. and Audell L. Degenerative hip joint disease was induced in dogs by<br />

extra-articular surgery that created a condition that mimics hip dysplasia.<br />

J Orthop Res 9(5):658-73 (1991).<br />

7. Response shift in self-reported functional scores after knee<br />

microfracture for full thickness cartilage lesions. Balain B., Ennis O.,<br />

Kanes G., Singhal R., Roberts S. N., Rees D. and Kuiper J. H. Osteoarthritis<br />

and <strong>Cartilage</strong> 17(8):1009-13 (2009)<br />

8. Does smoking influence outcome after autologous chondrocyte<br />

implantation?: A case-controlled study. Jaiswal P. K., Macmull S., Bentley<br />

G., Carrington R. W., Skinner J. A. and Briggs T. W. J Bone Joint Surg Br<br />

91(12):1575-8 (2009)<br />

9. Regenerative treatment with platelet-rich plasma combined with<br />

a bovine-derived xenograft in smokers and non-smokers: 12-month<br />

clinical and radiographic results. Yilmaz S., Cakar G., Ipci S. D., Kuru B.<br />

and Yildirim, B. Journal of clinical periodontology 37(1):80-7 (2010)<br />

10. May smokers and overweight patients be treated with a medial openwedge<br />

HTO? Risk factors for non-union. Meidinger G., Imhoff A. B., Paul<br />

J., Kirchhoff C., Sauerschnig M. and Hinterwimmer S. Knee Surg Sports<br />

Traumatol Arthrosc 19(3):333-9 (2011).<br />

11. <strong>Cartilage</strong> regeneration by gene therapy. Gelse K., von der Mark K. and<br />

Schneider H. Curr Gene Ther 3(4):305-17 (2003).<br />

12. Anticytokine therapy for osteoarthritis. Goldring M. B. Expert Opin<br />

Biol Ther 1(5):817-29 (2001).<br />

13. Beneficial effects of intra-articular caspase inhibition therapy<br />

following osteochondral injury<br />

Dang A. C., Warren A. P. and Kim H. T. Osteoarthritis and <strong>Cartilage</strong><br />

14(6):526-32 (2006).<br />

14. Weekly intra-articular injections of bone morphogenetic protein-7<br />

inhibits osteoarthritis progression. Hayashi M., Muneta T., Ju Y. J.,<br />

Mochizuki T. and Sekiya I. Arthritis Res Ther 10(5):R118 (2008).<br />

15. Efficacy of intra-articular injection of celecoxib in a rabbit model of<br />

osteoarthritis. Jiang D., Zou J., Huang L., Shi Q., Zhu X., Wang G. and Yang<br />

H. Int J Mol Sci 11(10):4106-13 (2010).<br />

16 .Repeated Platelet Concentrate Injections Enhance Reparative<br />

Response of Microfractures in the Treatment of Chondral Defects of the<br />

Knee: An Experimental Study in an Animal Model. Milano G., Deriu L.,<br />

Sanna Passino E., Masala G., Manunta A., Postacchini R., Saccomanno<br />

M. F. and Fabbriciani C. Arthroscopy (2012).<br />

Acknowledgments:<br />

The Canadian Arthritis Network and Canadian Institutes of Health<br />

Research contributed research funding.<br />

1.3<br />

<strong>Cartilage</strong> problems in the athlete: The scope of the problem<br />

K. Mithoefer<br />

Cambridge/United States of America<br />

Introduction: Injuries of the articular cartilage surfaces of the knee<br />

are observed with increasing frequency in athletes. Particularly<br />

participation in pivoting sports such as football, basketball, and<br />

soccer has been associated with a rising number of sports-related<br />

articular cartilage injuries with higher injury rates at the competitive<br />

and professional level. Injuries of the articular cartilage surface of the<br />

knee in the athlete can often occur in association with other acute<br />

injuries such as ligament or meniscal injuries, traumatic patellar<br />

dislocations, and osteochondral injuries and have been described<br />

in up to 50% of athletes undergoing anterior cruciate ligament<br />

reconstruction. Besides acute traumatic injury, articular cartilage<br />

injury can develop in the high-impact athletic population from<br />

chronic pathologic joint loading patterns such as joint instability<br />

or axis deviation. While intact articular cartilage adjusts to the<br />

increasing weightbearing activity in athletes by increasing cartilage<br />

volume and thickness in a linear dose-response relationship, recent<br />

studies indicate that this dose-response curve reaches a threshold<br />

and that activity beyond this threshold can result in maladaptation<br />

and injury of articular cartilage. High-impact joint loading above this<br />

threshold has been shown to decrease cartilage protoglycan content<br />

and to increase levels of degradative enzymes and chondrocyte<br />

apoptosis. Over time the integrity of the functional weight bearing<br />

unit is lost and a chondropenic response is initiated that can include<br />

loss of articular cartilage volume and stiffness, elevation of contact<br />

pressures, and development or progression of articular cartilage<br />

defects.<br />

Content: The limited spontaneous repair following acute or chronic<br />

articular cartilage injury is well documented. Recent reports<br />

demonstrated that hyaline cartilage defects in athletes resulted in<br />

significant pain and swelling and were associated with marked lifestyle<br />

changes and limitation of athletic activity. Some long-term data<br />

in athletes with isolated severe chondral or osteochondral damage<br />

in the weightbearing condyles showed a 75% initial return to sport<br />

initially, but a significant decline of athletic activity was observed over<br />

time with development of radiographic evidence of osteoarthritis in<br />

the 45-60% of athletes 14-34 years after the injury. These results are<br />

supported by the up to 12 fold increased risk of knee osteoarthritis in


high-impact athletes established by the National Institute of Health<br />

(NIH) and other independent studies. Untreated articular cartilage<br />

defects have been shown to result in significantly worse long-term<br />

joint function. The high demands on the joint surfaces in athletes<br />

make treatment of articular cartilage injuries and restoration of<br />

the injured joint surfaces critically important to facilitate continued<br />

athletic participation and to maintain a physically active lifestyle.<br />

The documented detrimental effect of high-impact articular loading<br />

in the athletic population requires cartilage surface restoration that<br />

can effectively withstand the significant mechanical joint stresses<br />

generated during high-impact, pivoting sports. Besides reducing<br />

pain, increasing mobility and improving knee function, the ability to<br />

return the athlete to sport and to continue to perform at the pre-injury<br />

athletic level presents one of the most important parameters for a<br />

successful outcome from articular cartilage repair in this challenging<br />

population. Treatment of articular cartilage injuries in the athletic<br />

population has traditionally presented a significant therapeutic<br />

challenge. However, development of new surgical techniques has<br />

created considerable clinical and scientific enthusiasm for articular<br />

cartilage repair. Based on the source of the cartilage repair tissue,<br />

these new surgical techniques can generally be categorized into<br />

three groups: marrow stimulation based techniques, osteochondral<br />

transplantation techniques, and cell-based repair techniques.<br />

Several studies have evaluated the microfracture technique<br />

specifically in the athletic population. These studies included<br />

recreational and professional athletes with follow-up ranging<br />

from 2-6 years. Activity scores and knee function scores increased<br />

significantly after microfracture in these athletes. Athletes were<br />

able to successfully return to high-impact, pivoting sports including<br />

football, soccer, alpine skiing, basketball, rugby, and tennis. Return<br />

to sports was reported at an average of 6.5-10 months. However,<br />

there was marked variability in the ability to return to sport after<br />

microfracture. The ability to return to athletics at the preoperative<br />

level also varied significantly. Return to professional and competitive<br />

level sports was much better than to recreational athletics. A<br />

recent study showed that while return to sports participation after<br />

microfracture can be achieved rapidly, performance and playing time<br />

will increase gradually to full participation. Return to high-impact<br />

sports after microfracture has been found to be higher in younger<br />

athletes with small lesion size, shorter preoperative symptoms, and<br />

without prior surgical intervention. A decline of initial improvement<br />

of postoperative sports participation was observed in some studies<br />

after microfracture in athletes and occurred between 24-37 months,<br />

however, activity levels and functional scores were still better than<br />

at baseline. Osteochondral mosaicplasty has also been specifically<br />

evaluated in athletes. Up to 95% good or excellent results with<br />

improved functional scores and MRI rating have been reported.<br />

Return to full athletic activity was reported in 61-93% of athletes at an<br />

average of 6.5 months. Longer preoperative symptoms and increased<br />

athlete age resulted in delayed return to sport after mosaicplasty.<br />

Preoperative radiographic or clinical evidence of joint degeneration<br />

predicted a return to sport at a lower level or even retirement from<br />

competitive sports following mosaicplasty. Prospective randomized<br />

comparison of mosaicplasty and microfracture in athletes reported<br />

significantly better results with mosaicplasty at an average of 36<br />

months. While studies have evaluated autologous osteochondral<br />

transfer in athletes, no specific information has been reported on this<br />

technique using allograft in this population. Autologous chondrocyte<br />

transplantation has recently been evaluated in the demanding<br />

athletic population. Good to excellent results were demonstrated in<br />

72-96% with significant improvement of activity score. Best results<br />

were obtained with single cartilage lesions of the medial femoral<br />

condyle. Return to high impact-athletics was higher in younger,<br />

competitive athletes, with short preoperative intervals while return in<br />

recreational athletes was less predictable. The time to return to sport<br />

was shorter in competitive level athletes. Athletes often returned<br />

to the same skill level and a high portion of returning athletes<br />

maintained their ability to perform 52 months after chondrocyte<br />

implantation. Return to athletics was better with fewer prior surgeries<br />

but return to sports was successful also with autologous cartilage<br />

transplantation as a salvage procedure. Combined pathology<br />

such as malalignment, ligamentous instability, or meniscal injury<br />

and deficiency is frequently encountered by the surgeon treating<br />

articular cartilage defects in the athletic knee. Surgically addressing<br />

these concomitant pathologies is critical for an effective and durable<br />

articular cartilage repair. Recent data demonstrated that isolated or<br />

combined adjuvant procedures have no significant negative effect<br />

on the ability to return to athletics after microfracture, mosaicplasty,<br />

or autologous chondrocyte transplantation. In conclusion, articular<br />

cartilage repair in athletes is aimed at returning the athlete to the<br />

pre-injury level of athletic participation without increased risk for<br />

long-term arthritic degeneration. Several surgical techniques have<br />

been shown to improve function and athletic activity after articular<br />

cartilage repair in this population but the rate of improvement and<br />

Extended Abstracts 105<br />

ability to return to athletic activity is dependent on several factors.<br />

The choice of repair technique should be tailored to individual<br />

patient and lesion characteristics using an established treatment<br />

algorithm. Long-term studies in this population will determine the<br />

efficacy of articular cartilage repair to reverse chondropenia and to<br />

prevent development of secondary arthritic degeneration.<br />

References:<br />

1. Della Villa S, Kon E, Filardo G, Ricci M, Vincentelli F, Delcogliano<br />

M, Marcacci M. Does intensive rehabilitation permit early return to<br />

sport without compromising the clinical outcome after arthroscopic<br />

autologous chondrocyte transplantation in highly competitive<br />

athletes? Am J Sports Med. 2010 Jan;38(1):68-77<br />

1. Drawer S, Fuller CW: Propensity for osteoarthritis and lower limb<br />

joint pain in retired professional soccer players. Br J Sports Med<br />

2001; 35:402-408.<br />

2. Flanigan DC, Harris JD, Trin TQ, Siston R, Brophy RH. Prevalence<br />

of chondral defects in athlete’s knees. A systematic review. Med Sci<br />

Sports Exerc 2010.<br />

3. Gudas R, Kelesinskas RJ, Kimtys V, Stankevicius E, Toliusis<br />

V, Benotavicius G, Smailys A: A prospective randomized clinical<br />

study of mosaic osteochondral autologous transplantation versus<br />

microfracture for the treatment of osteochondral defects in the knee<br />

joint in young athletes. Arthroscopy 2005; 21:1066-75.<br />

4. Kujala UM, Kettunen J, Paananen H, Aalto T, Battie MC, Impivaara<br />

O, et al. Knee Osteoarthritis in former runners, soccer players, weight<br />

lifters, and shooters. Arth Rheum 1995; 38:539-546.<br />

5. Levy AS, Lohnes J, Sculley S, LeCroy M, Garrett W. Chondral<br />

delamination of the knee in soccer players. Am J Sports Med 1996;<br />

24:634-39.<br />

6. Maletius W, Messner K: The long-term prognosis for severe<br />

damage to the weightbearing cartilage in the knee: A 14-year clinical<br />

and radiographic follow-up in 28 young athletes. Acta Orthop Scand<br />

1996; 165-168.<br />

7. Mithoefer K, Gill TJ, Cole BJ, Williams RJ, Mandelbaum BR. Clinical<br />

outcome and return to competition after microfracture in the<br />

athlete’s knee: An evidenc-based syetematic review. <strong>Cartilage</strong> 2010;<br />

1:113-20.<br />

8. Mithoefer K, Hambly K, Della Villa S, Silvers H, Mandelbaum<br />

BR. Return to sports participation after articular cartilage repair in<br />

the knee: scientific evidence. Am J Sports Med 2009; 37 Suppl 1:<br />

167S-76S.<br />

9. Mithöfer K, Minas T, Peterson L, Yeon H, Micheli LJ: Functional<br />

outcome of articular cartilage repair in adolescent athletes. Am J<br />

Sports Med 2005; 33:1147-1153.<br />

10. Mithöfer K, Peterson L, Mandelbaum BR, Minas T: Articular<br />

cartilage repair in soccer players with autologous chondrocyte<br />

transplantation: Functional outcome and return to competition. Am J<br />

Sports Med 2005; 33(11):1639-46.<br />

11. Mithoefer K, Williams RJ, Warren RF, Wickiewicz TL, Marx<br />

RG. High-impact athletics after knee articular cartilage repair: A<br />

prospective evaluation of the microfracture technique. Am J Sports<br />

Med. 2006;34:1413-8.<br />

12. Piasecki DD, Spindler KP, Warren TA, Andrish JT, Parker RD:<br />

Intraarticular injuries associated with anterior cruciate ligament tear:<br />

findings at ligament reconstruction in high school and recreational<br />

athletes. Am J Sports Med 2003; 31:601-5.<br />

13. Walzcac BE, McCulloch PC, Kang RW, Zelazny A, Tedeschi F,<br />

Cole BJ. Abnormal findings on knee magnetic resonance imaging in<br />

asymptomatic NBA players. J Knee Surg 2008; 21:27-33.


106<br />

Extended Abstracts<br />

2.1.1<br />

Growth factors pathways as regulators of chondrogenesis<br />

F. Beier<br />

London/<strong>Canada</strong><br />

Introduction: Growth factors and their downstream sign align<br />

pathways are essential regulators of chondrogenesis during<br />

development and thus prime candidates for applications in tissue<br />

engineering of cartilage. We will discuss general issues associated<br />

with these topics and present some of our recent findings.<br />

Content: The mechanisms regulating chondrogenesis during<br />

skeletal development are highly relevant both to our understanding<br />

of developmental diseases such as chondrodysplasias and to<br />

approaches towards tissue engineering of cartilage, for example to<br />

repair cartilage defects in osteoarthritis and other joint diseases.<br />

Growth factors are endogenous regulators of this process in vivo<br />

and a logical choice for use in tissue engineering, due to their<br />

relative ease of application and their extracellular mode of action<br />

(e.g. they can be added to the culture medium and don’t require<br />

cellular uptake). For example, bone morphogenetic proteins (BMPs)<br />

and other members of the transforming growth factor beta families<br />

generally act as promoters of chondrogenic differentiation (Bobick<br />

et al., 2009; Seo and Na, 2011). In contrast, Wnt proteins that signal<br />

through the canonical beta-catenin pathway promote osteogenic over<br />

chondrogenic differentiation of mesenchymal precursor cells. While<br />

these two families of growth factors received the bulk of attention in<br />

recent years, numerous other factors including hedgehog proteins,<br />

members of the fibroblast, insulin-like and epidermal growth factor<br />

(FGF, IGF and EGF) families and C-type natriuretic peptide have all<br />

been implicated in modulation of chondrogenesis in vivo and/or in<br />

vitro (Bobick et al., 2009; Seo and Na, 2011; Weiss et al., 2010; Woods<br />

et al., 2007a). Importantly, the specific effects of these growth factors<br />

are dependent on their concentration and their spatial and temporal<br />

patterns of action. Moreover, all these factors do not act in isolation,<br />

but cross-regulate each other and converge on key cellular signaling<br />

pathways and transcription factors. The cellular response therefore<br />

presents the integration of all the different, often contradictory<br />

signals the cells receive from the various growth factors. Further<br />

complexity is added by additional, non-growth factor signals that<br />

act on the cells, such as cell-matrix and cell-cell interactions and<br />

other signaling molecules including retinoic acid, nitric oxide and<br />

prostaglandins. Chondrogenesis is a multistep process that includes<br />

the steps of: 1) precursor cell condensation; 2) cellular commitment<br />

and subsequent differentiation to the chondrogenic fate; 3) secretion<br />

of a cartilage-specific extracellular matrix composed of collagen II and<br />

aggrecan (among other proteins and non-protein matrix components<br />

such as hyaluronic acid); 4) chondrocyte proliferation and, in the<br />

case of growth plate chondrocytes, 5) further differentiation towards<br />

hypertrophy followed by 6) apoptosis and replacement of cartilage<br />

by bone tissue(Bobick et al., 2009). All these steps are regulated<br />

by distinct (but overlapping) signals and mechanisms. In tissue<br />

engineering approaches, the signals that can be utilized depend<br />

on the desired application. For example, if one aims to generate<br />

articular cartilage for applications in degenerative joint disease, it is<br />

essential to promote early chondrocyte differentiation and possibly<br />

proliferation, but at the same time prevent differentiation further<br />

along the growth plate chondrocyte track towards hypertrophy.<br />

This makes BMPs unlikely candidates for such approaches as they<br />

ultimately cause hypertrophy and replacement of cartilage by<br />

bone. In contrast, for other applications (for example the repair of<br />

developmental defects, growth plate injuries or bone fractures),<br />

differentiation along the entire growth plate pathway might be<br />

desirable, which will then require a different approach. Thus, design<br />

and optimization of growth factor treatments for a particular tissue<br />

engineering strategy is complex and has to consider combination of<br />

different factors, timing and concentration of each individual factor.<br />

Thorough analyses of growth factor regulation of chondrogenesis<br />

during development can certainly be used to guide such designs.<br />

However, in addition to the complexities of growth factor effects<br />

on cells discussed above, other regulatory processes complicate<br />

analyses of these activities in vivo. These posttranslational<br />

mechanisms include the presence of extracellular antagonists and<br />

decoy receptors for many growth factors, sequestration of growth<br />

factors in the matrix, and proteolytic activation as well as inactivation<br />

of these factors. Thus, simple analyses of growth factor expression<br />

during development give, at best, an approximation of the potential<br />

function of these growth factors during chondrogenesis. Thorough<br />

and detailed functional analyses, such as through sophisticated<br />

conditional gene targeting in mice, is required to uncover how<br />

growth factors act together to control chondrogenesis. One<br />

potential drawback to the wide use of growth factors in cartilage<br />

tissue engineering is the high cost of recombinant growth factors.<br />

A potential strategy to overcome this limitation is the use of more<br />

economic small molecule modulators (inhibitors or activators) of<br />

the respective receptors or of downstream signaling molecules. The<br />

later also have the advantage that most major signaling molecules<br />

mediate cellular responses to many different growth factors. One<br />

group of signaling molecules that have been implicated in the<br />

control of chondrogenesis are the mitogen-activated protein (MAP)<br />

kinases (Bobick and Kulyk, 2008; Stanton et al., 2003). These<br />

proteins occupy a central role in the cell’s signaling network and are<br />

activated by many growth factors involved in chondrogenesis, such<br />

as TGFbeta family members, FGFs, EGFs etc. Of the major Map kinase<br />

subfamilies, ERK1/2 generally suppress, while p38 proteins promote<br />

chondrogenesis. MAP kinases exert their biological effects by<br />

controlling the activities of transcription factors and other proteins,<br />

but the specific downstream mediators responsible for the activities<br />

of ERK1/2 and p38 during chondrogenesis are not well understood.<br />

Another important group of signaling molecules that have been<br />

implicated in the control of chondrogenesis are small molecular<br />

switches of the Rho GTPase family (Woods et al., 2007a). Similar to<br />

MAP kinases, their activities are modulated by many different growth<br />

factors as well as by signals from cell-matrix and cell-cell interactions<br />

(through integrins, cadherins etc). Our work, and subsequently that<br />

of others, has shown that the classical member of the family, RhoA,<br />

suppresses chondrogenesis, in part through repression of Sox9, the<br />

chondrogenic ‘master’ transcription factor (Woods and Beier, 2006;<br />

Woods et al., 2005, 2007a). In contrast, two other Rho GTPases, Rac1<br />

and Cdc42, promote chondrogenesis (Woods et al., 2007b). A similar<br />

antagonism between RhoA and Rac1/Cdc42 applies to later stages<br />

of chondrocyte maturation, e.g. hypertrophy and apoptosis (Wang<br />

and Beier, 2005; Wang et al., 2004), at least in vitro. Many additional<br />

signaling pathways with important roles in chondrogenesis have been<br />

described. One pathway of particular interest is presented by glycogen<br />

synthase kinase 3 (GSK-3) which acts as a ‘brake’ on many of the<br />

discussed pathways (Kaidanovich-Beilin and Woodgett, 2011). The<br />

two GSK-3 proteins, GSK-3alpha and GSK-3beta, are best known for<br />

inducing the degradation of beta-catenin, the central component of the<br />

canonical Wnt pathway. However, these proteins are also involved in<br />

most of the other pathways controlling chondrogenesis. Manipulation<br />

of their activity thus has the potential to modulate the cell response<br />

to many of these factors and to drive the cells towards the desired<br />

chondrocyte phenotype. In closing, both the controlled application of<br />

growth factors and the manipulation of downstream pathways have<br />

the potential to further improve chondrogenesis for tissue engineering<br />

approaches. However, a better understanding of the endogenous role<br />

of these pathways in developmental chondrogenesis is likely required<br />

to optimize tissue engineering approaches so that chondrocytes of the<br />

desired phenotype and characteristics can be obtained.<br />

References:<br />

Bobick, B.E., Chen, F.H., Le, A.M., and Tuan, R.S. (2009). Regulation<br />

of the chondrogenic phenotype in culture. Birth Defects Res C Embryo<br />

Today 87, 351-371.<br />

Bobick, B.E., and Kulyk, W.M. (2008). Regulation of cartilage formation<br />

and maturation by mitogen-activated protein kinase signaling. Birth<br />

Defects Res C Embryo Today 84, 131-154.<br />

Kaidanovich-Beilin, O., and Woodgett, J.R. (2011). GSK-3: Functional<br />

Insights from Cell Biology and Animal Models. Front Mol Neurosci 4,<br />

40. Seo, S., and Na, K. (2011).<br />

Mesenchymal stem cell-based tissue engineering for chondrogenesis.<br />

J Biomed Biotechnol 2011, 806891. Stanton, L.A., Underhill, T.M., and<br />

Beier, F. (2003). MAP kinases in chondrocyte differentiation. Dev Biol<br />

263, 165-175.<br />

Wang, G., and Beier, F. (2005). Rac1/Cdc42 and RhoA GTPases<br />

antagonistically regulate chondrocyte proliferation, hypertrophy, and<br />

apoptosis. J Bone Miner Res 20, 1022-1031.<br />

Wang, G., Woods, A., Sabari, S., Pagnotta, L., Stanton, L.A., and Beier,<br />

F. (2004). RhoA/ROCK signaling suppresses hypertrophic chondrocyte<br />

differentiation. J Biol Chem 279, 13205-13214.<br />

Weiss, S., Hennig, T., Bock, R., Steck, E., and Richter, W. (2010).<br />

Impact of growth factors and PTHrP on early and late chondrogenic<br />

differentiation of human mesenchymal stem cells. J Cell Physiol 223,<br />

84-93.<br />

Woods, A., and Beier, F. (2006). RhoA/ROCK signaling regulates<br />

chondrogenesis in a context-dependent manner. J Biol Chem 281,<br />

13134-13140.


Woods, A., Wang, G., and Beier, F. (2005). RhoA/ROCK signaling regulates<br />

Sox9 expression and actin organization during chondrogenesis. J Biol<br />

Chem 280, 11626-11634.<br />

Woods, A., Wang, G., and Beier, F. (2007a). Regulation of chondrocyte<br />

differentiation by the actin cytoskeleton and adhesive interactions. J<br />

Cell Physiol 213, 1-8. Woods, A., Wang, G., Dupuis, H., Shao, Z., and<br />

Beier, F. (2007b). Rac1 signaling stimulates N-cadherin expression,<br />

mesenchymal condensation, and chondrogenesis. J Biol Chem 282,<br />

23500-23508.<br />

Acknowledgments:<br />

F.B. acknowledges funding from the Canadian Institutes of Health<br />

Research and the <strong>Canada</strong> Research Chair Program.<br />

2.1.2<br />

Signaling proteins and transcription factors in joint development<br />

and degeneration<br />

M. Pacifici, M. Iwamoto<br />

Philadelphia/United States of America<br />

Introduction: During limb skeletogenesis, chondrocytes located at the<br />

epiphyseal ends of long bone anlagen develop into permanent articular<br />

chondrocytes that sustain joint function though life (1). Instead,<br />

the chondrocytes constituting the shaft are transient cells, become<br />

organized in growth plates, undergo maturation and hypertrophy, and<br />

are replaced by bone via endochondral ossification (2). The essential<br />

nature of this developmental bifurcation in chondrocytes is widely<br />

appreciated, but its regulation remains poorly understood, particularly<br />

at the molecular level.<br />

We previously showed that the transcription factor Erg is specifically<br />

expressed in developing limb synovial joints during mouse and chick<br />

embryogenesis. Erg belongs to the ets family of transcription factors.<br />

The current 29 members regulate a variety of developmental processes<br />

and when mutated, can cause severe pathologies (3-4). They share the<br />

highly conserved 85-amino<br />

acid ets domain that binds to the core DNA core motif 5’-GGA(A/T)-3’<br />

and displays a winged helix-turn-helix<br />

organization with three α-helices and four β-sheets (5). Based on<br />

the structure of other domains and in particular the Pointed/SAM<br />

domain (6), the proteins are distinguished into ets sub-families. This<br />

is of significant relevance because the SAM domains mediate proteinprotein<br />

interactions and regulate interactions of Ets members with other<br />

SAM- and non-SAM containing nuclear proteins, thus exerting a major<br />

influence on function and bioactivity (7). Ets protein-protein interactions<br />

are also modulated by phosphorylation and conformational changes.<br />

Based on their similar SAM domain, Erg, Fli-1 and Fev constitute one<br />

such ets sub-family.<br />

We found that Erg is expressed in the mesenchymal interzone together<br />

with the signaling proteins Gdf5 and Wnt9a at very early stages of<br />

joint formation and is then expressed in articular chondrocytes (8-9).<br />

To study function, we over-expressed Erg in mice under the control<br />

of cartilage-specific Col2a1 regulatory sequences and found that the<br />

entire skeleton of the transgenic mice had remained cartilaginous and<br />

failed to undergo endochondral ossification (9). This led us to conclude<br />

that Erg may be important to establish and maintain the permanent<br />

phenotype of articular chondrocytes. To test this possibility directly, we<br />

have now created floxed Erg mice and examined the consequences of<br />

conditional Erg deletion on short and long term formation and function<br />

of articular cartilage.<br />

Content: To test the possible roles of Erg in joint formation, we first<br />

created floxed Erg mice by targeting an essential protein-encoding<br />

exon. The floxed Erg mice were then mated with mice that express Cre<br />

recombinase under the control of Gdf5 regulatory sequences. We had<br />

previously used the Gdf5-Cre mice to conditionally delete genes in early<br />

developing joints, including Ext1 (10). We had used them also to carry<br />

out genetic track-and-trace experiments after mating with RosaR26R<br />

mice to identify the origin of joint progenitor cells; we showed for the<br />

first time that the joint tissues are for the most produced by interzone<br />

mesenchymal cells only (11). The compound Gdf5-Cre/Erg-mutant<br />

mice were born at mendelian ratios but unexpectedly, did not exhibit<br />

overt joint defects when young. To account for absence of a major joint<br />

developmental phenotype, we asked whether the ets subfamily member<br />

Extended Abstracts 107<br />

Fli-1 was co-expressed and in fact it was, and may have compensated<br />

for Erg absence. To test possible postnatal roles of Erg, we subjected<br />

2 month-old Erg-deficient mice to knee’s medial collateral ligament<br />

(MCL) transection to induce experimental osteoarthritis (OA) (12).<br />

Strikingly, the Erg-deficient mice developed serious OA-like defects<br />

far sooner than operated wild type companions. Indeed, we observed<br />

similar severe OA-like defects in aging un-operated 7-11 month-old<br />

Erg-deficient mice, while control littermates displayed mild defects. To<br />

gain insights into how Erg maintains long-term articular chondrocyte<br />

function, we focused on parathyroid hormone-related protein (PTHrP)<br />

which is also expressed in developing joints, stabilizes the chondrocyte<br />

phenotype (13) and prevents chondrocyte hypertrophy when overexpressed<br />

in cartilage (just as transgenic Erg over-expression does).<br />

We found that Erg (as well as Fli-1) stimulates PTHrP expression and<br />

the PTHrP gene promoter contains several conserved ets binding sites<br />

needed for responsiveness.These and other data lead us to conclude<br />

that Erg regulates the development and long-term stabilization and<br />

function of articular chondrocytes and does so in cooperation with<br />

transcription factor subfamily member Fli-1 and with the signaling<br />

protein PTHrP.<br />

References:<br />

1. Pacifici M, Koyama E, Iwamoto M 2005 Mechanisms of synovial joint<br />

and articular cartilage formation: recent advances, but many lingering<br />

mysteries. Birth Defects Research, Pt. C 75:237-248.<br />

2. Kronenberg HM 2003 Developmental regulation of the growth plate.<br />

Nature 423:332-336.<br />

3. Sharrocks AD 2001 The ets-domain transcription factor family. Nature<br />

Reviews Mol. Cell Biol. 2:827-837.<br />

4. Gutierrez-Hartmann A, Duval DL, Bradford AP 2007 ETS transcription<br />

factors in endocrine systems. Trends Endocrinol. Metab. 18:150-158.<br />

5. Karim FD, Urness LD, Thummel CS, Klemsz MJ, McKercher SR, Celada<br />

A, Van Beveren C, Maki C, Gunther RA, Nye JA, Graves BJ 1990 The ETSdomain:<br />

a purine-rich core DNA sequence. Genes Dev. 4:1451-1453.<br />

6. Qiao F, Bowie JU 2005 The many faces of SAM. Science STKE<br />

286:re7<br />

7. Papoutsopolou S, Janknecht R 2000 Phosphorylation of ETS<br />

transcription factor ER81 in complex with its coactivators CREB-binding<br />

protein and p300. Mol. Cell. Biol. 20:7300-7310.<br />

8. Iwamoto M, Higuchi Y, Koyama E, Enomoto-Iwamoto M, Yeh H,<br />

Abrams WR, Rosenbloom J, Pacifici M 2000 Transcription factor ERG<br />

variants abone development. J. Cell Biol. 150:27-39.<br />

9. Iwamoto M, Tamamura Y, Koyama E, Komori T, Takeshita N, Williams<br />

JA, Nakamura T, Enomoto-Iwamoto M, Pacifici M 2007 Transcription<br />

factor ERG and joint and articular cartilage formation during mouse<br />

limb and spine skeletogenesis. Dev. Biol. 305:40-51.<br />

10. Mundy C, Yasuda T, Yamaguchi Y, Iwamoto, M. Enomoto-Iwamoto<br />

M, Koyama E, Pacifici M 2011 Synovial joint formation require slocal<br />

Ext1 expression and heprana sulfate production in developing mouse<br />

limbs and spine. Dev. Biol. 351:70-81.<br />

11. Koyama E, Shibukawa Y, Nagayama M, Sugito H, Young B, Yuasa T,<br />

Okabe T, Ochiai T, Kamiya N, Rountree RB, Kingsley DM, Iwamoto M,<br />

Enomoto-Iwamoto M, Pacifici M 2008 A distinct cohort of progenitor<br />

cells participates in synovial jskeletogenesis. Dev. Biol. 316:62-73.<br />

12. Kamekura S, Hoshi K, Shimoaka T, Chung U-I, Chikuda H, Yamada<br />

T, Uchida M, Ogata N, Seichi A, Nakamua K, Kawaguchi H 2005<br />

Osteoarthritis development ininduced by knee joint instability.<br />

Osteoarthr. Cart. 13:632-641.<br />

13. Macica CM, Liang G, Nasiri A, Broadus AE 2011 Genetic evidence of<br />

the regulatory role of parathyroid hormone-related protein in articular<br />

chondrocyte maintenance in an experimental mouse model. Arthr.<br />

Rheum. 63 3333-3343.<br />

Acknowledgments:<br />

We thank our colleagues for essential contributions to this research.<br />

We also thank Dr. D. Kingsley for providing the Gdf5-Cre mouse line.<br />

The research work was supported by NIH RO1 grants AR046000 and<br />

AR062908.


108<br />

Extended Abstracts<br />

2.1.3<br />

Growth factors in cartilage homeostasis in vitro / in vivo<br />

S. Chubinskaya<br />

Chicago/United States of America<br />

Introduction: <strong>Cartilage</strong> repair and regeneration is a major obstacle in<br />

orthopedics. The importance is enormous since osteoarthritis (OA) is<br />

a primary cause of disability among the adult population in the United<br />

States. OA is a disease of the joint, in which a process of attempted,<br />

but gradually failing, repair of damaged cartilage extracellular<br />

matrix occurs due to imbalance between synthesis and breakdown<br />

of matrix components. Currently, the only treatments available for<br />

cartilage damage are surgical interventions, such as microfracture,<br />

articular chondrocyte transplantation, autografting, allografting,<br />

debridement, lavage, and joint replacement at the end-stage<br />

disease. Innovations in bioengineering include the use of juvenile<br />

cartilage, isolated chondrocytes, stem cells together with scaffolds<br />

and various polymeric matrices, but those are still in development.<br />

To the best of our knowledge none of the existing approaches could<br />

regenerate normal adult hyaline cartilage that is able to perform<br />

required functions, sustain the load and integrate with the host<br />

tissue. Importantly, newly repaired tissue due to its imperfect<br />

structural organization may also be more susceptible to re-injury.<br />

From a therapeutic potential, there is no drug on the market that<br />

can halt the destruction of the joint seen in OA, though the progress<br />

has been made in disease-modifying OA drugs aimed to target<br />

several molecular pathways involved in the pathophysiology of OA.<br />

These include bisphosphonates, tetracyclines, metalloproteinase<br />

inhibitors, diacerein and cytokine antagonists, OA targeted gene<br />

therapy, and growth factors. In this presentation we will review the<br />

current knowledge on the use of growth factors for cartilage repair<br />

and their overall role in cartilage homeostasis.<br />

Content: Growth factors appear to be the most promising agents to<br />

stimulate anabolic responses and repair of articular cartilage. The<br />

most known are the members of the Transforming Growth Factor<br />

(TGF)-β superfamily including the Bone Morphogenetic Proteins<br />

(BMPs), Insulin-Like Growth Factor-1 (IGF-1), Fibroblast Growth<br />

Factors (FGF), especially FGF-2 and FGF-18, Epidermal Growth Factor,<br />

Vascular-Endothelial Growth Factors (VEGFs), and other. TGF-β<br />

is widely used in various animal models of OA and in stem cells<br />

research as the growth factor that drives chondrogenesis. In articular<br />

chondrocytes TGF-β has been shown to induce both catabolic and<br />

anabolic responses and its utilization for cartilage repair remains<br />

to be questionable due to its ability to stimulate tissue fibrosis and<br />

formation of osteophytes. IGF-1 is another anabolic factor, which<br />

chondroprotective activity was primarily demonstrated in various<br />

animal models of OA. In humans, however, the responsiveness to<br />

IGF-1 is significantly reduced with ageing, which limits its application<br />

for human cartilage repair. FGFs are important regulators of cartilage<br />

development and homeostasis. FGF-2 can stimulate cartilage repair<br />

responses, but its potent mitogenic effects may lead to chondrocyte<br />

cluster formation and poor extracellular matrix due to a relatively<br />

low level of type II collagen. FGF-2 has been also shown to induce<br />

pro-catabolic and pro-inflammatory responses. In a rabbit ACL<br />

transection model, however, sustained release formulations of<br />

FGF-2 reduced OA severity. Another member of the same family,<br />

FGF-18, has been shown to induce anabolic effects in chondrocytes<br />

and chondroprogenitor cells, and to stimulate cell proliferation<br />

and type II collagen production. In a rat meniscal tear model of OA,<br />

intra-articular FGF-18 injections induced a remarkable formation of<br />

new cartilage and reduced the severity of the experimental lesions.<br />

Thus far, only two anabolic factors, FGF-18 and BMP-7, are being<br />

tested in clinical studies in patients with established OA. FGF-18<br />

was investigated in a randomized, double-blind, placebo controlled<br />

multicenter studies in patients with primary knee OA, scheduled for<br />

total knee replacement. No local or systemic concerns were reported<br />

for single or multiple injections of FGF-18 and positive biological<br />

activity was documented on joint histology (cartilage damage/OA<br />

severity) and biomechanical properties (cartilage stiffness). However,<br />

no difference with placebo treatment was found in joint space<br />

narrowing and cartilage thickness. The members of the BMP family<br />

of proteins are important stimuli of mesenchymal cell differentiation<br />

and extracellular matrix formation. Few of them, BMP-2 and BMP-7,<br />

appear to be extremely potent in cartilage and bone repair. The most<br />

studied BMP for human and animal cartilage repair is BMP-7, also<br />

known as osteogenic protein-1 (OP-1). The results suggest that BMP-<br />

7 may be the best candidate thus far as a disease-modifying drug<br />

for OA and post-traumatic OA (PTOA). Unlike TGF-β and other BMPs,<br />

BMP-7 up-regulates chondrocyte metabolism and protein synthesis<br />

without creating uncontrolled cell proliferation and formation of<br />

osteophytes. BMP-7 prevents chondrocyte catabolism induced by<br />

IL-1 or fragments of matrix components. It has synergistic anabolic<br />

effects with other growth factors such as IGF-1, which in addition to<br />

its anabolic effect acts as a cell survival agent. Importantly, BMP-<br />

7 restores the responsiveness of human chondrocytes to IGF-1 lost<br />

with ageing through the regulation of IGF-1 and its signaling pathway.<br />

In our most recent studies on BMP-7 in acute ex vivo cartilage<br />

trauma model BMP-7 stimulated PG synthesis and preserved matrix<br />

integrity. The treatment with BMP-7 also significantly promoted cell<br />

survival in the impacted region and prevented the expansion of cell<br />

death and matrix degeneration into the adjacent, but not impacted<br />

regions. Furthermore, BMP-7 has been used in various PTOA animal<br />

models in dogs, sheep, goats and rabbits. In all these PTOA models<br />

(ACL transaction, osteochondral defect, and impaction), BMP-7<br />

regenerated articular cartilage, increased repair tissue formation<br />

and improved integrative repair between new cartilage and the<br />

surrounding articular surface. Critically, in a study with sheep<br />

impaction model a window of opportunity for the treatment with<br />

BMP-7 has been identified. It was found that therapeutic application<br />

of BMP-7 was the most effective in arresting progression of cartilage<br />

degeneration if administered twice with a weekly interval either<br />

immediately after trauma or delayed by three weeks. If delayed by<br />

three month, the treatment was ineffective. These important results<br />

suggest that the development and progression of PTOA could<br />

be arrested and maybe even prevented if the right treatment is<br />

administered at the right time. In addition, strong pro-anabolic and<br />

anti-catabolic properties of BMP-7 have been demonstrated in gene<br />

array studies with human chondrocytes and in disc degeneration<br />

animal models, where BMP-7 was also effective as an anti-pain<br />

treatment. Phase I clinical study produced very encouraging results<br />

by showing tolerability to the treatment, absence of toxic response,<br />

and a greater symptomatic improvement in patients that received a<br />

single injection of BMP-7. At this stage of our cumulative knowledge,<br />

BMP-7 appears to be one of the best candidate therapeutic agents<br />

for cartilage repair since it affects multiple catabolic and anabolic<br />

pathways.<br />

References:<br />

Anderson, DD; Chubinskaya, S; Guilak, F; Martin, JA; Oegema, TR;<br />

Olson, SA and Buckwalter, JA. (2011). Post-traumatic osteoarthritis:<br />

improved understanding and opportunities for early intervention.<br />

J Orthop Res, 29(6):802-9. Chubinskaya, S; Hurtig, M and Rueger,<br />

DC. (2007). OP-1/BMP-7 in cartilage repair. Int Orthop, 31(6):773-81.<br />

Chubinskaya, S; Otten, L; Soeder, S; Borgia, JA; Aigner, T; Rueger, DC<br />

and Loeser, RF. (2011). Regulation of chondrocyte gene expression<br />

by osteogenic protein-1. Arthritis Res Ther, 13(2):R55. Ellman,<br />

MB; An, HS; Muddasani, P and Im, HJ. (2008). Biological impact<br />

of the fibroblast growth factor family on articular cartilage and<br />

intervertebral disc homeostasis. Gene, 420(1):82-9. Ellsworth, JL;<br />

Berry, J; Bukowski, T; Claus, J; Feldhaus, A; Holderman, S; Holdren,<br />

MS; Lum, KD; Moore, EE; Raymond, F; Ren, H; Shea, P; Sprecher,<br />

C; Storey, H; Thompson, DL; Waggie, K; Yao, L; Fernandes, RJ; Eyre,<br />

DR and Hughes, SD. (2002). Fibroblast growth factor-18 is a trophic<br />

factor for mature chondrocytes and their progenitors. Osteoarthritis<br />

<strong>Cartilage</strong>, 10(4):308-20. Fortier, LA; Mohammed, HO; Lust, G and<br />

Nixon, AJ. (2002). Insulin-like growth factor-I enhances cell-based<br />

repair of articular cartilage. J Bone Joint Surg Br, 84(2):276-88. Lotz,<br />

MK and Kraus, VB. (2010). New developments in osteoarthritis.<br />

Posttraumatic osteoarthritis: pathogenesis and pharmacological<br />

treatment options. Arthritis Res Ther, 12(3):211. Dahlberg LE,<br />

Flechsenhar K, Gouteux S, Jurvelin JS. A randomized, double-blind,<br />

placebo controlled, multicenter center study of rhFGF-18 administered<br />

intra-articularly using single or multiple ascending doses in patients<br />

with primary knee osteoarthritis, scheduled for knee replacement.<br />

OARSI, San Diego, 2011.<br />

Acknowledgments:<br />

This work was supported by the NIH/NIAMS grants, Stryker Biotech<br />

research support, National Football League Charities, Ciba-Geigy<br />

Endowed Chair and Department of Biochemistry, Rush University<br />

Medical Center. The authors would like to acknowledge Dr. Arkady<br />

Margulis for tissue procurement and Dr. Lev Rappoport and Mrs.<br />

Arnavaz Hakimiyan for their technical assistance. The authors also<br />

would like to acknowledge the Gift of Hope Organ & Tissue Donor<br />

Network and donor’s families.


2.2.1<br />

Nerve dependence and cartilage development during Urodele<br />

limb regeneration and its relevance to mammals.<br />

M. Maden<br />

Gainesville/United States of America<br />

Content: When the Urodele limb is amputated anywhere along its<br />

length it can replace a perfect copy of itself in a remarkably rapid<br />

time. Following amputation and wound healing the internal tissues<br />

- muscle, cartilage/bone, dermis - dedifferentiate to liberate a<br />

population of apparently pluripotent cells, called the blastema, which<br />

proliferate and redifferentiate into the missing structures. Although<br />

initial studies suggested that these blastemal cells were pluripotent,<br />

for example, if the cartilage is removed from the stump, new cartilage<br />

nevertheless regenerates distal to the amputation plane, recent<br />

studies have revealed far less interchange between tissue types.<br />

We have shown, using transgenically labeled cells grafted into the<br />

regenerating limb, that there is a considerable degree of lineage<br />

preservation especially with regard to the muscle. <strong>Cartilage</strong> can,<br />

however, be regenerated from the dermal fibroblasts of the stump<br />

and these experiments will be described. The dermal fibroblast<br />

seems to be the ‘stem cell’ of the regenerating limb and in addition<br />

to being able to undergo this fate change to generate cartilage, has<br />

considerable organizing properties in terms of patterning.<br />

Proliferation of the blastemal cells in the regenerate has long been<br />

known to be a nerve dependent phenomenon. In 1823 it was revealed<br />

that severing of the nerves supplying the newt limb prevented<br />

regeneration and ideas on the role that nerves played were generated<br />

in the 1950’s in a careful series of experiments showing that the<br />

effect was quantitative and depended on the number of nerve fibres<br />

remaining at the amputation plane. Most recently the identity of the<br />

factor that the nerves provide has been found to be newt Anterior<br />

Gradient protein which is secreted by the Schwann cells of the distal<br />

nerve sheath and then by the gland cells in the wound epidermis<br />

covering the amputated limb. Our recent studies on the neural<br />

control of regeneration and the role of retinoic acid have led us to<br />

show that the maintenance of the differentiated cartilage phenotype<br />

is retinoic acid dependent since the application of an antagonist to<br />

retinoic acid signaling to the fully formed axolotl limb causes the<br />

cartilage to regress and the limb to shrink. We will describe our<br />

investigations into this aspect of cartilage maintenance.<br />

Our regeneration studies also encompass mammals and in particular<br />

the regeneration of ear tissue following circular punches. It is known<br />

that rabbits can fully regenerate after such damage, but we have<br />

found a species of mouse that also perform this surprising feat and<br />

can thus regenerate cartilage. We consider this regenerative property<br />

to be due to be the formation of a blastema which shows striking<br />

similarities to the Urodele limb blastema and these experiments will<br />

also be described.<br />

2.2.2<br />

Neurogenic factors in the etiopathogenesis of osteoarthritis<br />

D. Walsh<br />

Notthingham/United Kingdom<br />

Introduction: Osteoarthritis (OA) typically presents with pain, and<br />

yet how changes in the joint lead to pain in OA remains incompletely<br />

understood. Furthermore, the nervous system influences normal<br />

joint function, and may contribute to OA pathogenesis.<br />

Content: OA is often considered a disease of articular cartilage, but<br />

this tissue, as with the inner 2 thirds of the meniscus, contains no<br />

nerves in the normal joint in which it cannot therefore be a direct<br />

source of pain. Synovitis and subchondral bone marrow lesions<br />

have been associated with pain in imaging studies, although it<br />

remains unclear whether these lesions are themselves painful, or<br />

surrogate biomarkers of other pathology within the joint. Peripheral<br />

sensitization of nerves within the joint contributes to OA pain.<br />

Sensitization may be induced by inflammatory mediators (e.g. kinins,<br />

prostaglandins), or growth factors (e.g. nerve growth factor, NGF),<br />

produced both in OA synovium and subchondral bone. Clinical trials<br />

have confirmed the relative importance of such factors in OA pain.<br />

Subchondral nerves are normally protected from mechanical and<br />

chemical stimuli by force dissipation in the articular cartilage and<br />

subchondral bone plate, and by the relatively impermeable tidemark<br />

at the osteochondral junction. Exposure of subchondral nerves,<br />

Extended Abstracts 109<br />

due to abnormal biomechanical properties and permeabilisation<br />

of the osteochondral junction may further contribute to OA pain.<br />

In OA, sensory nerves invade through vascularised channels that<br />

extend from subchondral bone into articular cartilage and chondroosteophytes.<br />

Similarly nerves grow towards the meniscal tip during<br />

the meniscal degeneration that accompanies knee OA. Sensory<br />

innervation of previously non-innervated structures in the joint<br />

may also contribute to pain in OA, even if subjected only to normal<br />

mechanical stresses. Mechanisms of sensitization, joint damage<br />

and nerve growth are closely inter-related. Synovitis contributes<br />

to both joint damage and peripheral sensitization. Treatments that<br />

help maintain integrity of the osteochondral junction reduce pain<br />

in OA. Sensory nerve growth follows blood vessel growth, both in<br />

time and in space, and nerve growth is integrated with angiogenesis.<br />

Nerve growth and angiogenic factors are each upregulated in OA,<br />

and recent preclinical studies and clinical trials have demonstrated<br />

the potential that blocking NGF or angiogenesis may reduce OA<br />

pain. Much mechanistic understanding of OA derives from animal<br />

models, although existing models may have limitations in predicting<br />

responses to new treatments in man. OA is a heterogeneous disease,<br />

and no single model can mimic all aspects or stages of OA. Models,<br />

however, are helping to define the mechanisms, timecourse and<br />

interplay between peripheral and central sensitization, cartilage<br />

damage, osteochondral and subchondral pathology, osteophyte<br />

formation and nerve growth. Angiogenic and nerve growth factors<br />

overlap in their functions. NGF can stimulate angiogenesis.<br />

Neuropilin-1 on endothelial cells is a co-receptor for vascular<br />

endothelial growth factor (VEGF), and on sensory nerves binds the<br />

nerve guidance factors; semaphorins. Semaphorins inhibit both<br />

angiogenesis and sensory axonal growth. Angiogenesis facitiltates<br />

inflammation and is a prerequisite for endochondral ossification<br />

characteristic of osteophytes and at the osteochondral junction in<br />

OA. Factors that regulate nerve growth may also directly influence<br />

cartilage function in OA. NGF, its receptors and neuropilin-1<br />

are expressed in OA cartilage, and semaphorin-3A and -3F by<br />

hypertrophic chondrocytes. Furthermore, nerve-derived factors may<br />

additionally moderate chondrocyte function. Nerve-derived factors<br />

also regulate inflammation and vascular function in the synovium.<br />

The neuropeptides substance P and calcitonin gene-related peptide<br />

(CGRP) are released by peripheral terminals of sensory nerves within<br />

the joint, where they bind specific receptors on adjacent blood vessels<br />

and increase permeability and vasodilation respectively. These<br />

vasoactive neuropeptides can also initiate angiogenesis, and new<br />

blood vessel growth is emerging as one of the factors that determines<br />

persistence rather than resolution of synovitis. Neuropeptides may<br />

also regulate chondrocyte function, although whether this signifies<br />

neurogenic influences or autocrine or paracrine pathways from nonneuronal<br />

peptide sources remains unclear. Neurovascular factors<br />

are important in maintaining tissue phenotype. Angiogenesis into<br />

joint cartilage is associated with its ossification. Similarly, sensory<br />

innervation may be a beneficial facilitator of repair in some tissues<br />

(e.g. skin and bone), but may contribute to heterotypia in repairing<br />

cartilage defects. Inhibition of blood vessel and nerve growth may<br />

be important to ensure symptomatic benefit from tissue engineering<br />

approaches to cartilage repair. As well as having direct effects within<br />

the joint, neurogenic factors make important contributions to joint<br />

function through proprioceptive and neuromuscular pathways.<br />

Alterations in central processing of afferent signals in OA lead to<br />

altered body image and proprioceptive feedback. These contribute<br />

to symptoms, distress and disability, but also, potentially lead the<br />

joint to be subject to abnormal biomechanical stress. Furthermore,<br />

joint pain leads to muscle inhibition, both through moderation of<br />

spinal reflexes, and through changes in neurotrophic support to<br />

the musculature. Muscle weakness further contributes to disability,<br />

and exacerbates biomechanical stresses and consequential damage<br />

and pain. Neurogenic influences on the OA joint may be both<br />

beneficial and harmful. Nodal OA developing after neuronal damage<br />

may spare the affected joints, although it is unclear whether this<br />

effect is purely biomechanical due to reduced usage, or represents<br />

important neurogenic contributions to OA evolution. Conversely,<br />

sensory neuropathy may be associated with a severe destructive but<br />

hypertrophic arthropathy (named after Charcot), which may share<br />

some pathological features with OA. Analgesic-associated joint<br />

damage, however, may be more likely attributable to `off target’<br />

effects of specific agents, rather than exacerbation of OA pathology<br />

by sensory inhibition. Accelerated joint damage associated with<br />

corticosteroids, non-steroidal anti-inflammatory agents, and, most<br />

recently anti-NGF treatments is unlikely to be explained by a common<br />

analgesic mechanism. The precise contributions of neurogenic<br />

factors to OA etiopathogenesis remain to be completely elucidated.<br />

What is clearer is that nerves within the joint critically determine the<br />

symptoms of OA. Failure of total hip replacement to satisfactorily<br />

relieve OA pain in up to 15% of patients may point to important<br />

contributions of peri-articular structures and abnormal central pain


110<br />

Extended Abstracts<br />

processing. However, undoubted benefits for the large majority<br />

indicate pivotal roles for the articular surface and subchondral<br />

bone, at least in late disease. Disease modification must be directed<br />

at structural changes that are relevant to patients’ symptoms,<br />

particularly pain. Better understanding the symptomatic relevance<br />

of changes within the joint, and the interactions between nerves and<br />

their surrounding tissues, should lead to important advances in OA<br />

therapy in the near future.<br />

References:<br />

Mapp PI, Walsh DA. Angiogenesis in osteoarthritis: Mechanisms of<br />

disease and potential therapeutic targets. Nat. Rev. Rheumatol. (In<br />

press).<br />

Suri S, Walsh DA. Osteochondral alterations in osteoarthritis. Bone<br />

(In press) DOI: 10.1016/j.bone.2011.10.010<br />

Ashraf, S, Wibberley, H, Mapp, PI, Hill, R, Wilson, D, Walsh, DA.<br />

Increased vascular penetration and nerve growth in the meniscus:<br />

a potential source of pain in osteoarthritis. Ann. Rheum. Dis. 2011,<br />

70, 523-529.<br />

Walsh DA, McWilliams DF, Turley MJ, Dixon M, Fransès RE, Mapp PI,<br />

Wilson D. Angiogenesis and nerve growth factor at the osteochondral<br />

junction in rheumatoid arthritis and osteoarthritis Rheumatology<br />

2010, 49, 1852-61.<br />

Acknowledgments:<br />

I am grateful to the members of the Arthritis Research UK Pain Centre,<br />

and to patients and public who contribute to our research.<br />

2.2.3<br />

<strong>Cartilage</strong> lesions; which lesions are painful and what is the cause<br />

of pain ?<br />

M. Brittberg<br />

Kungsbacka/Sweden<br />

Introduction: There exist today a large number of techniques to treat<br />

and repair articular cartilage defects. However, there is no consensus<br />

of which defects that are needed to be treated and no information<br />

about which defects are causing the pain situation for the patients.<br />

When one detect a large cartilage defect in a joint surface and there<br />

are no other pathological findings, one may easily assume that<br />

this defect is the cause of the patient’s joint disability. The defect<br />

is repaired and in some patients the pain situation disappears<br />

but in some patients the pain remains even though arthroscopy<br />

examinations and MRI shows a nice repair. Pain development in a<br />

joint with isolated cartilage lesions is maybe different compared<br />

to pain in an organ disease like osteoarthritis. However, the pain<br />

mechanisms could partly use the same channels of information, the<br />

nerves and neural peptides.<br />

Content: A system to detect and show where the pain is and where<br />

it emanate from is crucial Elevated stress in the subchondral bone<br />

has been discussed as being a pain cause (Draper et al, 2011). To<br />

measure such stress is difficult. As part of pain studies, intraosseous<br />

pressure has been measured. Björkström et al. could note an increased<br />

pressure in a patella with chondromalcia and in patella OA versus a low<br />

pressure in a control normal patella (Björkström et al, 1980). Elevated<br />

subchondral stresses also induce an increased metabolic activity<br />

(Draper et al, 2011). Such an activity could be identified by scintigraphic<br />

examination. A MRI could show subchondral oedema which also could<br />

be due to such an increased metabolic activity. In chronic medial knee<br />

pain, increased tracer uptake in bone scintigraphy is more sensitive<br />

for medial knee pain than bone marrow oedema pattern on MRI (Buck<br />

et al. 2009). However, to use different sequences could help us better<br />

differentiate between the potential painful and not pain full lesions<br />

if pain could be related to a local subchondral oedema. Hayashi<br />

et al.(2011) studied semi quantitative assessment of subchondral<br />

bone marrow oedema-like lesions and subchondral cysts using<br />

intermediate-weighted (IW) fat-suppressed (fs) spin echo and Dual<br />

Echo Steady State (DESS) sequences on 3 T MRI. They showed that<br />

in a direct comparison an IW fs sequence depicted more subchondral<br />

bone marrow oedema-like lesions and better demonstrated the extent<br />

of their maximum size. A DESS sequence helped in the differentiation<br />

of subchondral bone marrow oedema-like lesions and subchondral<br />

cysts.Subchondral cysts may be involved in pain due to intra-cystic<br />

pain. The authors suggested that the IW fs sequence should be used<br />

for determination of lesion extent whenever the size of subchondral<br />

bone marrow oedema-like lesions is the focus of attention. Single<br />

photon emission computed tomography (SPECT) could be used to<br />

assess the physiology and homeostasis of subchondral bone adjacent<br />

to untreated and treated articular cartilage defects(Vellala et al, 2007,<br />

Hirschmann et al, 2011). Draper et al.(2012)found that painful knees<br />

exhibited increased tracer uptake when using PET/CT compared to<br />

the pain-free knees of four subjects with unilateral pain (P = 0.0006).<br />

They also also found a correlation between increasing tracer uptake<br />

and increasing pain intensity Possible sources of pain in patients<br />

with OA include the synovial membrane, joint capsule, periarticular<br />

ligaments,periosteum, and subchondral bone (Grubb et al, 2004). The<br />

subchondral bone related causes of pain include periostitis associated<br />

with osteophyte formation, subchondral micro fractures, bone angina<br />

due to decreased blood flow and elevated intraosseous pressure,<br />

and bone marrow lesions detected on magnetic resonance imaging<br />

(MRI). Such sources of pain could possibly also be part of the pain<br />

in isolated cartilage lesions. The synovial lining could also produce<br />

pain by a pain irritation of sensory nerve endings within the synovium<br />

from osteophytes and synovial inflammation caused by to the release<br />

of prostaglandins, leukotrienes, and cytokines. (Wong et al, 1993,<br />

Dirmeyer et al, 2008) The subchondral bone involvement is possibly<br />

much more important in the success of cartilage repair techniques and<br />

such bone involvement could be part of the development of OA and the<br />

following pain situation. Bone cells are under the influence of different<br />

systemic and local auto/paracrine factors. One such regulatory factor<br />

that can play both a sensory/ afferent and a regulatory/efferent role<br />

consists of neuropeptide-containing nerves. In particular, the calcitonin<br />

gene-related peptide (CGRP) (Brain et al. 1985, Kruger et al, 1999),<br />

substance P (Bjurholm et al, 1988, Halliday et al, 1993) and vasoactive<br />

intestinal peptide (VIP)(Rahmann et al. 1992) have been suggested to<br />

be involved in such regulatory loopes. Of interest to know is also the<br />

fact that chondrocytes have receptors for substance P (Millward-Sadler<br />

et al. 2004). In 1969, Greenwald and Hayes found that a pathway for<br />

dye from the medullary cavity to the articular cartilage in the human<br />

femoral head does exist. In 1994 Milz and Putz demonstrated also the<br />

existence of several channels between the subchondral region and<br />

the uncalcified cartilage into the cartilage. Early subchondral changes<br />

include redistribution of blood supply with marrow hypertension and<br />

bone marrow oedema. The chock absorption properties of articular<br />

cartilage depend on the combined unit of the cartilage layer and the<br />

subchondral bone layer giving the articular cartilage its visco elastic<br />

properties. Disturbances of the unit in isolated lesions as well as<br />

in widespread OA may elicit disturbances in the subchondral bone<br />

signalling system with a subsequent pain development. Highly<br />

vascularized areas in the human body are present in regions of high<br />

functional activity with high needs for nutrition. The subchondral bone<br />

in OA is such a region and it is connected with the basal layers of the<br />

cartilage via vascular channels. When the fine homeostatic balance is<br />

disturbed, secretions of neuropeptides in the subchondral region may<br />

occur and could be transported via the channels to the cartilage and<br />

subsequently direct influence receptors on the chondrocytes (Suri et<br />

al, 2007). A large number of dorsal root ganglion cells are small and<br />

medium sized, and sometimes called “type B” ganglion (BG) These<br />

neurons generally emit unmyelinated and thinly myelinated axons most<br />

commonly associated with nociception, and a substantial proportion of<br />

these neurons express one, or generally several neuropeptides Sensory<br />

nerve fibres (NFs) contain two major neuropeptides, substance P (SP)<br />

and calcitonin gene-related peptide (CGRP).(Edoff et al, 2000, 2001,<br />

2003). The distribution of axons displaying immunoreactivity (IR) for<br />

the peptide, substance (SP), has been examined in numerous tissues<br />

primarily by using immunofluorescence methods (Bjurholm et al,<br />

1988).CGRP immunoreactivity (CGRP-IR) is the most prevalent of BG<br />

neuropeptide markers, with neurons containing several other peptides<br />

constituting subsets of these CGRP-IR BG neurons (Buma at el. 1992).<br />

Not enough is known of the biological effects of CGRP, although it is<br />

the most potent vasodilator among endogenous peptides and CGRPIR<br />

nerve fibres are also associated with vascular edema. Vascularisation<br />

and the associated innervation of articular cartilage may contribute to<br />

tibiofemoral pain in OA as sympathetic and sensory nerves have been<br />

found to be present within vascular channels in the articular cartilage, in<br />

both mild and severe OA. Perivascular and free nerve fibres, and nerve<br />

trunks have bee observed within the subchondral bone marrow and<br />

within the marrow cavities of osteophytes. Such nerve ingrowth and<br />

neuropeptide influence via the channels as well as the direct contact<br />

seen in osteochondral defects may induce localized pain from the<br />

lesions. Future research on cartilage repair has to be focused also on<br />

the subchondral bone metabolic activity. The vascularity involvement<br />

is of great interest with subsequent neural ingrowth with possible<br />

effect on pain appearance (Neugebauer et al. 1995).


References:<br />

1.Bjurholm, A, A. Kreicbergs, E. Brodin, and M. Schultzberg (1988)<br />

Substance P and CGRP-immunoreactive nerves in bone. Peptides 165-<br />

171.<br />

2.Brain, S.D., and T.J. Williams (1985) Inflammatory oedema induced<br />

by synergism between calcitonin gene-related (CGRP) and mediators of<br />

increased vascular permeability. Br. J. Pharmacol. 855-860.<br />

3.Brain, S.D.,T.J. Williams, J.R. Tippins, H.R. Morris, andI. Maclntyre(1985)<br />

Calcitonin gene-related peptide is a potent vasodilator. Nature 313.54-<br />

56.<br />

4.Buck FM, Hoffmann A, Hofer B, Pfirrmann CW, Allgayer B. Chronic<br />

medial knee pain without history of prior trauma: correlation of pain<br />

at rest and during exercise using bone scintigraphy and MR imaging.<br />

Skeletal Radiol. 2009 Apr;38(4):339-47. Epub 2008 Dec<br />

5. Björkström S, Goldie IF, Wetterqvist H. Intramedullary pressure of the<br />

patella in Chondromalacia.Arch Orthop Trauma Surg. 1980;97(2):81-5.<br />

6. Buma P, Verschuren C, Versleyen D, Van der Kraan P, Oestreicher<br />

AB. Calcitonin gene-related peptide, substance P and GAP-43/B-50<br />

immunoreactivity in the normal and arthrotic knee joint of the mouse.<br />

Histochemistry. 1992 Dec;98(5):327-39.<br />

7. Lower density of synovial nerve fibres positive for calcitonin generelated<br />

peptide relative to substance P in rheumatoid arthritis but not in<br />

osteoarthritis. Rheumatology (Oxford). 2008 Jan;47(1):36-40<br />

8.Draper CE, Fredericson M, Gold GE, Besier TF, Delp SL, Beaupre GS,<br />

Quon A. Patients with patellofemoral pain exhibit elevated bone metabolic<br />

activity at the patellofemoral joint.J Orthop Res. 2012 Feb;30(2):209-13.<br />

doi: 10.1002/jor.21523. Epub 2011 Aug 2<br />

9. Neuropeptide content and physiological properties of rat cartilageprojecting<br />

sensory neurones co-cultured with perichondrial cells.<br />

Neurosci Lett. 2001 Nov 27;315(3):141-4<br />

10. Retrograde tracing and neuropeptide immunohistochemistry<br />

of sensory neurones projecting to the cartilaginous distal femoral<br />

epiphysis of young rats.Cell Tissue Res. 2000 Feb;299(2):193-200.<br />

11.Neuropeptide effects on rat chondrocytes and perichondrial cells<br />

in vitro.Neuropeptides. 2003 Oct;37(5):316<br />

12. A pathway for nutrients from the medullary cavity to the articular<br />

cartilage of the human femoral head. J Bone Joint Surg Br. 1969<br />

Nov;51(4):747-53<br />

13. Activation of sensory neurons in the arthritic joint. Novartis Found<br />

Symp. 2004;260:28-36; discussion 36-48, 100-4, 277-9.<br />

14. Hayashi D, Guermazi A, Kwoh CK, Hannon MJ, Moore C, Jakicic JM,<br />

Green SM, Roemer FW. Semiquantitative assessment of subchondral<br />

bone marrow edema-like lesions and subchondral cysts of the<br />

knee at 3T MRI: a comparison between intermediate-weighted fatsuppressed<br />

spin echo and Dual Echo Steady State sequences. BMC<br />

Musculoskelet Disord. 2011 Sep 9;12:198.<br />

15. The substance P fragment SP-(7-11) increases prostaglandin E2,<br />

intracellular Ca2+ and collagenase production in bovine articular<br />

chondrocytes.Biochem J. 1993 May 15;292 (Pt 1):57-62<br />

16.Hirschmann MT, Davda K, Rasch H, Arnold MP, Friederich NF.<br />

Clinical value of combined single photon emission computerized<br />

tomography and conventional computer tomography (SPECT/CT)<br />

in sports medicine. Sports Med Arthrosc. 2011 Jun;19(2):174-81.<br />

Review.<br />

17.Integrin-dependent signal cascades in chondrocyte<br />

mechanotransduction. Ann Biomed Eng. 2004 Mar;32(3):435-46.<br />

Review<br />

18. Peripheral patterns of calcitonin-gene-related peptide general<br />

somatic sensory innervation: cutaneous and deep terminations.J<br />

Comp Neurol. 1989 Feb 8;280(2):291-302.<br />

19. Involvement of substance P and neurokinin-1 receptors in the<br />

hyperexcitability of dorsal horn neurons during development of acute<br />

arthritis in rat‘s knee joint. J Neurophysiol. 1995 Apr;73(4):1574-83<br />

20. The regulation of connective tissue metabolism by vasoactive<br />

intestinal polypeptide. Regul Pept. 1992 Jan 23;37(2):111-21<br />

Extended Abstracts 111<br />

21. Neurovascular invasion at the osteochondral junction and in<br />

osteophytes in osteoarthritis. Ann Rheum Dis. 2007 Nov;66(11):1423-<br />

8. Epub 2007 Apr 19.<br />

22.Tiderius CJ, Svensson J, Leander P, Ola T, Dahlberg L. dGEMRIC<br />

(delayed gadolinium-enhanced MRI of cartilage) indicates adaptive<br />

capacity of human knee cartilage. Magn Reson Med. 2004<br />

Feb;51(2):286-90.<br />

23.Vellala RP, Mabjures S, Ryan PJ. Single photon emission computed<br />

tomography scanning in the diagnosis of knee pathology. J Orthop<br />

Syrg (HongKong) 2004; 12: 87-90 24. Neural mechanisms of joint<br />

pain. Ann Acad Med Singapore. 1993 Jul;22(4):646-50. Review<br />

2.3.1<br />

<strong>Cartilage</strong> defects in the femoropatellar joint<br />

B.J. Cole<br />

Chicago/United States of America<br />

Introduction: Chondral defects in the patellofemoral joint have<br />

varied etiologies. For example, the chondrosis may be genetically<br />

related, as with focal or diffuse degeneration secondary to trauma<br />

(direct impact or a result of patellofemoral instability) or secondary<br />

to repetitive microtrauma (e.g., excessive loads such as in jumping<br />

sports), or related to the cumulative microtrauma of biomechanical<br />

abnormalities (e.g., chronic patellar subluxation). High-grade<br />

(grades III and IV) focal chondral defects are reported to occur<br />

between 11% and 20% in patients undergoing knee arthroscopy.<br />

Of these defects, 11<strong>–</strong>23% involved the patella and 6<strong>–</strong>15% were<br />

trochlear. Not all of these lesions were symptomatic. In fact, some<br />

patients are asymptomatic even at very high functional levels.<br />

Kaplan et al. performed MRIs on asymptomatic NBA basketball<br />

players and found articular cartilage lesions in 47% of these<br />

players, with 50% of these lesions classified as high grade (III or IV).<br />

The patella was affected in 35%, and the trochlea in 25% of these<br />

players who were asymptomatic. Similarly, Walczak et al. found<br />

abnormal cartilage signal on MRI in 57% of asymptomatic NBA<br />

players with a 7% incidence of focal defects. Just as with other PF<br />

problems, symptoms and pathology have incomplete correlations.<br />

It is not entirely clear why some patients with PF chondral lesions<br />

present with pain while others can perform at a high level. Ficat<br />

and Hungeford proposed that the elevated intraosseous pressures<br />

seen in the face of an articular cartilage lesion could be the source<br />

of pain and today with MRI, it is not uncommon to see areas of<br />

bone overload associated with chondral lesions as evidenced by<br />

“bone bruises.” As noted, the articular cartilage is aneural, so pain<br />

other than bone may emanate from the soft tissues, including the<br />

joint capsule, ligaments, tendons, and synovium. In addition to<br />

mechanical factors, pain may be initiated in part by irritation from<br />

chondral debris, which activates an inflammatory and nociceptive<br />

response. As the true pain generator is often not well defined, it is<br />

crucial to thoroughly evaluate all potential sources of discomfort<br />

before attributing symptoms to a chondral defect.<br />

Content: Options for restoring articular cartilage are limited by the<br />

ability to replicate the complex three-dimensional anatomy of the<br />

PF joint. The decision of which cartilage restoration procedure to<br />

pursue depends on the size of the lesion, the level of demand the<br />

patient places on his/her knees, and whether a previous attempt<br />

at cartilage restoration has failed. In general, microfracture is the<br />

first-line therapy for low-demand patients and for smaller lesions.<br />

Though short-term results of microfracture of trochlear and patellar<br />

lesions have been acceptable, these results have deteriorate at<br />

midterm and long-term follow-up. The mechanical properties of the<br />

fibrocartilage created by microfracture may be poorly suited to the<br />

PF joint because of the high shear stresses. Caution in interpreting<br />

the literature describing the outcomes of microfracture is warranted.<br />

Because microfracture is relatively easy to perform and is often the<br />

first-line treatment for a symptomatic cartilage defect, comorbidities<br />

such as malalignment are often neglected at this early decisionmaking<br />

stage. If all the principles of comorbidity correction are<br />

adhered to, however, it is entirely possible that microfracture could<br />

perform more optimally in this patient population. Osteochondral<br />

allograft transplantation has also been used in the PF joint, although<br />

previous attempts have been limited to diffuse lesions as a salvage<br />

procedure. The complex topology of the PF joint and the requirement<br />

for high congruence have limited past attempts. Overall, results of<br />

osteochondral allograft transplantation have not been as successful<br />

for lesions on PF joint surfaces as they have been for lesions on


112<br />

Extended Abstracts<br />

the femoral condyles, but in some clinical scenarios osteochondral<br />

allograft transplantation remains an early treatment option. ACI is<br />

preferred for high-demand patients, patients with large lesions, and<br />

for those who have undergone a failed previous cartilage restoration<br />

attempt. ACI has been extensively studied, with the largest and most<br />

well-conducted trials of any cartilage restoration procedure used in<br />

the PF joint, and has the additional benefit of “self-conforming” to the<br />

topography of the joint. Contraindications to ACI in the PF joint include<br />

inflammatory arthritis, loss of subchondral bone, disease within the<br />

tibiofemoral joint, and a high likelihood of patient noncompliance with<br />

postoperative rehabilitation and restrictions. The potential benefits<br />

of ACI must be juxtaposed against the disadvantages, which include<br />

a high revision rate for débridement (33% for trochlear lesions). A<br />

bipolar lesion such as a “kissing lesion” is a relative contraindication<br />

to cartilage restoration and would likely benefit from PF arthroplasty.<br />

Notably, the complication rate for ACI, including the need for<br />

reoperation, has dramatically decreased as a result of the off-label<br />

usage of a synthetic collagen I/III membrane as an alternative to<br />

periosteum. AMZ is the most commonly used realignment procedure<br />

in the treatment of PF cartilage defects. The patients who are most<br />

likely to benefit from AMZ are those with (1) symptomatic lesions,<br />

(2) patellar maltracking, and (3) lesions that can be unloaded onto<br />

healthy cartilage through tibial tubercle osteotomy. Numerous<br />

authors have recommended AMZ, either alone or in association<br />

with a cartilage restoration procedure, if these three conditions<br />

are met. Prior to osteotomy, surgeons should document patellar<br />

maltracking via abnormal TT-TG values whenever possible, as well as<br />

healthy medial trochlear cartilage and, if possible, central trochlear<br />

and patellar cartilage as well. Symptoms must also be attributed to<br />

maltracking, otherwise the interventions upon the resulting articular<br />

cartilage lesions will fail to improve the patient’s outcome. In addition,<br />

while AMZ may work well for patients with articular cartilage defects<br />

and early-stage degeneration, advanced osteoarthritis may signal<br />

that this procedure is less likely to succeed, primarily because of the<br />

lack of healthy cartilage onto which the diseased cartilage can be<br />

unloaded. These patients may be better served by PF arthroplasty or<br />

total knee arthroplasty, depending on the condition of the tibiofemoral<br />

cartilage. The AMZ procedure itself can be customized to unload the<br />

diseased cartilage by changing the slope of the osteotomy to provide<br />

more anteriorization in patients with central or proximal lesions, or<br />

more medialization in patients with lateral lesions. If chondral lesions<br />

are proximal and medial, AMZ alone is contraindicated, and may<br />

worsen symptoms because the forces would be transferred to this<br />

area. Benefits of AMZ must be weighed against the additional risks<br />

of infection, symptomatic hardware, wound complications, nonunion<br />

(especially in obese patients, diabetic patients, and smokers), tibial<br />

fracture, compartment syndrome, and deep vein thrombosis. Removal<br />

of hardware may be required in up to 50% of patients. Several series<br />

have reported tibial fractures with AMZ, recommending extended<br />

weight-bearing limitations.<br />

References:<br />

Aroen A, Loken S, Heir S, et al. Articular cartilage lesions in 993<br />

consecutive knee arthroscopies. Am J Sports Med. 2004;32:211-215.<br />

Curl WW, Krome J, Gordon ES, et al. <strong>Cartilage</strong> injuries: a review of 31,516<br />

knee arthroscopies. Arthroscopy. 1997;13:456-460. Hjelle K, Solheim E,<br />

Strand T, et al. Articular cartilage defects in 1,000 knee arthroscopies.<br />

Arthroscopy. 2002;18:730-734<br />

Kaplan LD, Schurhoff MR, Selesnick H, et al. Magnetic resonance<br />

imaging of the knee in asymptomatic professional basketball players.<br />

Arthroscopy. 2005;21:557-561.<br />

Walczak BE, McCulloch PC, Kang RW, et al. Abnormal finding on<br />

magnetic resonance imaging in asymptomatic NBA players. J Knee Surg.<br />

2008;21:27-33.<br />

Nomura E, Inoue M, Kurimura M: Chondral and osteochondral injuries<br />

associated with acute patellar dislocation. Arthroscopy 2003;19(7):717-<br />

721.<br />

Gallo RA, Feeley BT: <strong>Cartilage</strong> defects of the femoral trochlea. Knee Surg<br />

Sports Traumatol Arthrosc 2009;17(11):1316-1325.<br />

Pascual-Garrido C, Slabaugh MA, L’Heureux DR, Friel NA, Cole BJ:<br />

Recommendations and treatment outcomes for patellofemoral articular<br />

cartilage defects with autologous chondrocyte implantation: Prospective<br />

evaluation at average 4-year follow-up. Am J Sports Med 2009;37(suppl<br />

1):33S-41S.<br />

Kreuz PC, Steinwachs MR, Erggelet C, et al: Results after microfracture<br />

of full-thickness chondral defects in different compartments in the knee.<br />

Osteoarthritis <strong>Cartilage</strong>2006;14(11):1119-1125.<br />

Jamali AA, Emmerson BC, Chung C, Convery FR, Bugbee WD: Fresh<br />

osteochondral allografts: Results in the patellofemoral joint. Clin Orthop<br />

Relat Res 2005;437:176-185.<br />

Farr J: Autologous chondrocyte implantation improves patellofemoral<br />

cartilage treatment outcomes. Clin Orthop Relat Res 2007;463:187-194.<br />

Mandelbaum B, Browne JE, Fu F, et al: Treatment outcomes of autologous<br />

chondrocyte implantation for full-thickness articular cartilage defects of<br />

the trochlea. Am J Sports Med2007;35(6):915-921.<br />

Acknowledgments:<br />

Geoffrey S. Van Thiel, MD/MBA Andrew Lee, MS<br />

3.1.1<br />

Basic concepts and potential application of tissue/organ printing<br />

D. Hutmacher<br />

Brisbane/Australia<br />

Introduction: The fundamental concept underlying tissue<br />

engineering is to combine a scaffold or matrix, with living cells,<br />

and/or biologically active molecules to form a tissue engineering<br />

construct (TEC) to promote the repair and/or regeneration of<br />

tissues. The scaffold (a cellular solid support structure comprising<br />

an interconnected pore network) or matrix (often a hydrogel in which<br />

cells can be encapsulated) is expected to perform various functions,<br />

including the support of cell colonization, migration, growth and<br />

differentiation. Further, for their design physicochemical properties,<br />

morphology and degradation kinetics need to be considered. External<br />

size and shape of the construct are of importance, particularly if<br />

it is customized for an individual patient [1]. Besides the physical<br />

properties of a scaffold or matrix material (e.g. stiffness, strength,<br />

surface chemistry, degradation kinetics), the micro-architecture of<br />

the constructs is of great importance for the tissue formation process<br />

[2]. In recent years, a number of automated fabrication methods have<br />

been employed to create scaffolds with well-defined architectures [3,<br />

4]. These have been classified as rapid prototyping (RP) technologies,<br />

solid freeform fabrication (SFF) techniques, or according to the latest<br />

ASTM standards, additive manufacturing (AM) techniques [5]. With<br />

AM techniques, scaffolds with precise geometries can be prepared<br />

[6], using computer-aided design combined with medical imaging<br />

techniques to make anatomically shaped implants [7]. Together with<br />

the development of biomaterials suitable for these techniques, the<br />

automated fabrication of scaffolds with tunable, reproducible and<br />

mathematically predictable physical properties has become a fastdeveloping<br />

research area.<br />

Content: The last few years have seen an upturn in economic activity<br />

and successful application of newly developed tissue engineering<br />

products, which for the largest part has resulted from identification<br />

of products that are translatable from bench to bedside with<br />

available technology and under existing regulatory guidelines [8].<br />

Cell-free scaffolds have shown clinical success, e.g. for bone (Fig. 1),<br />

osteochondral tissue repair, cartilage and skin [9]. Also, strategies to<br />

create new vasculature - a critical aspect of tissue engineering - are<br />

being developed by making use of the body’s self-healing capacity<br />

[10].<br />

Nevertheless, cell-based therapeutics have largely failed from both<br />

a clinical and financial perspective [12, 13]. The developed tissue<br />

engineering products were not necessarily inferior to previous<br />

alternatives, but the efficacy and efficiency were not sufficient<br />

to justify the associated increases in costs [14,15]. Manual cell<br />

seeding and culturing of pre-fabricated scaffolds is time-consuming,<br />

user-dependent, semi-efficient and, therefore, economically<br />

and logistically not feasible to achieve clinical application at an<br />

economical scale [16, 17]. Particular shortcomings of the current<br />

tissue engineering paradigm involving cell seeding of pre-fabricated<br />

scaffolds are the inabilities to: a)mimic the cellular organization<br />

of natural tissues b) upscale to (economically feasible) clinical<br />

application and c) address the issue of vascularization.<br />

The use of additive tissue manufacturing addresses these points<br />

by the incorporation of cells into a computer-controlled fabrication<br />

process, thus creating living cell/material constructs rather than


cell-free scaffolds. The fundamental premise of computer-controlled<br />

tissue fabrication is that tissue formation can be directed by the<br />

spatial placement of cells themselves (and their extracellular matrix),<br />

rather than by the spatial architecture of a solid support structure<br />

alone. Although still at an early stage of concept development and<br />

proof-of-principle experiments, it appears that endeavors following<br />

this approach are the most promising to deliver clinical solutions<br />

on the longer term where cell-free approaches cannot. Automated<br />

tissue assembly opens up a route to scalable and reproducible mass<br />

production of tissue precursors. Furthermore, implementing good<br />

manufacturing practices (GMP), quality control and legislation are<br />

facilitated by the use of automated processes.<br />

Additive manufacturing will enable the production of cell-containing<br />

constructs in a computer-controlled manner, thereby bypassing<br />

costly and poorly controlled manual cell seeding. Although big<br />

steps have been taken since the origins early in the past decade,<br />

the technology is still in its infancy. It is now critical to address<br />

key issues in biomaterials development (matching degradation<br />

to cellular production and providing adequate mechanical<br />

properties, while achieving rheological properties required for the<br />

manufacturing process), construct design (including vascularization<br />

of the construct), and system integration (inclusion of multiple cells,<br />

materials and manufacturing processes in a sterile and controlled<br />

environment). It is also important to pursue the development and<br />

commercialization of constructs in a manner that is acceptable to<br />

regulatory agencies, such as the Food and Drug Administration,<br />

where they will more than likely be classed as “combination<br />

products”, to efficiently translate research outcomes to clinical<br />

benefits. With the joint effort of researchers combining chemistry,<br />

mechanical engineering, information technology and cell biology,<br />

AM techniques can evolve into a technology platform that allows<br />

users to create tissue-engineered constructs with economics of<br />

scale in the years to come.<br />

References:<br />

1. Langer R, Vacanti JP. Tissue Engineering. Science 1993;260:920-6.<br />

2. Malda J, Woodfield TBF, van der Vloodt F, Wilson C, Martens DE,<br />

Tramper J, et al. The effect of PEGT/PBT scaffold architecture on the<br />

composition of tissue engineered cartilage. Biomaterials 2005;26:63-<br />

72.<br />

Acknowledgments:<br />

Australian Research Council (ARC) for funding.<br />

3.1.2<br />

Potential of natural and synthetic biomaterials for bioprinting<br />

F.P.W. Melchels 1 , W.J.A. Dhert 1 , D. Hutmacher 2 , J. Malda 1<br />

1 Utrecht/Netherlands, 2 Brisbane/Australia<br />

Introduction: Additive manufacturing techniques offer the potential<br />

to fabricate well-defined structures, combining computer-aided<br />

design and/or medical imaging with computer-controlled fabrication<br />

techniques to make anatomically shaped implants. The level of control<br />

offered by these computer-controlled technologies to design and<br />

fabricate tissues will accelerate our understanding of the governing<br />

factors of tissue formation and function [1]. Moreover, it will provide<br />

a valuable tool to study the effect of anatomy on graft performance.<br />

Bioprinting refers to the use of additive manufacturing techniques<br />

(particularly computer-controlled extrusion or dispensing) with<br />

incorporation of biology in the form of peptides, growth factors or<br />

even living cells. A major bottleneck in the development of bioprinting<br />

techniques is the limited availability of suitable biomaterials, which<br />

will be discussed in this paper.<br />

Content: Despite the promising results that have come out of tissue<br />

engineering research in the past 2 decades, the numbers of tissue<br />

engineering products and patients treated are still very low in light of<br />

the volume of funding in this research area. Obviously the complexity<br />

of engineering tissues is a main contributor to this fact, as tissue<br />

engineering is a knowledge-intensive area in which a multitude of<br />

expertises, materials and processes will have to be combined<br />

synergistically to come to the end product: a medical treatment.<br />

Traditionally, the preparation of a tissue engineered construct (TEC)<br />

Extended Abstracts 113<br />

involves many manual handlings, such as cell isolation, cell<br />

expansion, scaffold fabrication, cell seeding, cell culturing and<br />

implantation. This way of operation might work well in a research<br />

setting but is hard to translate to a clinical setting for the following<br />

reasons: · the logistics and resources required would be nearimpossible<br />

to realise · the labour-associated costs would be<br />

prohibitive · due to the many manual handlings, reproducible results<br />

are far from guaranteed · good manufacture practice (GMP) standards<br />

would be hard to meet in terms of standardisation and quality control<br />

For these reasons, automation of tissue engineering processes will<br />

be indispensible to raise its practical applicability to the level<br />

required for the clinic [2]. Over the last decade, a general trend can<br />

be seen towards higher levels of automation of several steps in the<br />

process of preparing TECs, generally with improved outcomes. The<br />

development of AM technologies and suitable biomaterials has<br />

created the ability to fabricate scaffolds with well-defined internal<br />

pore architectures based on medical imaging computer-aided design<br />

(CAD) [3]. Compared to scaffolds prepared by conventional<br />

techniques such as porogen leaching and gas foaming, AM-fabricated<br />

scaffolds exhibit improved mechanical properties, improved pore<br />

accessibility and (hence) better cell seeding and nutrient transport.<br />

Furthermore, the automation of the cell seeding and culturing phase<br />

using bioreactors has improved tissue uniformity [4]. Still, the current<br />

tissue engineering paradigm involving cell seeding of pre-fabricated<br />

scaffolds has some inherent shortcomings. For example, the complex<br />

anisotropic cellular organisation found in native tissues can not be<br />

mimicked. Furthermore, the very important issue of vascularisation<br />

(for most tissues other than cartilage at least) is insufficiently<br />

addressed. In bioprinting, the scaffold fabrication and cell seeding<br />

stages are combined and fully automated by concurrent deposition<br />

of cells and biomaterials [1]. This allows for the highly automated<br />

preparation of TECs in a scalable and reproducible manner, with<br />

designed construct architecture and computer-controlled placement<br />

of cells. Since cells require a moist environment, hydrogels are the<br />

most suited class of biomaterials to contain the cells in the bioprinting<br />

process. Hydrogels are networks of water-soluble polymers that are<br />

crosslinked through either physical (reversible) or chemical<br />

(irreversible) crosslinks. As a result, these highly hydrated<br />

environments (typically containing 75-99 % water) allow for diffusion<br />

of nutrients and metabolites, whilst immobilising the cells [5]. To be<br />

used in conjunction with cells in tissue engineering applications,<br />

hydrogels have to meet be non-toxic and allow cells in the gel to<br />

perform all their natural functions, which may include attachment,<br />

proliferation, (re)differentiation and production of ECM components.<br />

Finally, degradability of the hydrogel is desired to allow for cell<br />

motility and tissue remodelling. To this end, soft, loosely crosslinked<br />

gels with low polymer concentrations are often the most appropriate.<br />

Printing of well-defined structures poses completely different -if not<br />

opposite- requirements on hydrogel properties. The viscosity needs<br />

to be sufficiently high to enable drawing of thin filaments of material<br />

and to prevent cells from settling in the precursor suspension.<br />

Subsequently, a relatively quick gelation is instrumental to retain<br />

the shape of the fabricated structure. A last requirement is adequate<br />

mechanical properties to preserve shape during culture, and/or to<br />

withstand loads after implantation. The mentioned properties are<br />

optimal at high polymer concentrations and high crosslink densities,<br />

which are conditions that are counter-productive for cell survival and<br />

functioning. Therefore, one of the largest challenges is to establish<br />

the ‘bioprinting window’, i.e. to find the conditions which allow for<br />

printing as well as cell encapsulation [6]. Moreover, smart ways will<br />

have to be found to increase viscosity, shape retainment and<br />

mechanical properties, without necessarily increasing polymer<br />

concentration or crosslink density. For this, one has to make use of<br />

rheological phenomena such as yield stres and shear-thinning, and<br />

to design superstrong hydrogels with particular network topologies<br />

[7]. Hydrogels can be of either natural or synthetic origin. Naturally<br />

derived gels are generally good cell support materials, but intrinsically<br />

suffer from batch-to-batch variation, limited tunability and the<br />

possibility of disease transfer. Synthetic hydrogels do not bear these<br />

disadvantages, but often lack biofunctionality. Naturally derived<br />

hydrogels that have been employed for bioprinting include proteins,<br />

polysaccharides and glycosaminoglycans. Collagen is a protein<br />

derived from animal skin or bones that forms a good cell substrate<br />

as it presents binding peptide motifs and allows for cell-mediated<br />

tissue remodelling through enzymatic degradation. It has been used<br />

quite successfully for bioprinting endothelial cells [8]. Due to the low<br />

concentration (0.3 %) and low crosslink density, large structures<br />

could not be built; however the cells were motile and could proliferate<br />

fast (30 % increase in 24 hrs), making collagen a good candidate for<br />

cell delivery in bioprinting. Gelatine is denatured collagen bearing<br />

similar properties but with increased water solubility and lower<br />

antigenic risk than collagen due to its denatured state. It has been<br />

printed at 20 % into large structures, laden with hepatocytes that<br />

remained viable for as long as 3 months after aldehyde crosslinking


114<br />

Extended Abstracts<br />

[9]. Agarose and alginate are polysaccharides extracted from the cell<br />

walls of algae and have both been used for cell immobilisation and<br />

bioprinting. Bioprinting of warm agarose solutions is feasible (at 5<br />

%), although the gelation is relatively slow which causes some<br />

broadening and sagging of the printed filaments [10]. Alginate forms<br />

a gel when divalent cations (usually Ca2+) are mixed in, by<br />

electrostatic crosslinking. Therefore, sodium alginate solutions can<br />

be dispensed into a bath of CaCl2 solution resulting in the formation<br />

of gel strands [11]. Alternatively, the sodium alginate and CaCl2<br />

solution can be pre-mixed and printed within the time frame of<br />

gelation [12]. Dextran is another polysaccharide that has been<br />

applied in bioprinting. It has no intrinsic gelation property, so it can<br />

be modified with methacrylate groups to allow photo-initiated<br />

crosslinking to retain the printed shape. Furthermore, to obtain an<br />

appropriate viscosity for printing it has been mixed with more viscous<br />

components such as hyaluronan [13]. Hyaluronan (synonymous for<br />

hyaluronic acid or HA) is a glycosaminoglycan which has a relatively<br />

high occurrence in articular cartilage, and therefore seems a good<br />

candidate for chondrocyte encapsulation and bioprinting. Besides<br />

as a mixture with dextran, HA has been printed in conjunction with<br />

gelatine (both components methacrylate-functionalised for UVinitiated<br />

crosslinking) [14] and with star-shaped PEG-tetraacrylate<br />

[15]. In neither case evidence was given of high-resolution printed<br />

structures, however the cells (hepatoma cells, intestine epithelial<br />

cells and fibroblasts) did proliferate when encapsulated in the gels,<br />

indicating the suitability of HA as an environment for tissue<br />

development. Compared to the examples of bioprinting of naturally<br />

derived hydrogels above, far less examples of bioprinting of synthetic<br />

hydrogels exist. The mostly used polymer is sold under the brand<br />

name Lutrol F127 and is used in the pharmaceutical industry. It is an<br />

amphiphilic block copolymer consisting of two hydrophilic PEG<br />

blocks and one slightly hydrophobic poly(propylene oxide) (PPO)<br />

block. In water it is fluid at low temperatures and gels at around 30<br />

°C as the PPO blocks collapse and form physical crosslinks. Its<br />

rheological properties allow printing of up to 10 layers of well-defined<br />

gel strands [8, 11]. Initial cell viability after printing is good, but when<br />

incubated in medium at 37 °C the physical hydrogel slowly dissolves<br />

and most of the cells die within 3 days. Modification with alaninemethacrylate<br />

endgroups to allow for photo-initiated crosslinking<br />

enables at least 3 weeks culture with retainment of shape and<br />

reasonable cell viability (60 % 3 days post-printing, unchanged over<br />

up to 3 weeks) [16]. Except for this gel, currently only one other<br />

polymer has been reported specifically designed for bioprinting [17].<br />

It is also a PEG-based polymer with hydrophobic blocks that induce<br />

thermal gelation (hydroxypropylmethacrylamide-lactate in this case)<br />

and has methacrylate end-groups for photo-initiated crosslinking as<br />

well. Short-term viability of encapsulated cells was demonstrated,<br />

but longer-term cultures with assessment of differentiation and<br />

production of ECM components will have to prove the suitability of<br />

these gels for bioprinting of viable TECs. In summary, over the past<br />

few years researchers have made a start in identifying hydrogels that<br />

may be suitable for bioprinting. Computer-designed hydrogel<br />

structures with encapsulated cells have been prepared with<br />

reasonable accuracy. Over the next decade we can expect the<br />

development of biomaterials specifically engineered for bioprinting,<br />

and a considerable increase in the quality of printed constructs.<br />

Furthermore, the functionality of printed cell-laden constructs in<br />

terms of tissue development and regeneration capacity will have to<br />

be critically assessed.<br />

References:<br />

1. Melchels, F.P.W., et al., Progress in Polymer Science, 2012.<br />

doi:10.1016/j.progpolymsci.2011.11.007.<br />

2. Archer, R. and D.J. Williams, Nature Biotechnology, 2005. 23: p.<br />

1353-1355.<br />

3. Peltola, S.M., et al., Annals of Medicine, 2008. 40: p. 268-280.<br />

4. Martin, I., D. Wendt, and M. Heberer, Trends in Biotechnology,<br />

2004. 22: p. 80-86.<br />

5. Nicodemus, G.D. and S.J. Bryant, Tissue Engineering Part<br />

B-Reviews, 2008. 14: p. 149-165.<br />

6. Khalil, S. and W. Sun, Journal of Biomechanical Engineering-<br />

Transactions of the Asme, 2009. 131.<br />

7. Johnson, J.A., et al., Progress in Polymer Science, 2010. 35: p. 332-<br />

337.<br />

8. Smith, C.M., et al., Tissue Engineering, 2004. 10: p. 1566-1576.<br />

9. Wang, X.H., et al., Tissue Engineering, 2006. 12: p. 83-90.<br />

10. Fedorovich, N.E., et al., Tissue Engineering Part A, 2008. 14: p.<br />

127-133.<br />

11. Fedorovich, N.E., et al., Tissue Engineering, 2007. 13: p. 1905-<br />

1925.<br />

12. Cohen, D.L., et al., Tissue Engineering, 2006. 12: p. 1325-1335.<br />

13. Pescosolido, L., et al., Biomacromolecules, 2011. 12: p. 1831-<br />

1838.<br />

14. Skardal, A., et al., Tissue Engineering Part A, 2010. 16: p. 2675-<br />

2685.<br />

15. Skardal, A., J. Zhang, and G.D. Prestwich, Biomaterials, 2010. 31:<br />

p. 6173-81.<br />

16. Fedorovich, N.E., et al., Biomacromolecules, 2009. 10: p. 1689-<br />

1696.<br />

17. Censi, R., et al., Advanced Functional Materials, 2011. 21: p. 1833-<br />

1842.<br />

Acknowledgments:<br />

For funding we thank the European Union (Marie Curie <strong>International</strong><br />

Outgoing Fellowship)<br />

3.2.2<br />

In vitro models for development and screening of cartilage repair<br />

strategies<br />

G.J.V.M. Van Osch<br />

Rotterdam/Netherlands<br />

Introduction: Traumatic cartilage defects can be treated with<br />

autologous chondrocyte implantation or microfracture. The results<br />

of these treatments are encouraging but are not equally good for all<br />

patients, do not always last for years and the repair tissue does not<br />

completely resemble the original. So there is room for improvement<br />

of these treatments. For generalized cartilage degeneration,<br />

osteoarthritis, there is no treatment available that can stop, slow or<br />

cure the disease. NSAIDs, cox-2 inhibitors, glucosamine and stem<br />

cells, amongst other treatment options, continue to raise interest<br />

and are under investigation.<br />

Content: In early stages of development of a cartilage repair strategy,<br />

treatments are evaluated with in-vitro models as well as animal<br />

models. In-vitro models are being applied frequently especially in<br />

OA research. There is a broad range of culture models of isolated<br />

chondrocytes or cartilage explants, of animal or human origin. The<br />

fast developments in the field of cartilage tissue engineering is<br />

expected to reveal new models, but these are not yet frequently used<br />

as test models for new treatments. The choice for a certain culture<br />

model usually depends on robustness, costs and experience with<br />

the models. But in spite of the availability of these well characterised<br />

chondrocyte and cartilage culture systems one has to realize that<br />

in the joint cartilage in not the only tissue present. Continuous<br />

interactions exist between the different joint tissues, largely based<br />

on secreted factors. For a good representation of the situation in the<br />

joint, these interactions should be taken into account. This requires<br />

more complex model systems with multiple tissues. The last years<br />

the involvement of subchondral bone in cartilage degeneration and<br />

regeneration became more obvious. Subchondral bone contains<br />

many growth factors that will be released upon damage. Furthermore<br />

the development of a model including cartilage and subchondral<br />

bone allows studying repair strategies of chondral defects as well<br />

as osteochondral defects (de Vries-van Melle et al 2011). Although<br />

it is accepted for long time that synovial inflammation influences<br />

cartilage metabolism in rheumatoid arthritis, its role in cartilage<br />

repair and osteoarthritis was largely neglected till recently. The<br />

effect of synovium of patients with osteoarthritis on cartilage<br />

formation by mesenchymal stem cells was recently demonstrated<br />

(Heldens et al, 2011). We recently demonstrated that cartilage and<br />

synovium secrete different cytokines and that the combination of<br />

cartilage and synovium in culture better represents the situation<br />

in a joint (Beekhuizen et al 2011). With this model we were able to<br />

show that triamcinolone, that is for years being discussed for its


negative effects on cartilage metabolism, has a protective effect<br />

on cartilage in the presence of osteoarthritic synovium. Finally,<br />

joints contain a large amount of intra-articular adipose tissue. More<br />

recently the contribution of intra-articular adipose tissue received<br />

attention (Clockaerts et al 2010). Next to adipocytes, this intraarticular<br />

fat tissue contains immune cells and stem cells. We have<br />

recently shown that the immune cell composition is influenced by<br />

joint pathology (Klein-Wieringa et al 2011; Bastiaansen-Jenniskens<br />

et al 2011). Due to this altered cellular composition, the adipose<br />

tissue secretes different factors into the synovial fluid. These<br />

secreted factors do influence cartilage and synovium metabolism<br />

(Bastiaansen-Jenniskens et al 2011). These in-vitro models that<br />

contain multiple tissues better represent the conditions in a joint<br />

in-vivo. These are, however still models, thus simplifying the real<br />

situation. It is for example not clear if and how the relatively large<br />

cutting edges, which are created during preparation of the tissues,<br />

influence the responses. Whole organ cultures are being used, albeit<br />

infrequently; These used to involve developing embryonic bones of<br />

rodents, but also patellae of small animals (Schalkwijk et al 1985)<br />

and even sesamoid bones of cows (Korver et al 1989) can be cultured.<br />

Furthermore, long-term culturing will definitely have an effect on cell<br />

metabolism, but these effects are not completely clear. Finally, joint<br />

loading is an important condition that is only sometimes taken into<br />

account (Grad et al 2006) since it is difficult to apply physiological<br />

loading regimes in large series. Although animal models will remain<br />

an important tool in development of medications, each of the invitro<br />

models can be useful as first screening for a newly developed<br />

cartilage repair strategy, to investigate mechanisms of action or to<br />

find ways for further improvement of therapy. The use of in-vitro<br />

models can reduce costs and number of animals used and offers the<br />

opportunity to use a fully human system.<br />

References:<br />

Bastiaansen-Jenniskens YM, Clockaerts S, Feijt C, Zuurmond AM,<br />

Stojanovic-Susulic V, Bridts C, de Clerck L, Degroot J, Verhaar JA,<br />

Kloppenburg M, van Osch GJ. Infrapatellar fat pad of patients with<br />

end-stage osteoarthritis inhibits catabolic mediators in cartilage.<br />

Ann Rheum Dis. 2011 Oct 13. [Epub ahead of print]<br />

Beekhuizen M, Bastiaansen-Jenniskens YM, Koevoet W, Saris DB,<br />

Dhert WJ, Creemers LB, van Osch GJ. Osteoarthritic synovial tissue<br />

inhibition of proteoglycan production in human osteoarthritic<br />

knee cartilage: establishment and characterization of a long-term<br />

cartilage-synovium coculture. Arthritis Rheum. 2011 Jul;63(7):1918-<br />

27.<br />

Clockaerts S, Bastiaansen-Jenniskens YM, Runhaar J, Van Osch GJ,<br />

Van Offel JF, Verhaar JA, De Clerck LS, Somville J. The infrapatellar fat<br />

pad should be considered as an active osteoarthritic joint tissue: a<br />

narrative review. Osteoarthritis <strong>Cartilage</strong>. 2010 Jul;18(7):876-82.<br />

De Vries-van Melle ML, Mandl EW, Kops N, Koevoet WJ, Verhaar JA,<br />

van Osch GJ. An Osteochondral Culture Model to Study Mechanisms<br />

Involved in Articular <strong>Cartilage</strong> <strong>Repair</strong>. Tissue Eng Part C Methods.<br />

2011 Oct 18. [Epub ahead of print]<br />

Grad S, Gogolewski S, Alini M, Wimmer MA. Effects of simple and<br />

complex motion patterns on gene expression of chondrocytes<br />

seeded in 3D scaffolds. Tissue Eng. 2006 Nov;12(11):3171-9.<br />

Heldens GT, Blaney Davidson EN, Vitters EL, Schreurs BW, Piek<br />

E, van den Berg WB, van der Kraan PM. Catabolic Factors and<br />

Osteoarthritis-Conditioned Medium Inhibit Chondrogenesis of<br />

Human Mesenchymal Stem Cells. Tissue Eng Part A. 2011 Oct 17.<br />

[Epub ahead of print]<br />

Klein-Wieringa IR, Kloppenburg M, Bastiaansen-Jenniskens YM,<br />

Yusuf E, Kwekkeboom JC, El-Bannoudi H, Nelissen RG, Zuurmond<br />

A, Stojanovic-Susulic V, Van Osch GJ, Toes RE, Ioan-Facsinay A.<br />

The infrapatellar fat pad of patients with osteoarthritis has an<br />

inflammatory phenotype. Ann Rheum Dis. 2011 May;70(5):851-7.<br />

Korver GH, van de Stadt RJ, van Kampen GP, Kiljan E, van der Korst<br />

JK. Bovine sesamoid bones: a culture system for anatomically intact<br />

articular cartilage. In Vitro Cell Dev Biol. 1989 Dec;25(12):1099-106.<br />

Schalkwijk J, van den Berg WB, van de Putte LB, Joosten LA. Hydrogen<br />

peroxide suppresses the proteoglycan synthesis of intact articular<br />

cartilage. J Rheumatol. 1985 Apr;12(2):205-10.<br />

Acknowledgments:<br />

Extended Abstracts 115<br />

The research was financially supported by the Dutch Arthritis<br />

Association, Top Institute Pharma, BioMedical Materials Program,<br />

GAMBA (FP7)<br />

3.3.1<br />

Can meniscus reconstruction prevent or even cure OA?<br />

W. Gersoff<br />

Denver/United States of America<br />

Introduction: see content<br />

Content: Can Meniscus Reconstruction Prevent or Even Cure OA? The<br />

development of knee OA is a multifactorial process. Some factors<br />

are presently not controllable (genetics), while others are potentially<br />

controllable (weight and activity), and others are potentially<br />

modifiable(injury to the knee). In evaluating whether mensical<br />

allograft transplantation can modify the development of osteoarthritis<br />

of the knee several important factors must be considered: the role<br />

of the meniscus; the effects of menisectomy on knee function and<br />

biomechanics; and, the success of meniscal allograft transplantation<br />

as a procedure to restore meniscal function. The meniscus functions<br />

as the key component of the shock absorbency and force distribution<br />

system of the knee joint. In this capacity it acts to decrease the<br />

compressive and loading forces transmitted to the articular cartilage<br />

and subchondral bone therefore protecting them from excessive<br />

loads. The meniscus has an important biomechanical role in the knee<br />

joint contributing to joint stability. The development of osteoarthritis<br />

in the knee joint after menisectomy has been recognized as early<br />

as 1949 in Fairbank’s original work. Subsequent studies have<br />

also demonstrated the high potential for the development of<br />

osteoarthritis in the post-menisectomy knee. As arthroscopy has<br />

developed further studies have shown the varying roles of partial<br />

menisectomy and total meniscetomy in the development of knee<br />

osteoarthritis and therefore the need to maintain as much meniscus<br />

tissue as possible. Improved techniques of meniscal repair have<br />

facilitated the preservation of the meniscus as well.The concept<br />

of functional menisectomy has also been developed to delineate<br />

the importance of regions of the meniscus in regard to increasing<br />

risk of the development of osteoarthritis.The posterior horn areas<br />

of both the medial and lateral meniscus have been shown to bear<br />

the majority of load in the knee. The loss of the posterior horn<br />

will result in a functional menisectomy although there may still be<br />

over 50% of the meniscus tissue remaining.In general the loss of<br />

greater then 50% of the meniscal tissue is considered detrimental<br />

regardless of anatomical area. Meniscal allograft transplantation<br />

was first performed by Milachowski in 1986. Since that time there<br />

has been significant advancements in both surgical techniques and<br />

tissue preservation. During this time there has also been continued<br />

confirmation of the importance of the intact functioning meniscus.<br />

While the first meniscus allograft transplantation procedures<br />

were done as open procedures with formal arthrotomy and often<br />

collateral ligament release , the technique has now evolved to a<br />

minimally invasive procedure performed arthroscopically assisted..<br />

Instrumentation has been developed that allows for consistent<br />

placement of the allograft tissue based on the known landmarks of<br />

the anterior and posterior horn attachments of the meniscus.The<br />

techniques involve either the use of a bone bridge, double bone plugs,<br />

or all soft tissue. The bone bridge techniques either incorporate a<br />

slot or trough. Regardless of the technique performed, meticulous<br />

detail must be used to maintain the anatomical positioning of the<br />

meniscus. Meniscal repair fixation devices have also been developed<br />

that allows for all inside fixation of the majority of the meniscal<br />

segments. Some of these devices however are not well designed<br />

for the attachment of meniscus to synovium but rather meniscus<br />

to meniscus. Advances have also been made in tissue processing,<br />

preservation, sizing and availability. Early issues associated with<br />

tissue shrinkage and friability secondary to excessive radiation and<br />

freezing techniques have been eliminated. Meniscal allograft tissue<br />

is considered to be immunologically privileged. Therefore the risk of<br />

tissue rejection is minimal, although a subclinical immune response<br />

may occur.The post-operative rehabilitation has also improved as a<br />

better understanding of the science of the meniscal allograft healing<br />

has taken place. The patient will undergo minimal immobilization<br />

and start earlier weight bearing. The importance of knee ligament<br />

stability, femoral tibial alignment, and articular cartilage integrity<br />

in association with meniscal pathology has also been given greater<br />

consideration. Procedures to address any of these deficiencies are<br />

commonly combined with meniscal allograft transplantation.The<br />

patient receiving a meniscal allograft transplantation in 2012 is most


116<br />

Extended Abstracts<br />

likely receiving an overall improved surgical procedure with higher<br />

success rates and better long-term outcomes than 26 years ago.<br />

With these developments in consideration is it possible to critically<br />

look at the results of meniscal allograft transplantation? A recent<br />

meta-analysis published by ElAttar et al reviewed publications<br />

in the English speaking literature over the past 26 years. This<br />

would therefore include publications before,during and after the<br />

previously described developments. This publication fouty-four<br />

trials representing 1136 grafts and 1068 patients. Of these 678<br />

were medial and 458 were lateral with a mean age of 34,8 years.<br />

The authors concluded that meniscal allograft transplantation<br />

was a safe and reliable procedure with a consistent satisfactory<br />

outcome. However,does symptom improvement directly correlate<br />

with the prevention of osteoarthritis ? This is a extremely challenging<br />

question and there are no studies that could document this for an<br />

isolated meniscal allograft transplantation. Indeed, this study could<br />

be extremely difficult to design and implement as it would have to<br />

be designed to take in multiple factors that could contribute to the<br />

development of knee osteoarthritis. The study would also require<br />

a long period of follow-up to truly determine the prevention or<br />

development of knee ostaoarthritis. The post- menisecetomy knee<br />

has a 14-fold increase in the development of osteoarthritis as<br />

documented by radiograph. This is secondary to increased loading<br />

in the affected compartment of the knee and altered biomechanics<br />

of the knee. Mensical allograft transplantation has been shown<br />

to decrease these compressive forces in animal studies. Meniscal<br />

allograft transplantation has also been shown to be be a safe,reliable<br />

and effective operation in the clinical setting. Therefore, based on<br />

the information available in 2012 it is reasonable to assume that<br />

mensical allograft transplantation at the very least functions to<br />

alter the course of degeneration in the knee that would otherwise<br />

be destined for the development of osteoarthritis. This ultimately<br />

may not prevent osteoarthritis development but may prolong its<br />

development. The future development of improved techniques<br />

and tissue healing enhancement factors will certainly continue<br />

to improve the outcomes of the procedure. The development of<br />

synthetic meniscal replacement devices for either partial or total<br />

menisectomies will continue to revolutionize the ability to preserve<br />

mensical integrity and function in the knee.<br />

Can Meniscus Reconstruction Prevent or Even Cure OA? The<br />

development of knee OA is a multifactorial process. Some factors<br />

are presently not controllable (genetics), while others are potentially<br />

controllable (weight and activity), and others are potentially<br />

modifiable(injury to the knee). In evaluating whether mensical<br />

allograft transplantation can modify the development of osteoarthritis<br />

of the knee several important factors must be considered: the role<br />

of the meniscus; the effects of menisectomy on knee function and<br />

biomechanics; and, the success of meniscal allograft transplantation<br />

as a procedure to restore meniscal function. The meniscus functions<br />

as the key component of the shock absorbency and force distribution<br />

system of the knee joint. In this capacity it acts to decrease the<br />

compressive and loading forces transmitted to the articular cartilage<br />

and subchondral bone therefore protecting them from excessive<br />

loads. The meniscus has an important biomechanical role in the knee<br />

joint contributing to joint stability. The development of osteoarthritis<br />

in the knee joint after menisectomy has been recognized as early<br />

as 1949 in Fairbank’s original work. Subsequent studies have also<br />

demonstrated the high potential for the development of osteoarthritis<br />

in the post-menisectomy knee. As arthroscopy has developed further<br />

studies have shown the varying roles of partial menisectomy and total<br />

meniscetomy in the development of knee osteoarthritis and therefore<br />

the need to maintain as much meniscus tissue as possible. Improved<br />

techniques of meniscal repair have facilitated the preservation<br />

of the meniscus as well.The concept of functional menisectomy<br />

has also been developed to delineate the importance of regions<br />

of the meniscus in regard to increasing risk of the development<br />

of osteoarthritis.The posterior horn areas of both the medial and<br />

lateral meniscus have been shown to bear the majority of load in<br />

the knee. The loss of the posterior horn will result in a functional<br />

menisectomy although there may still be over 50% of the meniscus<br />

tissue remaining.In general the loss of greater then 50% of the<br />

meniscal tissue is considered detrimental regardless of anatomical<br />

area. Meniscal allograft transplantation was first performed by<br />

Milachowski in 1986. Since that time there has been significant<br />

advancements in both surgical techniques and tissue preservation.<br />

During this time there has also been continued confirmation of<br />

the importance of the intact functioning meniscus. While the first<br />

meniscus allograft transplantation procedures were done as open<br />

procedures with formal arthrotomy and often collateral ligament<br />

release , the technique has now evolved to a minimally invasive<br />

procedure performed arthroscopically assisted.. Instrumentation has<br />

been developed that allows for consistent placement of the allograft<br />

tissue based on the known landmarks of the anterior and posterior<br />

horn attachments of the meniscus.The techniques involve either the<br />

use of a bone bridge, double bone plugs, or all soft tissue. The bone<br />

bridge techniques either incorporate a slot or trough. Regardless of<br />

the technique performed, meticulous detail must be used to maintain<br />

the anatomical positioning of the meniscus. Meniscal repair fixation<br />

devices have also been developed that allows for all inside fixation<br />

of the majority of the meniscal segments. Some of these devices<br />

however are not well designed for the attachment of meniscus to<br />

synovium but rather meniscus to meniscus. Advances have also been<br />

made in tissue processing, preservation, sizing and availability. Early<br />

issues associated with tissue shrinkage and friability secondary to<br />

excessive radiation and freezing techniques have been eliminated.<br />

Meniscal allograft tissue is considered to be immunologically<br />

privileged. Therefore the risk of tissue rejection is minimal, although<br />

a subclinical immune response may occur.The post-operative<br />

rehabilitation has also improved as a better understanding of the<br />

science of the meniscal allograft healing has taken place. The patient<br />

will undergo minimal immobilization and start earlier weight bearing.<br />

The importance of knee ligament stability, femoral tibial alignment,<br />

and articular cartilage integrity in association with meniscal<br />

pathology has also been given greater consideration. Procedures<br />

to address any of these deficiencies are commonly combined with<br />

meniscal allograft transplantation.The patient receiving a meniscal<br />

allograft transplantation in 2012 is most likely receiving an overall<br />

improved surgical procedure with higher success rates and better<br />

long-term outcomes than 26 years ago. With these developments in<br />

consideration is it possible to critically look at the results of meniscal<br />

allograft transplantation? A recent meta-analysis published by ElAttar<br />

et al reviewed publications in the English speaking literature over the<br />

past 26 years. This would therefore include publications before,during<br />

and after the previously described developments. This publication<br />

fouty-four trials representing 1136 grafts and 1068 patients. Of these<br />

678 were medial and 458 were lateral with a mean age of 34,8 years.<br />

The authors concluded that meniscal allograft transplantation was a<br />

safe and reliable procedure with a consistent satisfactory outcome.<br />

However,does symptom improvement directly correlate with the<br />

prevention of osteoarthritis ? This is a extremely challenging question<br />

and there are no studies that could document this for an isolated<br />

meniscal allograft transplantation.This study could be extremely<br />

difficult to design and implement as it would have to be designed<br />

to take in multiple factors that could contribute to the development<br />

of knee osteoarthritis. The study would also require a long period of<br />

follow-up to truly determine the prevention or development of knee<br />

ostaoarthritis. The post- menisecetomy knee has a 14-fold increase<br />

in the development of osteoarthritis as documented by radiograph.<br />

This is secondary to increased loading in the affected compartment<br />

of the knee and altered biomechanics of the knee. Mensical allograft<br />

transplantation has been shown to decrease these compressive forces<br />

in animal studies. Meniscal allograft transplantation has also been<br />

shown to be be a safe,reliable and effective operation in the clinical<br />

setting. Based on the information available in 2012 it is reasonable<br />

to assume that mensical allograft transplantation at the very least<br />

functions to alter the course of degeneration in the knee that would<br />

otherwise be destined for the development of osteoarthritis. The<br />

development of synthetic meniscal replacement devices for either<br />

partial or total menisectomies will continue to revolutionize the<br />

ability to preserve mensical integrity and function in the knee.<br />

References:<br />

1. Milachowski KA, Weismeier K, Wirth CJ (1989) Homologous<br />

meniscus transplantation,experimental and clincical results. Int<br />

Orthop 13:1-11<br />

2. ElAttar M, Dhollander A, Verdonk R, Almquist KF, Verdonk P<br />

(2011) Twenty-six years of meniscal allograft transplantation: is it<br />

still experimental? A meta-analysis of 44 trials. Knee Surg Sports<br />

Traumatol Arthrosc 19:147-157.<br />

3. Roos H, Lauren M, Adalberth T, Roos EM, Jonsson K, Lohmander<br />

LS (1998) Knee osteoarthritis after menisectomy: prevalence of<br />

radiographic changes after twenty-one years, compared with<br />

matched controls. Arthritis Rheum 41:697-693.<br />

4. von Lewinski G, Milachowski KA, Weismeier K, Kohn D, Wirth<br />

CJ (2007) Twenty year results of combined meniscal allograft<br />

transplantation, anterior cruciate ligament reconstruction and<br />

advancement of the medial collateral ligament. Knee Surg Sports<br />

Traumatol Arthrosc 15:1072-1082.<br />

3.2.3<br />

Nutraceuticals & Osteoarthritis: Do omega-3 fatty acids,<br />

glucosamine & chondroitin sulphates have chondro-protective<br />

actions?


B. Caterson<br />

Cardiff/United Kingdom<br />

Introduction: See content.<br />

Content: Nutraceuticals such as omega-3 fatty acids, glucosamine<br />

and chondroitin sulphates (either separately or in combination) are<br />

widely used by people suffering a wide variety of musculoskeletal<br />

tissue diseases such as osteoarthritis (OA) that has arisen from<br />

a multiplicity of potential causes (e.g. genetic predispositions,<br />

obesity, sports injuries, trauma and ageing). The efficacy of using<br />

nutraceuticals to alleviate pain and OA disease progression has been<br />

claimed for many years without there being unequivocal biological,<br />

biochemical and/or medical evidence explaining their beneficial<br />

mechanisms of action. In this presentation I will review some of the<br />

recent research both supporting and refuting their claims of chondroprotection<br />

in the pathogenesis of OA. The beneficial actions of dietary<br />

cod liver oil (enriched in Omega-3 fatty acids) on alleviating arthritis<br />

were first described in the English scientific literature in the early<br />

1700’s. In more recent years (i.e. late 1980’s <strong>–</strong> present) there have<br />

now been numerous publications providing evidence for some of the<br />

potential targets underlying the mechanism(s) of action of omega-3<br />

fatty acid dietary supplementation. In general, these ‘recent’ basic<br />

biomedical science studies have suggested that omega-3 fatty acid<br />

metabolism acts by reducing the levels of prostaglandin-mediated<br />

inflammation as well as down-regulating the degradative actions of<br />

aggrecanases (ADAMTS-4 & -5) and other matrix metalloproteinases<br />

that destroy the articular cartilage that leads to joint space<br />

narrowing in later stage OA. A variety of clinical trials and metaanalyses<br />

of these trials have been conducted over the past 30 <strong>–</strong> 40<br />

years; however, none of these have provided indisputable medical<br />

data and evidence supporting the claims of chondro-protection and/<br />

or slowing disease progression. The clinical trial results have been<br />

hampered by the heterogeneous causes and nature of human OA<br />

and the meta-analyses of the trial results have been influenced by<br />

both the heterogeneous nature of human OA and the quality of<br />

some of the papers selected to generate the meta-analysis data.<br />

However, a recent study my Knott et al [Osteoarthritis & <strong>Cartilage</strong><br />

(2011) 19: 1150 <strong>–</strong> 1157] using a homogenous (natural genetic<br />

predisposition) model of knee OA have published results definitively<br />

demonstrating the beneficial effects of omega-3 fatty acid dietary<br />

supplementation on both cartilage and bone metabolism. The effects<br />

of dietary supplementation with Glucosamine on the pathogenesis of<br />

osteoarthritis were initially published in the late 1970s & early 1980s<br />

with increasing research occurring in the 1990s and early 21st century.<br />

Similar to the studies performed on the beneficial effects on omega-3<br />

fatty acids there have been a number of in vitro biochemical studies<br />

showing the beneficial effects of glucosamine on decreasing the<br />

expression and activity of cartilage matrix degrading enzymes (e.g.<br />

the aggrecanases); however, large concentrations of glucosamine<br />

were needed to produce these beneficial effects in vitro and these<br />

results thus explaining the large doses (> 1 gram per day) needed<br />

for treatment of human OA sufferers. In some recent studies from<br />

our laboratory [McGuigan et al (2008) J. Med. Chem. 51: 5807 <strong>–</strong> 5812]<br />

we have chemically modified glucosamine to make it more lipophilic;<br />

this biochemical change allowing significantly lower doses of this<br />

‘modified-glucosamine to enter the cell (where it is converted back<br />

to intracellular glucosamine) this delivery process thereby reducing<br />

the expression and activity of the aggrecanases that are responsible<br />

for cartilage degradation in OA. Recent studies from the van Osch<br />

lab [Rozendaal RM et al (2009) Osteoarthritis & <strong>Cartilage</strong> 17: 427<br />

<strong>–</strong> 432] have demonstrated the beneficial effects of glucosamine<br />

dietary supplementation, these effects occurring in both anabolic<br />

and catabolic aspects of synovial joint tissue metabolism. Similarly<br />

to the omega-3 fatty acid studies, the potential chondro-protective<br />

effects of dietary glucosamine supplementation have been tested<br />

in many clinical trials and subsequent meta-analyses. However,<br />

unequivocal conclusions have not been possible because of the<br />

heterogeneous nature of human OA as well as quality of some of<br />

the published papers selected for the meta-analyses. The beneficial<br />

effects in healthy living with dietary supplementation with different<br />

sources of chondroitin sulphates go way back in time and history<br />

(e.g. Asian shark fin soup). However, unlike omega-3 fatty acids and<br />

glucosamine, there have been very few definitive in vitro or in vivo<br />

basic science studies performed that can directly elucidate the effects<br />

(if any) chondroitin sulphates have on cartilage metabolism and/or<br />

it benefits in the treatment of OA. One of the biggest complications<br />

with understanding the effects of dietary supplementation with<br />

‘chondroitin sulphate’ is that there are numerous different structural<br />

types and sources of chondroitin sulphate and its epimerised partner<br />

dermatan sulphate; e.g. a penta-saccharide unit of a chondroitin/<br />

dermatan sulphate (CS/DS) glycosaminoglycan (GAG) chain has a<br />

possible 1008 different combinatorial structures [Caterson B (2012)<br />

Int. J. Exp. Path. 93: 1 <strong>–</strong> 10]. Many of these very specific and different<br />

Extended Abstracts 117<br />

sulphation motifs in CS/DS GAG oligosaccharides are specifically<br />

expressed in “stem/progenitor cell niches” of most tissues in<br />

many animal species [i.e. chicken <strong>–</strong> humans; Caterson 2012]. These<br />

oligosaccharide motifs are involved in binding growth factors,<br />

morphogens and chemokines and in tissue and organ development<br />

the establishment of morphogen & chemokine gradients. At present,<br />

we believe that any beneficial effects of dietary chondroitin sulphate<br />

ingestion may be due to systemic effects on many different tissues<br />

depending on where tissue injury and subsequent regeneration and<br />

repair is needed. Many human OA patient clinical trials have also<br />

attempted to determine the efficacy of chondroitin sulphates alone<br />

or in combination with other popular nutraceuticals. However, these<br />

trials and subsequent meta-analyses have had the same problems<br />

as those for omega-3 fatty acids and glucosamine described above.<br />

In summary, in my opinion, there is some reasonable to very good<br />

evidence to support the use of nutraceutical supplements to slow<br />

down the progression of OA in humans [for recent Review see:<br />

Jerosch J (2011) Int. J. Rheumatol. 2011:969012. E-pub 2011 Aug 2].<br />

However, in the future, researchers may probably benefit from using<br />

homogeneous models of OA to better decipher the biochemical and<br />

physiological mechanisms underpinning the onset of osteoarthritis<br />

in the heterogeneous human population. In addition, I believe that it<br />

would be worthwhile for researchers involved in tissue engineering/<br />

regeneration technologies to consider the potential benefits of<br />

post-operatively adding nutraceutical supplementation to their<br />

procedures investigating their potential benefits in expediting the<br />

outcomes of autologous cartilage implantation/transplantation<br />

technologies.<br />

3.3.2<br />

Clinical options for meniscus reconstruction and regeneration<br />

R. Jakob<br />

Fribourg/Switzerland<br />

Introduction: Thoughts regarding reconstruction, restoration and<br />

regeneration of meniscal volume, size and function after traumatic<br />

disruption of meniscal tissue<br />

Basically, after diagnosing a meniscal tear, fresh or old, you may<br />

decide to:<br />

1- Leave it alone<br />

2- Remove it, partially or totally<br />

3- Suture it (+/- unloading osteotomy in case of compartmental<br />

overload!)<br />

4- Replace it totally by allogenous tissue or partially by artificial<br />

material<br />

In the following we shall concentrate on the means to maintain<br />

meniscal tissue after traumatic lesions.<br />

Content: Meniscal tissue is made of fibrocartilage which is similar<br />

in vulnerability as hyaline articular cartilage. Malalignment,<br />

associated cartilage injuries and ligament instability are definite co-<br />

morbidities. Clinical and radiographic and patient-related outcome<br />

data has established a direct relation of loss of meniscal tissue to<br />

impairment of all these parameters mentioned (Englund et al; Lee et<br />

al). Baratz et al showed an increase in contact pressures of 75% and<br />

an overall increase of 235% in peak-contact pressures after subtotal<br />

meniscectomy.<br />

Consensus exists that in younger patients chances for reconstruction<br />

and healing of meniscal tears are higher than in older patients.<br />

However, ageing and degeneration of the meniscus starts already<br />

at 35 years. Therefore, following a similar principle as in a major<br />

fresh unipolar articular surface lesion or in chronic bipolar<br />

unicompartmental damage you may, in case of definite overload,<br />

decide for an unloading OT. This may as well be indicated in a<br />

difficult meniscal reconstruction which, in our opinion and as has<br />

been observed in the past in the clinical case, is easier than with<br />

marked overload of the treated compartment.


118<br />

Extended Abstracts<br />

Meniscal suture techniques are divided in “inside-out”, “outsidein”<br />

or “all-inside” techniques. While it is so far unsure if the three<br />

methods offer all the same mechanical stability it seems obvious<br />

that the speed of the procedure (surgical time) is in favor of the<br />

all-inside technique but its disadvantage is the higher price. 500 US<br />

$ for two sutures using the modern devices versus 30 US $ using<br />

a couple of “outside-in” sutures! Nerve entrapments or vascular<br />

injuries are the risk and disadvantage of the “outside-in” and<br />

“inside-out” technique. But also the “all-inside” techniques are<br />

not without hazard. Anchors may displace or damage vessels and<br />

nerves if too deeply inserted. Shorter surgical time may weigh a lot<br />

but without any doubt the elegant surgeon may do a solid refixation<br />

using the “outside-in” technique in 25 minutes for 10% of the cost<br />

of a few of the modern anchor systems. As long as the tear site is<br />

more peripherally and not too posteriorly located this may be valid.<br />

Some critical tears are very posteriorly located starting right at the<br />

posterior root so that only an “all-inside” technique using one of the<br />

modern anchor or suture systems is capable, without too high a risk<br />

for neurovascular structures, to lead to a mechanically successful<br />

and solid fixation. Indication for suture is critical and it may need a<br />

sound ethical judgment since meniscal suture associated with an<br />

ACL tear will most likely not be “extra” reimbursed in case the ACL<br />

is reconstructed. To escape this dilemma and submit the patient to<br />

a staged procedure with two surgeries within 6 weeks should not be<br />

the first choice but it is sometimes unavoidable but it carries more<br />

and obvious risks with infection or thromboembolic complications.<br />

We fear that this paramedical conflict of today’s hospital practice<br />

may unfortunately lead the surgeon, who stays under pressure of<br />

the administration and keen hospital directors, to remove a torn<br />

meniscus or leave it unfixed, the latter of the two still being better<br />

because of the inherent chance of a spontaneous healing “despite<br />

the surgeon”. The financial pressure in today’s hospital policy is<br />

high and the remuneration systems are not rendering easy the fate<br />

of the poor meniscus.<br />

Although it is well known that the future of a torn and sutured<br />

meniscus to successfully heal is higher with a stable knee one is<br />

nevertheless allowed to question the relevance of which structure<br />

to be salvaged at every cost or price? Is it the meniscus or the ACL?<br />

The answer comes readily when we ask the question which absent or<br />

poorly functioning structure is more “arthrogenic” (OA producing), a<br />

torn or absent meniscus or an absent ACL? This question, although<br />

justified, may remain open. Among others, three main factors play<br />

hereby a role: The type of meniscal tear, medial or lateral (the lateral<br />

being more rapidly followed by articular damage), concomitant<br />

cartilage surface lesions, morphotype and others, maybe not yet<br />

discovered and described, and the type and intensity of activity of<br />

a given patient.<br />

A few situations merit mentioning. Not only the reconstruction<br />

of meniscal “volume” but the restoration of its function with<br />

maintained circumferential fiber tension must remain the target<br />

and the two cannot be separated from each other. This is easily<br />

understandable with the situation of a posterior root avulsion<br />

or a complete posterior horn radial tear (Seung Beom Han et<br />

al.). Both do basically not decrease volume but allow the entire<br />

meniscus consequently to displace in the periphery and lose a lot<br />

of its function. In other words, mere existence and maintenance or<br />

recreation of meniscal volume is not enough, if it is not invited or<br />

allowed to function in the right location and position! As soon as we<br />

observe peripheral subluxation there must be doubt about having<br />

reached the original purpose and goal of ”shape and function”<br />

which cannot be separated. Correct diagnosis and interpretation<br />

of posterior horn avulsion and adequate, if needed transosseous<br />

refixation is therefore mandatory to give such a meniscus a chance<br />

for anatomic healing and maintenance of function.<br />

Lee et al showed that the periphery of the meniscus is more<br />

important for the overall pressure distribution in the compartment<br />

than the central portion which may indicate that patients after<br />

partial meniscectomy still have a nearly normal pressure distribution<br />

in the joint. An isolated partial meniscectomy therefore may not<br />

pose a significant short-term risk for a cartilage repair procedure.<br />

However, long-term data exists linking partial meniscectomies to<br />

the development of OA over a 15-year time span. This data are even<br />

more compelling in conjunction with ligamentous instability.<br />

Special attention needs to be given to white on white tears which<br />

make up to about 30% of all tears (own estimation) and which still get<br />

sacrificed too loosely and frequently by the average arthroscopist in<br />

a 20 minute, well paid procedure. With the justification that healing<br />

is impossible such a tear too easily serves as the learning path for<br />

the second year orthopaedic resident. And that often marks the start<br />

of a “knee history”. We think that this attitude needs to be revised.<br />

Our own experience over the last 10 years has taught us that the<br />

potential for healing of such tears is real as long as a few principles<br />

are followed.<br />

1. Suture alone of avascular tears may suffice although the success<br />

rate may not be as high as in red-red or red-white tears (Noyes and<br />

Westin).<br />

2. Wrapping the meniscus in a fascial sleeve as demonstrated by<br />

Henning already in 1991 may be successful and although technically<br />

difficult and time-intensive be rewarding with 70% of healing.<br />

Despite these promising results that technique has not really be<br />

followed.<br />

3. Since 2003 we wrap avascular meniscus tears in a Collagen Matrix<br />

which can act as an internal bioreactor attracting cells of the synovial<br />

fluid and the blood, preferably from intra-osseous source richer in<br />

MSC’s (Jacobi and Jakob; Juelke at al). In animal experiments other<br />

authors have shown the superior effect for healing when autologous<br />

cells were embedded in the suture gap with an impregnated matrix<br />

(Pabbruwe et al; Peretti et al).<br />

In summary, future will allow us to focus more attention on the<br />

maintenance of meniscal tissue and value it higher than in the<br />

past so as to save menisci by reversing today’s tendency where<br />

most efforts are focused on the reconstruction of the ACL. We<br />

would welcome when in future torn ACL’s would be allowed to heal<br />

naturally, maybe by adding some cells and growth factors and when<br />

on the other hand careful surgical assistance would be offered to<br />

avascular meniscus tears. It is time to change the thinking in this<br />

regard!<br />

Picture 1<br />

Avascular Meniscus tear site is covered with collagen I/III Matrix<br />

(Geistlich Biomaterials). The insertion and fixation can be done using<br />

any of the classical fixation methods.<br />

Picture 2<br />

Invasion of meniscal synoviocytes and MSCs of synovial fluid and<br />

bone onto matrix that attracts them like a sponge and into rupture<br />

gap.


References:<br />

Baratz ME, Rehak DC, Fu FH, Rudert MJ. Peripheral tear of the<br />

meniscus. The effect of open versus arthroscopic repair on<br />

intraarticular contact stresses in the human knee. Am J Sports Med<br />

1988;16(1):1<strong>–</strong>6<br />

Baratz ME, Fu FH, Mengato R. Meniscal tears: the effect of<br />

meniscectomy and of repair on intraarticular contact areas and<br />

stress in the human knee. A preliminary report. Am J Sports Med<br />

1986;14(4):270<strong>–</strong>275<br />

Englund M, Guermazi A, Lohmander LS. The meniscus in knee<br />

osteoarthritis. Rheum Dis Clin North Am 2009;35(3):579-590<br />

Henning CE, Yearout KM, Vequist SW, et al (1991). Use of the fascia<br />

sheath coverage and exogenous fibrin clot in the treatment of<br />

complex meniscal tears. Am J Sports Med 19:626-631<br />

Jacobi M, Jakob RP (2010)Meniscal repair: enhancement of healing<br />

process. In: The meniscus (R.V. Philippe Beaufils, ed), Springer<br />

Verlag, Berlin Heidelberg, pp 129-135.<br />

Jülke H, Jakob RP, Brehm W, Mainil-Varlet P, Schäfer B, Nesic D.<br />

Meniscus repair in a goat model: an Autologous Chondrocyte<br />

Implantation (ACI)-like approach for tears in the avascular zone (In<br />

preparation)<br />

Lee SJ, Aadalen KJ, Malaviya P, et al. Tibiofemoral contact mechanics<br />

after serial medial meniscectomies in the human cadaveric knee. Am<br />

J Sports Med 2006;34(8):1334<strong>–</strong>1344<br />

Noyes F, Barber, Westin, BW. Arthroscopic repair of meniscus tears<br />

extending into the avascular zone with or without anterior cruciate<br />

reconstruction in patients 4 years of age or older. The Journal of<br />

Arthroscopy and Related Surgery. Vol 16, Issue 8, November 2000,<br />

pages 822-829<br />

Noyes F, Barber-Westin SD. Arthroscopic <strong>Repair</strong> of Meniscal Tears<br />

Extending into the Avascular Zone in Patients Younger Than Twenty<br />

Years of Age. Presented at the interim meeting of the AOSSM, San<br />

Francisco, California, March 2001. Am J Sports Med July 2002 vol. 30<br />

no. 4 589-600<br />

Noyes F, Barber, Westin, BW. <strong>Repair</strong> of Complex and Avascular<br />

Meniscal Tears and Meniscal Transplantation. The Journal of Bone &<br />

Joint Surgery. 2010; 92:1012-1029<br />

Peretti GM, Caruso EM, Randolph MA, et al (2001) Meniscal repair<br />

using engineered tissue. J Orthop Res 19:278-285<br />

Peretti GM, Gill TJ, Xu JW, et al (2004) Cell-based therapy for meniscal<br />

repair: a large animal study. Am J Sports Med 32:146-158<br />

Pabbruwe MB, Esfandiari E, Kafienah W, Tarlton JF, Hollander AP<br />

(2009). Induction of cartilage integration by a chondrocyte/collagenscaffold<br />

implant. Biomaterials 30(26):4277-86<br />

Pabbruwe MB, Kafienah W, Tarlton JF, Mistry S, Fox DJ, Hollander AP<br />

(2010). <strong>Repair</strong> of meniscal cartilage white zone tears using a stem<br />

cell/collagen-scaffold implant. Biomaterials 31(9):2583-91<br />

Seung Beom Han et al. Unfavorable Results of Partial Meniscectomy<br />

for Complete Posterior Medial Meniscus Root Tear With Early<br />

Osteoarthritis: A 5- to 8-Year Follow-Up Study. The Journal of<br />

Arthroscopic and Related Surgery, Vol 26, No 10 (October), 2010: pp<br />

1326-1332<br />

Extended Abstracts 119<br />

3.3.3<br />

How critical is the meniscus to the pathology of OA?<br />

P. Lavigne<br />

<strong>Montreal</strong>/<strong>Canada</strong><br />

Introduction: In 1897, Bland and Sutton described the meniscus as<br />

“a functionless remains of a leg muscle”. It was not until 1936, that a<br />

specific role for the meniscus proposed using a canine model. In his<br />

classic study, King showed for the first time a chondroprotective role<br />

for the meniscus. He suggested that “The amount of degeneration<br />

appears to be roughly proportional to the size of the segment<br />

removed”. King further suggested that the amount of meniscus<br />

removed during surgery should be minimized.This was the first of<br />

many studies associating meniscus pathology and later development<br />

of osteoarthritis, which is the topic of this review.<br />

Content: Fairbanks, in 1948, described roentgenographic changes<br />

of osteoarthritis following meniscectomy (1). He concluded that<br />

meniscectomy was not totally innocuous because it interferes at<br />

least temporarily with joint biomechanics. Many years later, basic<br />

science studies combined with clinical observations have identified<br />

several meniscal functions, such as load distribution, shock<br />

absorption, lubrication, assistance in articular cartilage nutrition and<br />

maintenance of joint stability. Despite these evidences, open total<br />

meniscectomy remained the standard of care for meniscal tears untill<br />

the mid 1970s. With the advent of arthroscopy and the mounting<br />

literature regarding menisci function, partial meniscectomy began<br />

to be discussed in the late 1970s. Today, arthroscopic meniscal<br />

surgery is one of the most commonly performed orthopaedic<br />

procedures. The annual incidence of meniscal tears is 60 to 70 per<br />

100 000 general population, with a peak incidence between 21 and<br />

30 years of age in male and 11 and 20 years in female. An estimated<br />

850 000 meniscal procedures are performed in the United States<br />

yearly. In the 1980s, Baratz et al showed decrease in joint contact<br />

surface areas and increase in peak contact pressures (up to 235%)<br />

when total meniscectomy was performed (2). Clinical observations<br />

of increased susceptibility to osteoarthritis following meniscectomy<br />

later supported biomechanical studies. In 1998, Roos et al reported<br />

that total meniscectomy resulted in a relative risk of 14 of developing<br />

knee osteoarthritis 21 years later (3). Many studies also reported<br />

radiographic signs of knee osteoarthritis in about 50% of patients<br />

10 to 20 years after meniscectomy and the extent of meniscal<br />

resection is now believe to relate to the degree of radiographic<br />

osteoarthritis (4,5). A better understanding of meniscus structure,<br />

intrinsic meniscal healing capacity and function in the last 20 years<br />

has led to a “repair/replace” instead of a “remove” approach.<br />

This change in clinical practice led to the introduction of meniscal<br />

repair techniques and meniscus allograft transplantation in hopes<br />

of preventing degenerative joint disease resulting from partial or<br />

subtotal meniscectomy. While some studies have shown fewer<br />

radiologic signs of osteoarthritis with meniscal repair, others have<br />

failed to report a substantial protection of degenerative changes<br />

progression following meniscus repair (6,7).<br />

Recently, Paxton et al systematically reviewed the literature<br />

comparing meniscal repair and partial meniscectomy (8). They were<br />

able to compare 6 studies with a minimum follow up of 10 years that<br />

reported either radiographic or MRI progression of osteoarthritis<br />

following meniscus repair or partial meniscectomy. They found<br />

that 78% of patients undergoing meniscus repair showed no<br />

radiographic signs of osteoarthritis compared to 64% in the partial<br />

meniscectomy patients at a minimum of ten years. Further, Fairbank<br />

grade 0 or 1 changes were found in 97% of patients after meniscus<br />

repair compared to 87% in the partial meniscectomy patients. These<br />

results are in accordance with the study of Stein et al, published in<br />

2010 (9). In a cohort study including 42 medial meniscus repairs<br />

compared to 39 medial meniscectomy, Stein reported that 80%<br />

of patients in the repair group showed no radiographic signs of<br />

osteoarthritis compared to 40% in the meniscectomy group at an<br />

average follow up of 8.8 years. Although recent reports suggest an<br />

apparent chondroprotective effect following meniscal repair, there is<br />

an evident need for a prospective study that would take into account<br />

various variables such as age, type of tear, mechanical alignment,<br />

history of previous trauma and surgery to name a few. Similarly,<br />

very little evidences currently exist in the literature supporting the<br />

chondroprotective effect of meniscus allograft transplantation (10).<br />

Verdonk et al have reported the outcome of 47 meniscus allograft<br />

transplantations at a minimum follow up of 10 years (11). Looking at<br />

osteoarthritis progression, 48% of knees showed signs of joint space<br />

narrowing and MRI analysis showed no progression of cartilage<br />

degeneration in 35% of operated knees. In a recent study, Marcacci<br />

et al studied 32 meniscal allograft transplantations at a minimum<br />

follow up of 3 years (12). They reported a chondroprotective effect on


120<br />

Extended Abstracts<br />

knee joint cartilage using the Yulish score both on the femoral and<br />

tibial surfaces. It will be interesting to see if the reported protective<br />

effect on cartilage degradation is maintained at a longer follow up.<br />

While articular cartilage degeneration following meniscectomy is<br />

well described, there is a relative paucity of literature on the potential<br />

chondroprotective effect following meniscal repair or meniscal<br />

allograft transplantation. It remains difficult to date, to determine<br />

to what extent these surgical techniques protect articular cartilage<br />

against osteoarthritis development. There is a need for longer<br />

clinical studies assessing the protective role of meniscus preserving<br />

surgery in the development of knee osteoarthritis.<br />

References:<br />

1. Fairbank TJ. Knee joint changes after meniscectomy. J Bone Joint<br />

Surg Br 1948; 30B:664-670.<br />

2. Baratz ME, Fu FH, Mengato R. Meniscal tears: the effect of<br />

meniscectomy and of repair on intraarticular contact areas and<br />

stress in the human knee. A preliminary report. Am J Sports Med<br />

1986; 14(4):270-275.<br />

3. Roos H, Laurén M, Adalberth T, Roos EM, Jonsson K, Lohmander LS.<br />

Knee osteoarthritis after meniscectomy: prevalence of radiographic<br />

changes after twenty-one years, compared with matched controls.<br />

Arthritis Rheum 1998; 41(4):687-693.<br />

4. Lohmander LS, Englund PM, Dahl LL, Roos EM. The long-term<br />

consequence of anterior cruciate ligament and meniscus injuries:<br />

osteoarthritis. Am J Sports Med 2007; 35(10):1756-1769.<br />

5. Englund M, Lohmander LS. Risk factors for symptomatic knee<br />

osteoarthritis fifteen to twenty-two years after meniscectomy.<br />

Arthritis Rheum 2004; 50(9):2811-2819<br />

6. Sommerlath KG. Results of meniscal repair and partial<br />

meniscectomy in stable knees. Int Orthop 1991; 15(4):347-350.<br />

7. Shelbourne KD, Carr DR. Meniscal repair compared with<br />

meniscectomy for bucket-handle medial meniscal tears in anterior<br />

cruciate ligament-reconstructed knees. Am J Sports Med 2003;<br />

31(5):718-723. 8- Paxton ES, Stock MV, Brophy RH. Meniscal repair<br />

versus partial meniscectomy: a systematic review comparing<br />

reoperation rates and clinical outcomes. Arthroscopy. 2011<br />

Sep;27(9):1275-1288.<br />

9. Stein T, Mehling AP, Welsch F, von Eisenhart-Rothe R, Jäger A. Longterm<br />

outcome after arthroscopic meniscal repair versus arthroscopic<br />

partial meniscectomy for traumatic meniscal tears. Am J Sports Med<br />

2010;38(8):1542-1548.<br />

10. McDermott I. Meniscal tears, repairs and replacement:<br />

their relevance to osteoarthritis of the knee. Br J Sports Med<br />

2011;45(4):292-297.<br />

11. Verdonk PC, Verstraete KL, Almqvist KF, De Cuyper K, Veys EM,<br />

Verbruggen G, Verdonk R. Meniscal allograft transplantation: longterm<br />

clinical results with radiological and magnetic resonance<br />

imaging correlations. Knee Surg Sports Traumatol Arthrosc<br />

2006;14(8):694-706.<br />

12. Marcacci M, Zaffagnini S, Marcheggiani Muccioli GM, Grassi A,<br />

Bonanzinga T, Nitri M, Bondi A, Molinari M, Rimondi E. Meniscal<br />

Allograft Transplantation Without Bone Plugs: A 3-Year Minimum<br />

Follow-up Study. Am J Sports Med 2012;40(2):395-403.<br />

Acknowledgments:<br />

The author wishes to thank the Fonds de Recherche en Santé du<br />

Québec (FRSQ) and University of <strong>Montreal</strong> - Department of Surgery<br />

for their support.<br />

5.10<br />

Advances in understanding of post-traumatic osteoarthritis <strong>–</strong><br />

Implications for treatment of joint injuries<br />

J. Buckwalter<br />

Iowa City/United States of America<br />

Introduction: Advances in Understanding of Post-Traumatic<br />

Osteoarthritis Implications for Treatment of Joint Injuries University of<br />

Iowa Department of Orthopaedics Joseph Buckwalter Excessive joint<br />

loadings, either single (acute contact stress) or repetitive (cumulative<br />

contact stress), cause progressive joint degeneration and subsequent<br />

development of the clinical syndrome of osteoarthritis (OA). Joint<br />

injuries causing acute excessive contact stress are common and often<br />

affect young adults: each year one in people between the ages of -<br />

seeks medical attention for treatment of joint injury, and more than<br />

% of all lower limb OA is caused by joint trauma. Despite advances<br />

in surgical treatment and rehabilitation of injured joints, the risk<br />

of OA following joint fractures has not decreased in the last years.<br />

Cumulative excessive articular surface contact stress that leads to OA<br />

results from joint dysplasia, incongruity and instability, but also may<br />

cause OA in patients without known joint abnormalities. Advances in<br />

understanding of the thresholds for mechanical damage to articular<br />

cartilage, and of the biologic mediators that cause progressive loss<br />

of articular cartilage due to excessive mechanical stress, will lead to<br />

better treatments of joint injuries and improved strategies for restoring<br />

damaged joint surfaces. Recent in vitro investigations show that<br />

reactive oxygen species (ROS) released from mitochondria following<br />

excessive articular cartilage loading can cause chondrocyte death<br />

and matrix degradation. Preventing the release of ROS or inhibiting<br />

their effects preserves chondrocytes and their matrix. Fibronectin<br />

fragments released from articular cartilage subjected to excessive<br />

loads also stimulate matrix degradation; inhibition of the molecular<br />

pathways initiated by these fragments prevents this effect. Distraction<br />

and motion of osteoarthritic articular surfaces in humans can promote<br />

joint remodeling, decrease pain and improve joint function in patients<br />

with end-stage post-traumatic OA This result, combined with the<br />

observation that chondroprogenitor cells are active in osteoarthritic<br />

joints, suggests that altered loading creates an environment that<br />

promotes beneficial joint remodeling. Taken together, these recent<br />

advances in understanding of how mechanical forces cause loss of<br />

articular cartilage, including identification of mechanically induced<br />

mediators of cartilage loss, and of how changing joint loading can<br />

promote joint remodeling provide the basis for new biologic and<br />

mechanical approaches to the prevention and treatment of OA.<br />

Content: Advances in Understanding of Post-Traumatic Osteoarthritis<br />

Implications for Treatment of Joint Injuries University of Iowa<br />

Department of Orthopaedics Joseph Buckwalter Excessive joint<br />

loadings, either single (acute contact stress) or repetitive (cumulative<br />

contact stress), cause progressive joint degeneration and subsequent<br />

development of the clinical syndrome of osteoarthritis (OA). Joint<br />

injuries causing acute excessive contact stress are common and often<br />

affect young adults: each year one in people between the ages of -<br />

seeks medical attention for treatment of joint injury, and more than<br />

% of all lower limb OA is caused by joint trauma. Despite advances<br />

in surgical treatment and rehabilitation of injured joints, the risk of<br />

OA following joint fractures has not decreased in the last years.<br />

Cumulative excessive articular surface contact stress that leads to OA<br />

results from joint dysplasia, incongruity and instability, but also may<br />

cause OA in patients without known joint abnormalities. Advances in<br />

understanding of the thresholds for mechanical damage to articular<br />

cartilage, and of the biologic mediators that cause progressive loss<br />

of articular cartilage due to excessive mechanical stress, will lead to<br />

better treatments of joint injuries and improved strategies for restoring<br />

damaged joint surfaces. Recent in vitro investigations show that<br />

reactive oxygen species (ROS) released from mitochondria following<br />

excessive articular cartilage loading can cause chondrocyte death<br />

and matrix degradation. Preventing the release of ROS or inhibiting<br />

their effects preserves chondrocytes and their matrix. Fibronectin<br />

fragments released from articular cartilage subjected to excessive<br />

loads also stimulate matrix degradation; inhibition of the molecular<br />

pathways initiated by these fragments prevents this effect. Distraction<br />

and motion of osteoarthritic articular surfaces in humans can promote<br />

joint remodeling, decrease pain and improve joint function in patients<br />

with end-stage post-traumatic OA This result, combined with the<br />

observation that chondroprogenitor cells are active in osteoarthritic<br />

joints, suggests that altered loading creates an environment that<br />

promotes beneficial joint remodeling. Taken together, these recent<br />

advances in understanding of how mechanical forces cause loss of<br />

articular cartilage, including identification of mechanically induced<br />

mediators of cartilage loss, and of how changing joint loading can<br />

promote joint remodeling provide the basis for new biologic and<br />

mechanical approaches to the prevention and treatment of OA.


References:<br />

Adkisson HDt, Martin JA, Amendola RL, Milliman C, Mauch KA, Katwal<br />

AB, Seyedin M, Amendola A, Streeter PR, Buckwalter JA. 2010. The<br />

potential of human allogeneic juvenile chondrocytes for restoraion<br />

of articular cartilage. Am J Sports Med 38:1324-1333.<br />

Anderson DD, Chubinskaya S, Guilak F, Martin JA, Oegema TR, Olson<br />

SA, Buckwalter JA. 2011. Post-traumatic osteoarthritis: Improved<br />

understanding and opportunities for early intervention. J Orthop Res<br />

29:802-809.<br />

Anderson DD, Iyer KS, Segal NA, Lynch JA, Brown TD. 2010.<br />

Implementation of discrete element analysis for subject-specific,<br />

population-wide investigations of habitual contact stress exposure.<br />

J Appl Biomech 26:215-223.<br />

Anderson DD, Van Hofwegen C, Marsh JL, Brown TD. 2011. Is<br />

elevated contact stress predictive of post-traumatic osteoarthritis<br />

for imprecisely reduced tibial plafond fractures? J Orthop Res 29:33-<br />

39.<br />

Beecher BR, Martin JA, Pedersen DR, Heiner AD, Buckwalter JA. 2007.<br />

Antioxidants block cyclic loading induced chondrocyte death. Iowa<br />

Orthop J 27:1-8.<br />

Bonasia DE, Martin JA, Marmotti A, Amendola RL, Buckwalter JA,<br />

Rossi R, Blonna D, Adkisson HD, Amendola A. 2011) Cocultures of<br />

adult and juvenile chondrocytes compared with adult and juvenile<br />

chondral fragments: in vitro matrix production. Amer J Sports Med:<br />

Electronic Publication. Brown TD, Johnston JC, Saltzman CL, Marsh<br />

JL, Buckwalter JA. 2006. Posttraumatic osteoarthritis: a first estimate<br />

of incidence, prevalence, and burden of disease. J Orthop Trauma<br />

20:739-744.<br />

Buckwalter JA. 1992. Mechanical Injuries of Articular <strong>Cartilage</strong>. In:<br />

Finerman, G. editor, Biology and Biomechanics of the Traumatized<br />

Synovial Joint. Park Ridge IL: American Academy of Orthopaedic<br />

Surgeons, pp. 83-96.<br />

Buckwalter JA, Brown TD. 2004. Joint injury, repair and remodeling:<br />

Roles in post-traumatic osteoarthritis. Clin Ortho Rel Res 423:7-<br />

16. Buckwalter JA, Martin JA, Brown TD. 2006. Perspectives on<br />

chondrocyte mechanobiology and osteoarthritis. Biorheology<br />

43:603-609.<br />

Cross JD, Wenke JC, Buckwalter JA, Ficke JR, Johnson AE. Post-<br />

Traumatic Osteoarthritis Caused by Battlefield Injuries is the Primary<br />

Source of Disability in Warriors. J American Academy of Orthopaedic<br />

Surgeons (submitted)<br />

Ding L, Heying E, Nicholson N, Stroud NJ, Homandberg GA, Buckwalter<br />

JA, Guo D, Martin JA. 2010. Mechanical impact induces cartilage<br />

degradation via mitogen activated protein kinases. Osteoarthritis<br />

<strong>Cartilage</strong> 18:1509-1517.<br />

Goodwin W, McCabe D, Sauter E, Reese E, Walter M, Buckwalter JA,<br />

Martin JA. 2010. Rotenone prevents impact-induced chondrocyte<br />

death. J Orthop Res 28:1057-1063. Heiner AD, Martin JA, McKinley<br />

TO, Goetz JE, Thedens DR, Brown TD (in press) Frequency Content<br />

of <strong>Cartilage</strong> Impact Force Signal Reflects Acute Histologic Structural<br />

Damage. <strong>Cartilage</strong>.<br />

Intema F, Thomas TP, Anderson DD, Elkins JM, Brown TD, Amendola<br />

A, Lafeber FP, Saltzman CL. 2011. Subchondral bone remodeling is<br />

related to clinical improvement after joint distraction in the treatment<br />

of ankle osteoarthritis. Osteoarthritis <strong>Cartilage</strong>.<br />

Koelling S, Kruegel J, Irmer M, Path JR, Sadowski B, Miro X, Miosge<br />

N. 2009. Migratory chondrogenic progenitor cells from repair tissue<br />

during the later stages of human osteoarthritis. Cell Stem Cell 4:324-<br />

335.<br />

Martin JA, Brown T, Heiner A, Buckwalter JA. 2004. Post-traumatic<br />

osteoarthritis: the role of accelerated chondrocyte senescence.<br />

Biorheology 41:479-491.<br />

Martin JA, Klingelhutz AJ, Moussavi-Harmi F, Buckwalter JA. 2004.<br />

Effects of oxidative damage and telomerase activity on human<br />

articular cartilage chondrocyte senscence. J Gerontology: Biol<br />

Sciences 59:B324-36.<br />

Martin JA, Buckwalter JA. 2006. Post-traumatic osteoarthritis: The role<br />

of stress induced chondrocyte damage. Bioreheology 43:517-521.<br />

Extended Abstracts 121<br />

Martin JA, McCabe D, Walter M, Buckwalter JA, McKinley TO.<br />

2009. N-acetylcysteine inhibits post-impact chondrocyte death in<br />

osteochondral explants. J Bone Joint Surg Am 91:1890-1897.<br />

Maxian TA, Brown TD, Weinstein SL. 1995. Chronic stress tolerance<br />

levels for human articular cartilage: two nonuniform contact models<br />

applied to long-term follow-up of CDH. J Biomech 28:159-166.<br />

McKinley TO, Rudert MJ, Koos DC, Brown TD. 2004. Incongruity<br />

versus instability in the etiology of posttraumatic arthritis. Clin<br />

Orthop Relat Res:44-51.<br />

McKinley TO, Tochigi Y, Rudert MJ, Brown TD. 2008. The effect of<br />

incongruity and instability on contact stress directional gradients in<br />

human cadaveric ankles. Osteoarthritis <strong>Cartilage</strong> 16:1363-1369.<br />

McKinley TO, Tochigi Y, Rudert MJ, Brown TD. 2008. Instabilityassociated<br />

changes in contact stress and contact stress rates near a<br />

step-off incongruity. J Bone Joint Surg Am 90:375-383.<br />

Ramakrishnan P, Hecht BA, Pedersen DR, Lavery MR, Maynard<br />

J, Buckwalter JA, Martin JA. 2010. Oxidant conditioning protects<br />

cartilage from mechanically induced damage. J Orthop Res 28:914-<br />

920.<br />

Segal NA, Anderson DD, Iyer KS, Baker J, Torner JC, Lynch JA, Felson<br />

DT, Lewis CE, Brown TD. 2009. Baseline articular contact stress levels<br />

predict incident symptomatic knee osteoarthritis development in the<br />

MOST cohort. J Orthop Res 27:1562-1568.<br />

Seol D,. McCabe DJ, Choe H, Zheng H, Jang K, Walter M, Yu Y, Lehman<br />

A, Ramakrishnan PS, Buckwalter JA, Martin JA. Chondrogenic<br />

Progenitor Cell Responses to <strong>Cartilage</strong> Injury. Osteoarthritis <strong>Cartilage</strong><br />

(submitted) Thomas TP, Anderson DD, Mosqueda TV, Van Hofwegen<br />

CJ, S. L. Hillis SL, Marsh JL T. D. Brown TD. 2010. Objective CT-based<br />

metrics of articular fracture severity to assess risk for posttraumatic<br />

osteoarthritis. J Orthop Trauma 24(12): 764-769.<br />

Tochigi Y, Buckwalter JA, Martin JA, Hillis SL, Zhang P, Vaseenon<br />

T, Lehman AD, Brown TD. 2011. Distribution and progression of<br />

chondrocyte damage in a whole-organ model of human ankle intraarticular<br />

fracture. J Bone Joint Surg Am 93:533-539.<br />

Tochigi Y, Vaseenon T, Heiner AD, Fredericks DC, Martin JA, Rudert<br />

MJ, Hillis SL, Brown TD, McKinley TO. 2011. Instability dependency<br />

of osteoarthritis development in a rabbit model of graded anterior<br />

cruciate ligament transection. J Bone Joint Surg Am 93:640-647.<br />

Acknowledgments:<br />

The work described in this presentation, the abstract and the<br />

references was supported by the National Insitutes of Health -<br />

National Institute of Arthritis, Musculskeletal and Skin Diseases and<br />

the Veterans Adminstration.


122<br />

Extended Abstracts<br />

5.20<br />

<strong>Cartilage</strong> <strong>Repair</strong>: Where are we now? Where are we going?<br />

D. Grande<br />

Manhasset/United States of America<br />

Introduction: The Clinical Problem of <strong>Cartilage</strong> Damage<br />

Lesions in articular cartilage can cause considerable musculoskeletal<br />

morbidity, with significant economic and social implications.<br />

Osteoarthritis (OA) has a significant impact on human health, particularly<br />

in the active military and veteran’s population who are at higher risk for<br />

cartilage trauma over the course of active duty. Although cartilage has<br />

a relatively simple structure compared to other tissues, cartilaginous<br />

injuries can be extremely unforgiving. The limited blood supply in<br />

cartilage is thought to be responsible for the inadequate repair post<br />

injury. A substantial fraction (~12%) of the overall burden of OA arises<br />

secondary to joint trauma, where the risk of post-traumatic OA (PTOA)<br />

ranges from 20% to 50% [1, 2]. Currently, 9% of the U.S. population<br />

aged 30 and older has OA of the hip or knee, costing an estimated $28.6<br />

billion dollars with >400,000 primary knee replacements currently<br />

being performed each year in the U.S. alone [3].<br />

Content: History of a Cell Based Strategy for <strong>Cartilage</strong> <strong>Repair</strong>: Autologous<br />

Chondrocyte Transplantation<br />

In the early 1980’s the concept of healing cartilage with predominantly<br />

hyaline tissue was largely considered a myth. Popular procedures at the<br />

time included Pirdie drilling and abrasion arthroplasty which resulted<br />

in largely fibrocartilage to fibrous tissue. It was at that time I first met<br />

Lars Peterson,MD who was on a sports medicine sabbatical while I was<br />

a graduate student at the Hospital for Joint Diseases in New York City.<br />

While conducting a journal club on the subject of articular cartilage he<br />

posed this question to me: What if it was possible to properly repair<br />

articular cartilage? This clearly piqued my interest and we began a<br />

collaboration to try and develop a new method for achieving this goal.<br />

The motivation for pursuing this project were patients who had sustained<br />

cartilage injury but were still deemed to young for total joint arthroplasty<br />

which resulted in pain and disability for young active individuals. The<br />

concept of a cell based strategy was explored and determined to be<br />

a viable option. After several experiments we concluded that articular<br />

chondrocytes exhibited several intrinsic properties of the tissue we<br />

hoped to repair: They already synthesized type II collagen and aggrecan.<br />

At that time the alternate cell source was mesenchymal stem cells<br />

pioneered by Arnold Caplan at Case-Western Reserve University. These<br />

were the early 1980’s and the term tissue engineering was not yet used<br />

to define this new branch of science. The clinical strategy developed<br />

around the need to first obtain a biopsy of cartilage which would then<br />

be used to isolate free chondrocytes and expanded in culture. Based on<br />

earlier work by Benya and Shaffer, we hypothesized that chondrocyte<br />

phenotype was plastic a limited culture time and could be reestablished<br />

by return to a 3-D environment. Optimizing cell delivery and a technique<br />

for maintaining the chondrocytes within a defect was problematic as<br />

suitable biomaterial membranes were scarce at that time. The decision<br />

to use periosteum was based on its anatomical proximity to the surgical<br />

site as well as its historical use in many orthopaedic applications such<br />

interpositional arthroplasty. The first results of rabbit experiments were<br />

decidedly superior then expectations and our team realized that a new<br />

chapter in orthopaedic research had been opened. The first reports of<br />

the technique were presented at the annual meeting of the Orthopaedic<br />

Research <strong>Society</strong> in 1985 and were promptly met with skepticism as<br />

the promising results were in conflict with over two hundred years of<br />

dogma. This was followed up by two seminal publications, one in the<br />

journal of orthopaedic research in 1989 received significant attention. In<br />

spite of initial skepticism, Lars Peterson remained undeterred and was<br />

confident in our preclinical studies he then conducted the first human<br />

clinical trials using the exact same protocols.<br />

Other <strong>Cartilage</strong> <strong>Repair</strong> Strategies Developed During this Era: 1980-1990<br />

The use of immature, neonatal chondrocytes for cartilage repair was<br />

based on the higher metabolic rates of these cells compared to those<br />

of adult. These were shown to be capable of excellent repair in an avian<br />

model by Nevo and Itay [4]. Although not developed further at that time<br />

the use of young cartilage has recently been adapted by Zimmer as their<br />

product DeNovo-NT.<br />

The archetypes of plug type scaffolds for arthroscopic delivery were<br />

fabricated of carbon fiber by Dunlop Corp. in Birmingham, UK, and<br />

investigated in clinical trials by McMinn. Coutts and Amiel [5] studied<br />

cartilage repair using the bioabsorbable scaffold material poly-L-lactic<br />

acid [PLLA] with the addition of perichondrial derived chondrocytes.<br />

Their descendants include the True-Fit plug along with other similar<br />

plugs like the Chondromimetic product.<br />

The prototype for a tissue engineered strategy was developed by [6]<br />

using vicryl suture [polylactic glycolide; PLGA] formed into a rudimentary<br />

nonwoven scaffold and seeded with chondrocytes. They demonstrated<br />

that chondrocytes could generate cartilage tissue de novo as the scaffold<br />

degraded leaving only the cells and their synthesized extracellular<br />

matrix. The field of tissue engineering has seen a prolific amount of<br />

activity with respect to cell types explored [Stem cells; marrow, muscle,<br />

adipose, synovial, embryonic, IPS] and scaffold fabrication.<br />

Current State of the Articular <strong>Cartilage</strong> <strong>Repair</strong><br />

While the etiology of joint degeneration leading to PTOA occur at the<br />

molecular, cellular and tissue level, current treatments for PTOA are<br />

primarily surgical [7,8,9]. Several procedures, such as microfracture<br />

(MFX), osteochondral autograft transfer system, mosaicplasty,<br />

autologous chondrocyte implantation (ACI), and matrix-induced<br />

autologous chondrocyte implantation (MACI) [10-13] have been devised<br />

to relieve pain, restore function, and delay or halt the progression of focal<br />

cartilaginous defects. Each of these methods has its own characteristic<br />

advantages and limitations [14-17]. MFX involves the piercing of the<br />

subchondral bone to allow marrow and its host stem cells to colonize<br />

the wound bed, promoting cartilage formation that is more fibrous than<br />

hyaline in quality. Osteochondral allografting involves the transfer of<br />

bone-cartilage units from ‘healthy’ regions to damaged regions and<br />

rapidly restores load-bearing capacity and cartilage structure; however<br />

limitations arise due to donor site morbidity, lack of healthy donor tissue,<br />

and insufficient integration. ACI (injection of chondrocytes in suspension<br />

under a periosteal flap) has shown promise in small defects in non- and<br />

low-load bearing sites, however it employs adult human chondrocytes<br />

from OA cartilage, which possess a limited capacity to form a hyaline<br />

rich tissue. In MACI, a scaffold cut to the shape and size of the defect is<br />

seeded with autologous chondrocytes and secured in the defect using<br />

a fibrin glue [18]. New techniques involving tissue engineering use cells<br />

in combination with scaffolds to regenerate a cartilage plug in vitro, for<br />

implantation into the joint (e.g. [19,20]. However, these approaches<br />

generally provide a biological replacement of cartilaginous plugs, as an<br />

alternative to chondral autografts, ACI or MACI.<br />

The Need for Early Intervention in <strong>Cartilage</strong> <strong>Repair</strong><br />

The early phase of inflammation post joint trauma triggers a cascade<br />

of catabolic changes in cartilage, synovial tissue and underlying bone.<br />

While acute inflammation can be part of the normal healing process,<br />

chronic inflammation in PTOA is associated with a positive feedback<br />

cycle that augments the destructive and degenerative pathways<br />

mediated by matrix degrading enzymes, primarily MMPs. The use of<br />

an MMP inhibitor as an early intervention shortly after the incidence of<br />

injury is attractive because it may be deployed outside of a surgical unit,<br />

where oral administration may be preferred. By specifically inhibiting<br />

MMPs, we are targeting the pathophysiologic enzymes responsible<br />

for ECM breakdown, without inhibiting the other mediators of normal<br />

inflammatory responses, associated with physiological healing. MMP<br />

inhibition can reduce or potentially delay the onset of PTOA, thus<br />

decreasing the need for massively invasive procedure such as total joint<br />

replacement. Moreover, MMP interventions early in the acute posttraumatic<br />

period can significantly improve the therapeutic outcomes<br />

of treatments administered later during surgical repair, by reducing<br />

the severity of the disease. MMP inhibition is also expected to reduce<br />

the production of fibrous cartilage (inferior quality ‘scar like’ tissue) in<br />

favor of improved production of hyaline (type II collagen rich) cartilage<br />

with mechanical properties significantly improved over existing repair<br />

techniques. The production of a natural hyaline cartilage can also<br />

improve the integration of the repair tissue with surrounding tissue and<br />

underlying bone.<br />

Successful Large Defect Resurfacing<br />

While chondrocytes have been instrumental in repair of focal cartilage<br />

defects, it is unlikely that they will be available in sufficient quantity for<br />

clinical applications involving large whole condylar defects. The success<br />

of MFX is dependent on the premise that MSCs are recruited to the<br />

repair site. However, the presence of multiple populations of cells in the<br />

marrow limits the specific contribution of MSCs, normally represented<br />

in less than 0.1% of all marrow cells. The use of the homing agent SDF-<br />

1 offers a specific augmentation of MFX, resulting in an environment<br />

enriched with MSCs. The biomechanical importance of a properly<br />

functioning articular surface presents a unique challenge when treating<br />

articular cartilage pathology. The use of mechanically stable scaffolds<br />

coated with SDF-1 provides a large porous substrate for the enriched<br />

MSCs to attach and form cartilage, de novo.


In Situ Tissue Engineering as the New Paradigm for Rapid Clinical<br />

Translation<br />

MFX is successful in regeneration of small cartilage defects, but has a<br />

wide variance in the quality of the cartilage fill. <strong>Cartilage</strong> repair quality<br />

is thought to be directly proportional to the recruitment of MSCs to the<br />

wound site. In-vitro studies by our group have shown that SDF-1 can<br />

affect the recruitment of MSCs by acting as a homing molecule, and can<br />

also affect their growth and differentiation. Moreover, we examined<br />

the efficacy of SDF-1 for cartilage repair in vivo. Defects were repaired<br />

with either scaffold only (PLLA or collagen sponge), scaffold coated<br />

with SDF-1 (50 ng/ml), or left empty. The quality of repair was analyzed<br />

histologically after 4 weeks with safranin-O/fast green staining and<br />

quantified with the O’Driscoll score. The quality of repair in the empty<br />

defect was poor, with fibrillation at articular surface, and degenerative<br />

changes in surrounding tissue. The SDF-1 coated scaffolds had<br />

congruent and smooth cartilage surfaces that were well integrated with<br />

the surrounding cartilage. Quantitatively, the scaffold only group had<br />

modest improvement in cartilaginous filling compared to empty defect.<br />

However, the SDF-1 coated scaffold had significantly improved quality<br />

of cartilage repair with GAG rich cartilage ECM. Interestingly, we saw no<br />

major differences in repair quality between PLLA and collagen scaffold<br />

treated groups. These findings on small defects support the hypothesis<br />

that SDF-1 promotes significant improvement in cartilaginous repair.<br />

We theorize that this is mechanistically due to increased recruitment<br />

of MSCs into the injury site. For large defect repairs, a clinical strategy<br />

would be to administer allogenic MSCs to increase the number of<br />

reparative cells available for contiguous repair across the defect<br />

Summary: Effective and comprehensive treatment of all phases of<br />

injury is essential in order to address the initial structural joint injury<br />

and as well the inflammatory and destructive processes that follow<br />

and can result in more diffuse joint pathology. Acute, subacute and<br />

chronic surgical resurfacing of larger or multifocal symptomatic hyaline<br />

tissue traumatic defects as well as associated osteochondral defects<br />

is essential. Although current marrow stimulation (microfracture) and<br />

autogenous osteochondral transplantation techniques have been<br />

available, these methods have had less effective application in treating<br />

larger sizes defects (> 2 centimeter 2). Use of volume stable scaffolds<br />

coated to chemotactically enhance mesenchymal stem cell recruitment<br />

to the repair construct is an attractive option. The concurrent use<br />

of biochemical catabolic inhibitors that can reduce degradative<br />

inflammatory mechanisms that can biologically expedite recovery and<br />

improve the quality of structural repair is a promising strategy as there<br />

are few surgical techniques that result in superior and durable clinical<br />

outcomes in young active patients.<br />

References:<br />

Literature Cited<br />

1. Brown, T.D., et al., Posttraumatic osteoarthritis: a first estimate of<br />

incidence, prevalence, and burden of disease. J Orthop Trauma, 2006.<br />

20(10): p. 739-44. PMC 17106388. http://www.ncbi.nlm.nih.gov/<br />

entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_<br />

uids=17106388<br />

2. Dirschl, D.R., et al., Articular fractures. J Am Acad Orthop Surg, 2004. 12(6):<br />

p. 416-23. PMC 15615507. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi<br />

?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=15615507<br />

3. Kurtz, S., et al., Projections of primary and revision hip and knee<br />

arthroplasty in the United States from 2005 to 2030. J Bone Joint Surg<br />

Am, 2007. 89(4): p. 780-5. PMC 17403800. http://www.ncbi.nlm.nih.<br />

gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&lis<br />

t_uids=17403800<br />

4. Nevo Z., Itay S., Abramovici A. Use of cultured embryonal chick epiphyseal<br />

chondrocytes as grafts for defects in chick articular cartilage Clin Orthop<br />

Relat Res. 1987 Jul;(220):284-303.<br />

5. Chu C.R., Coutts R.D., Yokshioka M., Harwood F.L., Monosov A.Z., Amiel<br />

D., Articular cartilage repair using allogeneic perichondrocyteseeded<br />

biodegradable porous polylactic acid (PLA): A tissue-engineering study,<br />

Journal of Biomedical Materials Research, 13 SEP 2004 P 1147-1154 V29<br />

Issue 9, DOI: 10.1002/jbm.820290915<br />

6. Cima L.G., Vacanti J.P., Cacanti C., Ingber D., Mooney D., Langer R.;<br />

Journal of Biomechanical Engineering, Vol. 113, No. 2. (1991), pp. 143-151,<br />

doi:10.1115/1.2891228<br />

7. Anderson, D.D., et al., Post-traumatic osteoarthritis: improved<br />

understanding and opportunities for early intervention. J Orthop Res. 29(6):<br />

Extended Abstracts 123<br />

p. 802-9. PMC 21520254. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi<br />

?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=21520254<br />

8. Furman, B.D., S.A. Olson, and F. Guilak, The development of<br />

posttraumatic arthritis after articular fracture. J Orthop Trauma, 2006.<br />

20(10): p. 719-25. PMC 17106385. http://www.ncbi.nlm.nih.gov/<br />

entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&lis<br />

t_uids=17106385<br />

9. Daher, R.J., et al., New methods to diagnose and treat cartilage<br />

degeneration. Nat Rev Rheumatol, 2009. 5(11): p. 599-607. PMC<br />

19786989. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retr<br />

ieve&db=PubMed&dopt=Citation&list_uids=19786989<br />

10. Evans, P.J., A. Miniaci, and M.B. Hurtig, Manual punch versus power<br />

harvesting of osteochondral grafts. Arthroscopy, 2004. 20(3): p. 306-<br />

10. PMC 15007320. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?c<br />

md=Retrieve&db=PubMed&dopt=Citation&list_uids=15007320<br />

11. Mithoefer, K., et al., The microfracture technique for the treatment<br />

of articular cartilage lesions in the knee. A prospective cohort study. J<br />

Bone Joint Surg Am, 2005. 87(9): p. 1911-20. PMC 16140804. http://<br />

www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMe<br />

d&dopt=Citation&list_uids=16140804<br />

12. Hangody, L., et al., Mosaicplasty for the treatment of articular defects<br />

of the knee and ankle. Clin Orthop Relat Res, 2001(391 Suppl): p. S328-<br />

36. PMC 11603716. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?c<br />

md=Retrieve&db=PubMed&dopt=Citation&list_uids=11603716<br />

13. Brittberg, M., et al., Treatment of deep cartilage defects in the knee<br />

with autologous chondrocyte transplantation. N Engl J Med, 1994.<br />

331(14): p. 889-95. PMC 8078550. http://www.ncbi.nlm.nih.gov/<br />

entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&lis<br />

t_uids=8078550<br />

14. Knutsen, G., et al., A randomized trial comparing autologous<br />

chondrocyte implantation with microfracture. Findings at five years. J<br />

Bone Joint Surg Am, 2007. 89(10): p. 2105-12. PMC 17908884. http://<br />

www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMe<br />

d&dopt=Citation&list_uids=17908884<br />

15. Bentley, G., et al., A prospective, randomised comparison of<br />

autologous chondrocyte implantation versus mosaicplasty for<br />

osteochondral defects in the knee. J Bone Joint Surg Br, 2003. 85(2): p.<br />

223-30. PMC 12678357. http://www.ncbi.nlm.nih.gov/entrez/query.fc<br />

gi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=12678357<br />

16. Saris, D.B., et al., Characterized chondrocyte implantation results<br />

in better structural repair when treating symptomatic cartilage defects<br />

of the knee in a randomized controlled trial versus microfracture. Am J<br />

Sports Med, 2008. 36(2): p. 235-46. PMC 18202295. http://www.ncbi.<br />

nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Ci<br />

tation&list_uids=18202295<br />

17. Knutsen, G., et al., Autologous chondrocyte implantation compared<br />

with microfracture in the knee. A randomized trial. J Bone Joint Surg<br />

Am, 2004. 86-A(3): p. 455-64. PMC 14996869. http://www.ncbi.nlm.<br />

nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citatio<br />

n&list_uids=14996869<br />

18. Steinwachs, M., New technique for cell-seeded collagen-matrixsupported<br />

autologous chondrocyte transplantation. Arthroscopy,<br />

2009. 25(2): p. 208-11. PMC 19171282. http://www.ncbi.nlm.nih.gov/<br />

entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&lis<br />

t_uids=19171282<br />

19. Hung, C.T., et al., A paradigm for functional tissue engineering of<br />

articular cartilage via applied physiologic deformational loading. Ann<br />

Biomed Eng, 2004. 32(1): p. 35-49. PMC 14964720. http://www.ncbi.<br />

nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Ci<br />

tation&list_uids=14964720<br />

20. Mauck, R.L., et al., Functional tissue engineering of articular<br />

cartilage through dynamic loading of chondrocyte-seeded agarose<br />

gels. J Biomech Eng, 2000. 122(3): p. 252-60. PMC 10923293. http://<br />

www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMe<br />

d&dopt=Citation&list_uids=10923293


124<br />

Extended Abstracts<br />

7.1<br />

The use of autologous cartilage fragments - clinical studies<br />

J. Farr<br />

Indianapolis/United States of America<br />

Introduction: Current surgical treatment options for symptomatic<br />

cartilage lesions include debridement/lavage, marrow stimulation<br />

with or without augmentation, osteochondral autograft implantation,<br />

stored osteochondral allograft implantation, and autologous<br />

chondrocyte implantation first and subsequent generations.7-12<br />

There are two new approaches that use minced/particulated<br />

cartilage to treat chondral defects. One investigational technique<br />

uses autograft cartilage (<strong>Cartilage</strong> Autograft Implantation System<br />

[CAIS]; DePuy Mitek Rayham MA USA) and the other uses juvenile<br />

allograft cartilage (DeNovo NT; Zimmer Warsaw IN USA).3,13 This<br />

report will focus on CAIS (originally reported at the Warsaw ICRS) as<br />

Dr. Caborn will report on DeNovo NT.<br />

Content: Even a few years ago, the concept that cartilage could be<br />

implanted and heal without underlying bone would be considered ill<br />

conceived by most cartilage surgeons. While using minced articular<br />

cartilage to repair a chondral defect is relatively new in US journals,<br />

Albrecht, a German scientist, reported the technique in 1983.19 He<br />

demonstrated that in the rabbit cartilage autograft cut into small<br />

pieces without bone led to cartilage defect healing noting, however,<br />

the subchondral plate was breached. United States based scientists<br />

Francois Binette and Ed Lu noted (as others in the field of investigating<br />

chondrocytes had) that during preparation to enzymatically free the<br />

chondroctyes, after mincing and before enzymatic digestion of the<br />

extracellular matrix, some chondrcytes appeared outside of the<br />

matrix and new matrix formation appeared adjacent to the minced<br />

cartilage fragments. Binette, Lu and their associates began a series<br />

of experiments to further investigate these findings first with in vitro<br />

experiments and then in the mouse, goat, and finally horse preclinical<br />

models.20,21. In essence, these studies showed that chondrocytes in<br />

the cartilage fragments could “escape” from the dense extra-cellular<br />

matrix, migrate, multiply, and form new hyaline-like cartilage tissue<br />

matrix that would integrate with the surrounding host tissue. This new<br />

tissue demonstrated both basilar and marginal integration. Unlike<br />

cultured chondrocytes that assume a spindle shape morphology<br />

during ex vivo culture, the chondrocytes from the minced cartilage<br />

retained the standard chondrocyte spheroid shape.21 In aggregate<br />

these studies demonstrated that autograft cartilage defect repair<br />

was possible when autograft cartilage was mechanically minced into<br />

cubes of 1-2 mm and then implanted into the defect.19,21<br />

This preclinical data was compelling enough for the FDA to approve<br />

a safety pilot study of what the sponsor referred to as CAIS (<strong>Cartilage</strong><br />

Autograft Implantation System).3 The clinical outcomes are now<br />

published at 2 years and an extension follow up study is complete<br />

to 4 years post-op with publication to follow. A parallel pilot study<br />

has been completed in Europe. In the US, the FDA has approved a<br />

pivotal study of the technique, which began recruiting patients in<br />

2010 and will enroll over 300 patients for a randomized prospective<br />

comparison of CAIS to MFX (that is, CAIS is not available for general<br />

use but limited to clinical study patients).<br />

INDICATIONS/CONTRAINDICATIONS<br />

CAIS is being investigated for treatment of symptomatic articular<br />

cartilage defects of the knee in patients from age 18-55. Arthroscopic<br />

evaluation should confirm one or two ICRS grade 3a to 4a chondral<br />

lesions of the femoral condyles or trochlea that after debridement<br />

measure from 1 cm2 to 10 cm2. As is standard for cartilage restoration<br />

in general, the meniscal tissue should be functional, the knee stable<br />

and limb alignment neutral. Potential contraindications to CAIS<br />

include bipolar lesions, significant underlying subchondral bony<br />

edema, or subchondral bone defect > 6mm subchondral bone loss.<br />

SURGICAL TECHNIQUE<br />

In the pilot study, standard arthroscopy confirmed CAIS inclusion/<br />

exclusion criteria, and hyaline cartilage similar to the amount harvested<br />

for ACI (roughly 200 mg) was then harvested arthroscopically from a<br />

low load-bearing surface using a proprietary device (DePuy Mitek)<br />

that minced the cartilage into 1- to 2-mm pieces. The device then<br />

uniformly dispersed the minced cartilage onto a biodegradable<br />

scaffold that is comprised of an absorbable co-polymer foam of<br />

35% polycaprolactone [PCL] and 65% polyglycolic acid [PGA] and<br />

reinforced with a polydioxanone [PDO mesh] [DePuy Mitek]). The<br />

minced cartilage was then glued to the scaffold using a commercially<br />

available fibrin sealant (Tisseel, Baxter, Illinois). A mini-arthrotomy<br />

was performed, and the defect was identified and prepared similar to<br />

the technique utilized for ACI. A template was cut to match the lesion<br />

and guided cutting the minced cartilage/scaffold construct. The sized<br />

CAIS scaffold implant was transferred to the defect with the cartilage<br />

fragments facing the subchondral bone and affixed with 2 or more<br />

biodegradable polyglycolic acid (PGA)/polydioxanone (PDO) staple<br />

anchors (DePuy Mitek.<br />

Rehabilitation Protocol<br />

The rehabilitation program for the pilot study purposely mimicked<br />

the Steadman microfracture rehabilitation program. This program<br />

begins with protection of the cartilage repair process and then<br />

progresses toward controlled loading, increased range of motion,<br />

and progressive muscle strengthening.3 Rehabilitation is specific<br />

for the patellofemoral and tibiofemoral compartment. Standard limb<br />

strengthening then follows with functional progression to activity.<br />

Results<br />

The FDA approved the pilot study in the United States to assess the<br />

safety of CAIS and to test whether CAIS improves quality of life by<br />

using standardized outcomes assessment tools. In the study, 29<br />

patients were randomized with the intent to treat with either MFX<br />

or CAIS. Patients were followed at pre-determined time points for 2<br />

years using several standardized outcomes assessment tools (SF-36,<br />

<strong>International</strong> Knee Documentation Committee [IKDC], Knee injury and<br />

Osteoarthritis Outcome Score [KOOS]). Magnetic resonance imaging<br />

was performed at baseline, 3 weeks, and 6, 12, and 24 months.<br />

Lesion size and <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong> (ICRS) grade<br />

were similar in both groups. General outcome measures (e.g. physical<br />

component score of the SF-36) indicated an overall improvement in<br />

both groups, and no differences in the number of adverse effects<br />

were noted in comparisons between the CAIS and MFX groups. The<br />

IKDC score of the CAIS group was significantly higher compared with<br />

the MFX group at both 12 and 24 months. Select subdomains (4/5) in<br />

the KOOS instrument were significantly different at 12 and 18 months,<br />

and all subdomains (Symptoms and Stiffness, Pain, Activities of Daily<br />

Living, Sports and Recreation, Knee-related Quality of Life) were<br />

significantly increased at 24 months in CAIS versus MFX.<br />

Qualitative analysis of the imaging data did not note differences<br />

between the 2 groups in fill of the graft bed, tissue integration, or<br />

presence of subchondral cysts. Patients treated with MFX had a<br />

significantly higher incidence of intralesional osteophyte formation<br />

(54% and 70% of total number of lesions treated) at 6 and 12 months<br />

when compared with CAIS (8% and 25% of total number of lesions<br />

treated). To be reported in 2012, the results at 3 years in Europe and<br />

4 years in the US demonstrate durability compared to the 2 year<br />

outcomes.<br />

DISCUSSION<br />

In short term clinical studies, CAIS appears to be safe and feasible, with<br />

improvements in subjective patient scores, and with MRI evidence of<br />

good defect fill.2,3,26 There are several potential advantages to this<br />

technique. CAIS does not require the violation of the subchondral<br />

bone, as is necessary for marrow stimulation procedures that may<br />

compromise subsequent revision surgeries as reported by Minas.27<br />

Obviously, CAIS does not create an osteochondral defect for treatment<br />

of a cartilage only lesion as is necessary for osteochondral auto and<br />

allograft transplantation. CAIS and DeNovo NT utilize a strategy of<br />

cartilage-cartilage healing in the defect bed. This may help to avoid<br />

problems of bony healing as seen in failed osteochondral allograft<br />

procedures, including lack of bone incorporation, necrosis, and<br />

avascular necrosis like collapse. As CAIS is single stage procedureit<br />

avoids the cost of cell culturing and the need for a second surgery<br />

for implantation. A disadvantage specific to CAIS is the potential for<br />

donor site morbidity at cell harvest; however, this risk is minimal and<br />

is, in theory, similar to the risk involved in ACI harvest.<br />

CONCLUSION<br />

CAIS appears to be a promising new treatment option for patients<br />

with symptomatic focal chondral defects in the knee. Further studies<br />

including formal reporting of the 4 year follow up of US patients, the<br />

3 year follow up of the European patients and the ongoing US pivotal<br />

study will improve evidence based recommendations.<br />

Financial Disclosure:<br />

Farr is a consultant for Depuy Mitek and is a principle investigator for<br />

the CAIS pivotal study


References:<br />

1. Chondral Defect <strong>Repair</strong> with Particulated Juvenile <strong>Cartilage</strong><br />

Allograft. Updated 2010;Available from: http://www.zimmer.<br />

com/web/enUS/pdf/DeNovo_Chondral_Defect_<strong>Repair</strong>_White_<br />

Paper_05_2010.pdf<br />

2. Farr J, Yao JQ. Chondral defect repair with particulated juvenile<br />

cartilage allograft. <strong>Cartilage</strong> 2011;epub ahead of print.<br />

3. Cole BJ, Farr J, Winalski CS, Hosea T. Outcomes after a single-stage<br />

procedure for cell-based cartilage repair: a prospective clinical safety<br />

trial with 2-year follow-up. Am J Sports Med 2011;39:1170-1179<br />

4. Hjelle K, Solheim E, Strand T, Muri R, Brittberg M. Articular cartilage<br />

defects in 1,000 knee arthroscopies. Arthroscopy 2002;18:730-734<br />

5. Maletius W, Messner K. The effect of partial meniscectomy on the<br />

long-term prognosis of knees with localized, severe chondral damage.<br />

A twelve- to fifteen-year follow up. Am J Sports Med 1996;24:258-<br />

262<br />

6. Saris DBF, Vanlauwe J, Victor J, Almqvist KF. Treatment of<br />

symptomatic cartilage defects of the knee: characterized chondrocyte<br />

implantation results in better clinical outcome at 36 months in<br />

a randomized trial compared to microfracture. Am J Sports Med<br />

2009;37 Suppl 1:10S-19S<br />

7. Brittberg M, Lindahl A, Nilsson A, Ohlsson C. Treatment of<br />

deep cartilage defects in the knee with autologous chondrocyte<br />

transplantation. N Engl J Med 1994;331:889-895<br />

8. Zaslav K, Cole B, Brewster R, DeBerardino T. A prospective study<br />

of autologous chondrocyte implantation in patients with failed prior<br />

treatment for articular cartilage defect of the knee: results of the<br />

Study of the Treatment of Articular <strong>Repair</strong> (STAR) clinical trial. Am J<br />

Sports Med 2009;37:42-55<br />

9. Gross AE, Shasha N, Aubin P. Long-term followup of the use of<br />

fresh osteochondral allografts for posttraumatic knee defects. Clin<br />

Orthop Relat Res 2005;435:79-87<br />

10. Hangody L, Kish G, Kárpáti Z, Szerb I, Udvarhelyi I. Arthroscopic<br />

autogenous osteochondral mosaicplasty for the treatment of femoral<br />

condylar articular defects. A preliminary report. Knee Surg Sports<br />

Traumatol Arthrosc 1997;5:262-267<br />

11. Moseley JB, O’Malley K, Petersen NJ, Menke TJ. A controlled trial<br />

of arthroscopic surgery for osteoarthritis of the knee. N Engl J Med<br />

2002;347:81-88<br />

12. Steadman JR, Rodkey WG, Singleton SB, Briggs KK. Microfracture<br />

technique for full-thickness chondral defects: technique and clinical<br />

results. Operative techniques in orthopaedics 1997;7:300-304<br />

13. Farr J. DeNovo NT natural graft tissue. Poster presented at: 8th<br />

World Congress of the <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong>; 2009;<br />

Miami, FL<br />

14. Asik M, Ciftci F, Sen C, Erdil M, Atalar A. The microfracture technique<br />

for the treatment of full-thickness articular cartilage lesions of the<br />

knee: midterm results. Arthroscopy 2008;24:1214-1220<br />

15. Cole BJ, Pascual-Garrido C, Grumet RC. Surgical management<br />

of articular cartilage defects in the knee. J Bone Joint Surg Am<br />

2009;91:1778-1790<br />

16. Saris DBF, Vanlauwe J, Victor J, Haspl M. Characterized chondrocyte<br />

implantation results in better structural repair when treating<br />

symptomatic cartilage defects of the knee in a randomized controlled<br />

trial versus microfracture. Am J Sports Med 2008;36:235-246<br />

17. Knutsen G, Drogset JO, Engebretsen L, Grøntvedt T. A<br />

randomized trial comparing autologous chondrocyte implantation<br />

with microfracture. Findings at five years. J Bone Joint Surg Am<br />

2007;89:2105-2112<br />

18. Knutsen G, Engebretsen L, Ludvigsen TC, Drogset JO. Autologous<br />

chondrocyte implantation compared with microfracture in the knee. A<br />

randomized trial. J Bone Joint Surg Am 2004;86-A:455-464<br />

19. Albrecht F, Roessner A, Zimmermann E. Closure of osteochondral<br />

lesions using chondral fragments and fibrin adhesive. Arch Orthop<br />

Trauma Surg 1983;101:213-217<br />

Extended Abstracts 125<br />

20. Frisbie DD, Lu Y, Kawcak CE, DiCarlo EF. In vivo evaluation of<br />

autologous cartilage fragment-loaded scaffolds implanted into<br />

equine articular defects and compared with autologous chondrocyte<br />

implantation. Am J Sports Med 2009;37 Suppl 1:71S-80S<br />

21. Lu Y, Dhanaraj S, Wang Z, Bradley DM. Minced cartilage without<br />

cell culture serves as an effective intraoperative cell source for<br />

cartilage repair. J Orthop Res 2006;24:1261-1270<br />

22. Cheung HS, Cottrell WH, Stephenson K, Nimni ME. In vitro collagen<br />

biosynthesis in healing and normal rabbit articular cartilage. J Bone<br />

Joint Surg Am 1978;60:1076-1081<br />

23. Adkisson HD, Martin JA, Amendola RL, Milliman C. The potential<br />

of human allogeneic juvenile chondrocytes for restoration of articular<br />

cartilage. Am J Sports Med 2010;38:1324-1333<br />

24. Stockwell RA. The interrelationship of cell density and cartilage<br />

thickness in mammalian articular cartilage. J Anat 1971;109:411-421<br />

25. Namba RS, Meuli M, Sullivan KM, Le AX, Adzick NS. Spontaneous<br />

repair of superficial defects in articular cartilage in a fetal lamb<br />

model. J Bone Joint Surg Am 1998;80:4-10<br />

26. Bonner KF, Daner W, Yao JQ. 2-year postoperative evaluation of<br />

a patient with a symptomatic full-thickness patellar cartilage defect<br />

repaired with particulated juvenile cartilage tissue. J Knee Surg<br />

2010;23:109-114<br />

27. Minas T, Gomoll AH, Rosenberger R, Royce RO, Bryant T.<br />

Increased failure rate of autologous chondrocyte implantation after<br />

previous treatment with marrow stimulation techniques. Am J Sports<br />

Med 2009;37:902-908<br />

28. Gomoll A, Probst C, Farr J, Cole B, Minas T. Use of a type I/III bilayer<br />

collagen membrane decreases reoperation rates for symptomatic<br />

hypertrophy after autologous chondrocyte implantation. Am J Sports<br />

Med 2009;37:205-235<br />

29. Gibson T, Brian Davis W, Curran R. The long-term survival of<br />

cartilage homografts in man. Br J Plast Surg 1959;11:177-177<br />

30. Stone KR, Walgenbach AW, Freyer A, Turek TJ, Speer DP. Articular<br />

cartilage paste grafting to full-thickness articular cartilage knee joint<br />

lesions: a 2- to 12-year follow-up. Arthroscopy 2006;22:291-299<br />

31. Crawford DC, Heveran CM, Cannon WD, Foo LF, Potter HG. An<br />

autologous cartilage tissue implant NeoCart for treatment of grade<br />

III chondral injury to the distal femur: prospective clinical safety trial<br />

at 2 years. Am J Sports Med 2009;37:1334-1343<br />

8.1.1<br />

Treatment of Osteochondral Lesions of the Talus<br />

E. Giza<br />

Davis/United States of America<br />

Introduction: Epidemiology of Talus Injury Ankle sprains are common<br />

injuries with a daily incidence of one per 10,000 people per day.<br />

There are 23,000 ankle injuries each day in the USA with basketball,<br />

soccer, and volleyball having the highest incidence per hour of play.<br />

Both acute injury and chronic lateral ligament insufficiency can lead<br />

to talar osteochondral lesions.<br />

Content: Talus Anatomy The talus articulates with the tibia, fibula,<br />

calcaneus, and navicular and of its surface 60% is covered in cartilage.<br />

It serves as a link to transmit load to the foot and experiences up to<br />

six times body weight with each step. It affords pronation-supination<br />

and dorsiflexion-plantarflexion through multiple facets, a unique<br />

bony architecture and multiple ligament attachments. The talus<br />

has no tendonous attachments. The chondrocytes have a limited<br />

capacity for intrinsic repair due to encasement in matrix proteins and<br />

limited vascularity. Full thickness injuries below the subchondral<br />

bone allow recruitment of marrow elements but injuries greater than<br />

two to four millimeters have a poor potential to heal with normal<br />

appearing cartilage. A chondrocyte’s biosynthetic machinery is<br />

impaired with senescence and further hampers healing of defects<br />

regardless of technique. Evaluation of talus injury Symptoms Patients<br />

usually present with persistent pain after an ankle sprain. Time to<br />

presentation may be prolonged. Pain is typically localized to the<br />

side of the lesion and may be accompanied by intermittent swelling,


126<br />

Extended Abstracts<br />

stiffness, and weakness. Mechanical symptoms such as locking and<br />

catching are variable. High level athletes can usually tolerate the pain<br />

and may present after numerous injury events. A complete history<br />

pertaining to both medial and lateral ankle instability is paramount<br />

as findings on exam can be subtle. Alignment of the hindfoot should<br />

be noted as pes cavus deformity can contribute to inversion injuries.<br />

Stability should be tested with both anterior drawer testing and<br />

bilateral prone inversion challenge testing of the ankle and subtalar<br />

joints. Surgical Treatment For talus OCD, consistency in reporting<br />

of outcomes are inconsistent, and may studies have small sample<br />

sizes. Acute restoration is performed on the rare lesion with a bony<br />

defined fragment that is technically repairable. True osteochondritis<br />

dissecans lesions are possible candidates but overall relatively<br />

rare. Delamination must not be present and the best candidates are<br />

young athletes with good healing potential who are able to comply<br />

with weight bearing restrictions. Techniques include open reduction<br />

and internal fixation with recessed transchondral screws, retrograde<br />

internal fixation and biodegradable fixation devices. Arthroscopy,<br />

Curettage and Microfracture/Drilling The initial surgical treatment for<br />

most lesions involves arthroscopy with curettage and microfracture.<br />

Arthroscopy is indicated for unstable lesions and those that have<br />

failed conservative treatment with stable lesions. Best results are<br />

seen in patients with small lesions (<br />

1.5 cm², age less than 50 years old and failure of either non-operative<br />

treatment or an index cartilage surgery. Kissing talo-tibial lesions<br />

and axial defects or deformity are contraindications. Autografts are<br />

harvested as full thickness cartilage with subchondral bone. Multiple<br />

small sized cylindrical plugs of varying small diameters (2-4 mm) are<br />

harvested to implant and cover a larger area in the mosaicplasty<br />

technique. The intervening spaces are filled with newly generated<br />

fibrocartilage but hyaline transplanted cartilage populates the bulk<br />

remainder. Advantages for the OATS or mosiacplasty technique<br />

include the use of autograft tissue with robust fixation and a single<br />

stage procedure that can be performed open or arthroscopically.<br />

Surgical exposure of the posterior talar dome, large defect size, and<br />

matching cartilage shape, curvature and depth of resection are all<br />

technically demanding skills that need to be practiced on a cadaver<br />

specimen with appropriate preoperative planning. Both techniques<br />

necessitate an orthogonal approach to the articular surface. Bulk<br />

fresh osteochondral allografts are replacement options for multifocal<br />

and large defects (>1cm2) particularly in the setting of avascular<br />

necrosis. Indications include subchondral and extensive disease,<br />

possibly necessitating a bipolar graft and salvage procedures<br />

for failed autografting restoration attempts. Patients seeking to<br />

minimize the donor site morbidity of autograft procedures can be<br />

considered. Both fresh frozen and fresh osteochondral allografts are<br />

available however cryopreservation results in a loss of chondrocyte<br />

viability. Autologuous Chondrocyte Implantation (ACI) Autologuous<br />

Chondrocyte Implantation (ACI) uses cultured chondrocytes with a<br />

periosteal patch. Harvesting is first accomplished from either the<br />

knee or ankle and can include the damaged area of chondral tissue.<br />

The biopsy typically yields 2-3 million cells and these cells can<br />

be stored for greater than one year. Once the decision is made to<br />

transplant the cells are cultured for 6-8 weeks and results in at least<br />

12 million cells available for implantation. The process of growing the<br />

cells increases the number of viable cells for implantation by at least<br />

ten fold. The ability to culture large numbers of cells for implantation<br />

is critical for the success of the procedure. Matrix-Based Chondrocyte<br />

Implantation Matrix-Based Chondrocyte Implantation (MACI) is<br />

similar to ACI. Cells are first harvested from the ankle or knee. Next,<br />

chondrocyte cells are enzymatically separated from the matrix and<br />

cultured to produce 15-20 million cells. Instead of injecting the<br />

cultured chondrocytes under periosteam, the cells are imbedded<br />

in a type I/III collagen membrane bilayer. Similar to ACI, the MACI<br />

technique allows the surgeon to increase by at least ten fold the cells<br />

available for implantation into the defect. A second stage operation is<br />

used to implant the membrane over the talar defect. The advantages<br />

of MACI include that it is technically easier than ACI and no tibial/<br />

malleloar osteotomy is needed. Two operations are required and<br />

the cells can be stored for greater than one year after initial harvest.<br />

Summary Osteochondral defects of the talus continue to be a<br />

challenging entity. The role of ankle instability as both an acute and<br />

chronic etiology is evident. Early attempts at stimulating healing,<br />

whether by curettage and/or drilling and microfracture have been<br />

shown to be equivalent. Patients with failed first line treatments are<br />

candidates for cartilage replacement surgeries such as OATS, ACI<br />

and MACI. Larger, non-healing lesions are potential candidates for<br />

bulk osteochondral allografts. Development of precise indications,<br />

optimal rehabilitation, all-arthroscopic techniques, and advanced<br />

biomaterials to accelerate healing are current topics of ongoing<br />

investigation.<br />

8.1.2<br />

Clinical options for cartilage reconstruction in the ankle.<br />

R. Ferkel<br />

Van Nuys/United States of America<br />

Introduction: In 1743, Hunter stated that “From Hippocrates down to<br />

the present age, we shall find that an ulcerated cartilage is universally<br />

allowed to be very troublesome disease; that amidst of a cure with<br />

more difficulty than a carious bone; and that, when destroyed it is<br />

never recovered B. PRIMARY PROBLEM 1. Fibrocartilage repair not<br />

been shown to withstand mechanical wear over time 2. Fibrocartilage<br />

might degenerate and lesion progress into osteoarthritis<br />

Content: I. Treatment Alternatives A. Osteochondral autograft plugs<br />

(OATS) 1. “Robs Peter to pay Paul” 2. Question of long term problems<br />

with femoral plug holes; subsequent pain 3. Size limitations for autograft<br />

transplantation 4. Advantage of single setting procedure 5. Scranton et<br />

al. (2005): 50 patients, significant improvement; 90% good/excellent.<br />

Karlsson scores increased 24 to 83 B. Osteochondral allografts 1. Chu<br />

(1999): 55 knees, average follow-up 75 months 2. Overall success 76%<br />

3. Gross (2001): 9 patients; 6 patients remain in situ, mean survival 11<br />

years. 3 cases required fusion 4. Bugbee et al. (2010): 12 ankles a. Mean<br />

follow-up 38 months b. Average previous surgery 1.8 c. Graft survival<br />

83% d. Mean OMAS improved 28 to 71 5. Kelikian et al. (2011): allografts<br />

on 38 patients a. Mean follow-up 38 months b. Graft survival 89% c.<br />

AOFAS score from 52 to 79 II. Autologous Chondrocyte Implantation A.<br />

Definition <strong>–</strong> implantation of in vitro cultured autologous chondrocytes<br />

using periosteal tissue or membrane cover after expansion of isolated<br />

chondrocytes B. ACI <strong>–</strong> Generations 1. Generation 1 <strong>–</strong> Carticel suspended<br />

under periosteal flap or membrane 2. Generation 2 <strong>–</strong> Carticel inserted<br />

under tissue patch or onto carrier scaffold 3. Generation 3 <strong>–</strong> Carrier-free,<br />

immature cartilage tissue III. Indications A. Indications 1. Age 15 to 55<br />

2. Focal defect > 1 cm2 3. Unipolar (only talus affected) 4. Contained<br />

5. Edge loading 6. Failed previous surgery 7. Large lesions, extensive<br />

subchondral cystic changes B. Relative 1. Multifocal unipolar lesions 2.<br />

Uncontained lesions IV. Contraindications A. Relative 1. Kissing (bipolar)<br />

lesions 2. No previous surgery 3. Early degenerative changes B. Absolute<br />

1. Osteoarthritis 2. Uncorrected malalignment 3. Uncorrected instability<br />

V. Surgical Procedure A. Biopsy procedure <strong>–</strong> Step 1 1. Chondral biopsy<br />

200-300 mg 2. Biopsy done in intercondylar notch of knee or ankle<br />

arthroscopically B. Surgical Technique <strong>–</strong> Step 2 1. Medial or lateral<br />

malleolar osteotomy performed under fluoroscopy 2. Defect preparation<br />

- vertically incise and remove defect and all damaged cartilage from<br />

subchondral bone 3. Harvest periosteum from medial distal tibia,<br />

measure appropriately 4. Biogide a. Absorbable porcine bilayer<br />

collagen I/III membrane has been used in knee and ankle ACI in place of<br />

periosteum b. Has a rough and smooth layer c. Made by same company<br />

as Chondro-gide and very similar 5. Apply fibrin glue circumferentially<br />

along periosteum-cartilage margin or at suture lines 6. Cell implantation<br />

a. Insert soft catheter tip through opening of periosteum and inject<br />

cells, then close defect and put additional fibrin glue over it VI. Surgical<br />

Technique with “Sandwich Procedure” A. Done for large cystic lesions<br />

>6 mm deep B. Osteochondral lesion is excised and cyst curetted out<br />

C. Bone graft obtained from proximal or distal tibia, depending on size<br />

of cyst. Place 2 layers of periosteum or BioGide VII. Postoperative Care<br />

A. Early phase, day 1 to week 8 B. Transition phase, 8-12 weeks C. Mid<br />

Phase, 3-5 months D. Final phase, 6-12 months E. Time line for activities<br />

1. Low impact activities 4-6 months 2. Repetitive impact activities 6-8<br />

months 3. High level activities 10-12 months VIII. Results of ACI <strong>–</strong> First<br />

Generation (ACI-P) A. Whittaker et al. (2005): 10 patients 1. Mean followup<br />

23 months 2. Mazur score increased 23 points 3. Increased donor site<br />

morbidity 4. “Second looks” done on 9 patients; showed filled defects,<br />

stable cartilage 5. Biopsies showed mostly fibrocartilage with some<br />

hyaline cartilage B. Baums et al. (2006): 12 patients 1. Mean follow-up<br />

63 months 2. Hannover score increased from 40 to 86 points 3. AOFAS<br />

increased 45 points 4. Patients involved in competitive sports able to<br />

return to full activity level C. Ferkel (2011): 32 patients 1. First 11 patients<br />

have been reviewed and published in AJSM 2. Current: Follow-up 29 of


32 (91%) 3. Average age: 34 (18-54) 4. Average follow-up: 70 months<br />

(24-129) 5. 9 “sandwich” procedures done, with bone grafting of large<br />

cystic underlying defect and use of two periosteal grafts back to back<br />

6. 2nd look arthroscopy 90% of patients (26/29) 7. Results: Excellent:<br />

8; Good: 15; Fair: 5; Poor: 1 8. Entire paper presented at AAOS 2011 X.<br />

Second Generation ACI A. Variety of scaffolds being used in Europe,<br />

implanted through small arthrotomy or arthroscopically. 1. Used as patch<br />

and cells inserted underneath 2. Cells seeded onto scaffold membrane<br />

B. Collagen-covered autologous chondrocyte implantation (CACI or<br />

ACI-C) 1. Absorbable porcine bilayer collagen I/III membrane 2. Chondro-<br />

Gide membrane with one compact and one porous surface 3. Gooding<br />

found no difference in results between periosteum and membrane cover<br />

in knees with CACI C. Hyalograft C 1. Benzyl ester of Hyaluronic acid 2.<br />

Bioabsorbs in 3 months 3. Marcacci et al.: 175 patients with grafts in knee,<br />

46 month mean follow-up. Results: 93% improvement at ICRS 2006 4.<br />

Giannini et al. (2008) <strong>–</strong> 46 patients in ankle a. Mean age 32; follow-up<br />

3 years b. Preop 57; postop 90 mean AOFAS score c. Biopsies collagen<br />

type II D. Membrane/matrix autologous chondrocyte implantation (MACI)<br />

1. Highly purified type I/III collagen membrane 2. Guillen and Abelow<br />

presented first 50 cases (42 knees; 8 ankles) 3. 8 ankles (ages 22-46)<br />

4. Large full thickness cartilage lesions of talus (2-6 cm) 5. 5/6 good/<br />

excellent results with follow-up 4 months-2 1/2 years 6. Giza et al.: MACI<br />

on 10 patients; AOFAS 61 to 73 XI. Third generation ACI A. Use carrierfree,<br />

immature cartilage tissue B. Lack of carrier scaffold C. Avoids carrier<br />

integration, degradation and biocompatibility complications D. Jubel et<br />

al. used alginate matrix to produce cell-rich chondrocyte disc in MFC of 48<br />

sheep E. Chondral defects treated with De Novo cartilage transplantation<br />

showed qualitatively better micro and macroscopic regeneration than<br />

those with periosteal flaps alone 1. Fair and Yao reported early good<br />

results in knee 2. Adams et al. reported particulated juvenile articular<br />

cartilage transplant for OLT with early good results XII. Summary A.<br />

Zengerink et al. has nicely summarized when to use which treatment for<br />

OLT B. Is there a critical defect size for poor outcome? 1. Choi et al (2009)<br />

found initial defect size is important prognostic factor for OLT and can<br />

serve as a basis for preop surgical decisions 2. OLT defect of 150 mm2<br />

or greater as calculated from MRI has a high correlation of having a poor<br />

clinical outcome<br />

References:<br />

Adams SB, Yao JQ, Schon LC: Particulated juvenile articular cartilage<br />

allograft transplantation for osteochondral lesions of the talus. Tech Foot<br />

Ankle Surg 2011;10:92-98.<br />

Assenmacher JA, Kelikian AS, Gottlob C, Kodros S: Arthroscopically<br />

assisted autologous osteochondral transplantation for osteochondral<br />

lesions of the talar dome: an MRI and clinical follow-up study. Foot Ankle<br />

Int 2001;22:544-551.<br />

Bazaz R, Ferkel RD: Treatment of osteochondral lesions of the talus with<br />

autologous chondrocyte implantation. Tech Foot Ankle Surg 2004;3:45-<br />

52.<br />

Baums MH, Heidrich G, Schultz W, et al.: Autologous chondrocyte<br />

transplantation for treating cartilage defects of the talus. J Bone Joint Surg<br />

2006;88A:303-308.<br />

Choi et al. Osteochondral lesion of the talus: Is there a critical defect size<br />

for poor outcome? Am J Sports Med 2009;37:1974-1980. Easley ME, Latt<br />

LD, Santangelo JR, et al.: Osteochondral lesions of the talus. J Amer Acad<br />

Orthop Surg 2010;18:616-630.<br />

Easley ME, Scranton PE: Osteochondral autologous transfer system. Foot<br />

Ankle Clin North Am 2003;8:275-290. El-Rashidy H, Villacis D, et al.: Fresh<br />

osteochondral allograft for the treatment of cartilage defects of the talus: a<br />

retrospective review. J Bone Joint Surg Am 2011;93:1634-1640.<br />

Farr J, Yao JQ: Chondral defect repair with particulated juvenile cartilage<br />

allograft. <strong>Cartilage</strong> 2011;2:346-353.<br />

Ferkel RD: Arthroscopic Surgery: The Foot and Ankle. Philadelphia;<br />

Lippincott, 1996. Ferkel RD, Scranton PE Jr., Stone JW, Kern B: Chronic<br />

osteochondral lesions of the talus: therapeutic dilemmas. Part II:<br />

Surgical treatment of osteochondral lesions of the talus. Inst Course Lect<br />

2010;59:387-206.<br />

Ferkel RD, Zanotti RM, Komenda GA, et al.: Arthroscopic treatment of<br />

chronic osteochondral lesions of the talus. Am J Sports Med 2008;36:1750-<br />

1762.<br />

Getgood A, Brooks R, Fortier L, Rushton N: Articular cartilage tissue<br />

engineering. J Bone Joint Surg 2009;91B:565-576.<br />

Giannini S, Buda R, Grigolo B, Vannini F: Autologous chondrocyte<br />

transplantation in osteochondral lesions of the ankle joint. Foot Ankle Int<br />

2001;22:513-517.<br />

Extended Abstracts 127<br />

Giannini S, Buda R, Vannini F, et al.: One-step bone marrow-derived cell<br />

transplantation in talar osteochondral lesions. Clin Orthop Relat Res<br />

2009;467:33073320.<br />

Giannini S, Buda R, Faldini C, et al.: Surgical treatment of osteochondral<br />

lesions of the talus in young active patients. J Bone Joint Surg<br />

2005;87A(suppl 2):28-41.<br />

Giannini S, Buda R, Vannini F, et al. Arthroscopic autologous chondrocyte<br />

implantation in osteochondral lesions of the talus: surgical technique and<br />

results. Am J Sports Med 2008;36:873-880.<br />

Gikas PD, Bayliss L, Bentley G, Briggs TWR: An overview of autologous<br />

chondrocyte implantation. J Bone Joint Surg 2009;91B:997-1006.<br />

Giza E, Sullivan M, Ocel D, et al.: Matrix-induced autologous chondrocyte<br />

implantation of talus articular defects. Foot Ankle Int 2010;31:747-753.<br />

Görtz S, De Young AJ, Bugbee WD: Fresh osteochondral allografting for<br />

osteochondral lesions of the talus. Foot Ankle Int 2010;31:283-90.<br />

Gross AE, Agnidis Z, Hutchison CR: Osteochondral defects of the talus<br />

treated with fresh osteochondral allograft transplantation. Foot Ankle Int<br />

2001;22:385-391.<br />

Guo Q-W, Hu Y-L, Jiao C, et al.: Arthroscopic treatment for osteochondral<br />

lesions of the talus: analysis of outcome predictors. Clin Med J 2010;123:296-<br />

300.<br />

Hahn DB, Aanstoos ME, Wilkins RM: Osteochondral lesions of the talus<br />

treated with fresh talar allografts. Foot Ankle Int 2010;31:277-282.<br />

Harris JD, Siston RA, Pan X, Flanigan DC: Autologous chondrocyte<br />

implantation: A systematic review. J Bone Joint Surg 2010;92A:2220-<br />

2233.<br />

Hangody L, Kish G, Modis L, et al.: Mosaicplasty for the treatment of<br />

osteochondritis dissecans of the talus: two to seven year results in 36<br />

patients. Foot Ankle Int 2001;22:552-558, 2001.<br />

Jackson DW, Scheer MJ, Simon TM: <strong>Cartilage</strong> substitutes: overview of basic<br />

science and treatment options. J Am Acad Orthop Surg 2001;9:37-52.<br />

Jones DG, Peterson L: Autologous chondrocyte implantation. J Bone Joint<br />

Surg 2006;88A:2501-2520.<br />

McGahan PJ, Pinney SJ: Current concept review: Osteochondral lesions of<br />

the talus. Foot Ankle Int 2010;31:90-101.<br />

Mitchell ME, Giza E, Sullivan MR: <strong>Cartilage</strong> transplantation techniques for<br />

talar cartilage lesions. J Am Acad Orthop Surg 2009;17:407-414.<br />

Nam EK, Ferkel RD, Applegate GR: Autologous chondrocyte implantation<br />

of the ankle: a 2 to 5 year follow-up. Am J Sports Med 2009;37:274-284.<br />

O’Loughlin PF, Heyworth BE, Kennedy JG: Current concepts in the diagnosis<br />

and treatment of osteochondral lesions of the ankle. Am J Sports Med<br />

2010;38:392-404.<br />

Peterson L, Brittberg M, Lindahal A: Autologous chondrocyte transplantation<br />

of the ankle. Foot Ankle Clin North Am 2003;8:291-303.<br />

Peterson L, Vasiliadis HS, Brittberg M, Lindahl A: Autologous chondrocyte<br />

implantation: a long-term follow-up. Am J Sports Med 2010;38:1117-1124.<br />

Raikin SM: Fresh osteochondral allografts for large-volume cystic<br />

osteochondral defects of the talus. J Bone Joint Surg 2009;91A:2818-<br />

2826.<br />

Ronga M, Grassi FA, Bulgheroni P: Arthroscopic autologous chondrocyte<br />

implantation for the treatment of a chondral defect in the tibial plateau of<br />

the knee. Arthroscopy 2004;20:79-84.<br />

Safran MR, Kim H, Zaffagnini S: The use of scaffolds in the management of<br />

articular cartilage injury. J Am Acad Orthop Surg 2008;16:306-311.<br />

Scranton PE Jr., McDermott JE: Treatment of type V osteochondral lesions<br />

of the talus with ipsilateral knee osteochondral autografts. Foot Ankle Int<br />

2001;22:380384.<br />

Verhagen RAW, Struijs PAA, Bossuyt PMM, van Dijk CN: Systematic review<br />

of treatments strategies for osteochondral defects of the talar dome. Foot<br />

Ankle Clin North Am 2003;8:233-242.<br />

Whittaker J-P, Smith G, Makwana N, et al.: Early results of autologous<br />

chondrocyte implantation in the talus. J Bone Joint Surg 2005;87B:179-<br />

183.<br />

Zengerink M, Szerb I, Hangody L, et al.: Current concepts: treatment of<br />

osteochondral ankle defects. Foot Ankle Clin 2006;11:331-359.


128<br />

Extended Abstracts<br />

8.1.3<br />

Dermal graft application in Hallux Limitus / Rigidus for salvage<br />

of the 1st metatarsophallangeal joint<br />

L.J. Sanchez<br />

Orlando/United States of America<br />

Introduction: Hallux Limitus / Hallux Rigidus presents as a<br />

gradual “deformity” of the foot Greater toe metatarsophallangeal<br />

joint. Its presentation includes a partial luxation of the first<br />

metatarsophallangeal joint, where the Hallux proximal phallanx<br />

base presents a plantar attitude or luxation in reference to the<br />

first metatarsal head. This foot pathology has been well described<br />

in a pletora of medical writtings in literature. It was first described<br />

as “Hammerzehenplattfuss” or “Hallux-hammertoe-flatfoot”, by<br />

Nicoladoni, in 1881. And it has been given other names as well,<br />

but it was Cotteril who denomiated it as “Hallux Rigidus”, in 1887.<br />

Later, the term of “Hallux Limitus” was given in 1937, by Hiss, as<br />

to describe the limited capasity of the hallux toe to dorsiflex on the<br />

first metatarsophallangeal joint. Part of the definition includes the<br />

inability of the hallux to dorsiflex on the first metatarsal head past<br />

60 degrees, as an initial presentation. Coughlin and Shumas, (1999)<br />

provided a radigraphical classification of this pathology. Up to<br />

date, this classification is most used to describe the Hallux Limitus<br />

/ Rigidus pathology . This classification starts with a Grade”0”,<br />

as the first metatarsophallageal joint looses 20% of dorsiflexion<br />

capasity, and is able to dorsiflex 40-60 degrees, and the X-Rays<br />

on this joint are normal, and there is no pain. The classification<br />

goes up to Grade “4” , where there is a “stiff joint” or ankylosis of<br />

the metatarsophallangeal joint, with “loose body fragments” or<br />

“OCD’s”. Hallux limitus / rigidus presents multi factorial causes<br />

and ethiologic factors. Its ethiology is divide in Traumatic vs Nontraumatic<br />

causes. A description of the non-traumatic ethiologic<br />

causes will be mentioned as part of this article. Even thou<br />

“arthrodesis of the first metatarsophalangeal joint” has become the<br />

“gold standard” in the surgical approach to Coughlin stage 3 and<br />

4 of hallux Limitus/Rigidus, there are other operative options that<br />

offers a less agressive approach with better initial outcomes and<br />

provides the patient with less post operative complications. This<br />

article presents 22 cases of Hallux Limitus/ Rigidus patients foot<br />

pathology treated with a combination of a first metatarsal distal<br />

decompression osteotomy, a cheilectomy of the joint in question,<br />

a mechanism to perform a full surgical release of the contracted /<br />

fibrotic tissue surrounding the joint and the plantar sesamoids, and<br />

the use a dermal allogenic scaffold graft to cover the first metatarsal<br />

head and mantain the glidding mechanism after surgical correction.<br />

These surgical interventions have been performaed in a two years<br />

period , between January of 2009, and January 2011. A postoperative<br />

“Modified Hallux-Metatarsophalangeal joint and Interphallangeal<br />

Joint Scale” have not been yet finished , and thus , not provided<br />

at the time this article / presentation is realised. age on patients<br />

ranged from 34 to 70 years of age, with mean average age of 54<br />

years of age. A postoperative patient pain / function / satisfaction<br />

results is provided as a preliminary information for which all 22<br />

patients responded to “possibly have the same surgery if the other<br />

foot was involved with the same pathology”. All patients were able<br />

to initiate full protected walking in a surgical shoe, within a period<br />

of three weeks of the surgery. There has been no surgical revitions,<br />

and minor complications of wound closure slight necrosis was seen<br />

in two patients, which resolved promptly upon diligent in office<br />

wound care. One patient had a moderate cellulitis / skin infection<br />

that resolvedas well upon diligent local care and antibiotic therapy.<br />

In all , (when compared to the gold standard “ joint arthrodesis”),<br />

this surgical procedure provides the patient with Hallux Limitus /<br />

Rigidus: a prompt recovery time, a reduced post surgical sequelae<br />

of possible complications, a functional ability to use normal shoe<br />

wear with orthoses, minimal bone loss in metatarsal or phallangeal<br />

components of the affected joint, and no excessive internal fixation<br />

hardware that may need to be removed.<br />

Content: The use of a”Dermal Acellular Scafold” as a joint spacer<br />

in the surgical treatment of Hallux Limitus / Rigidus has been<br />

documented in literature by several authors. It has been classify as<br />

an “Interpossitional Implant” and it has similar results to the use of<br />

“Autogenous first metatarsal capsular/periosteum interpositional<br />

grafting technique”, described in literature as well. All 22 patients<br />

in this preliminary study received Dermal acellular cadaveric scafold<br />

grafts from two different medical companies in the US: Wright<br />

Medical , “Graft Jacket” non fenestrated grafts, and Stryker US ,<br />

MMI non fenestrated acellular dermal grafts. All patients surgically<br />

intervened had a “Coughlin Hallux Limitus/rigidus staging” 2 to 4<br />

with respect to their first metatarsal arthritic disease progress. All<br />

22 patients were surgically treated equally, with minimal alterations<br />

in the surgical technique described. Steps in the surgical technique:<br />

1. A longitudinal incision approach is done dorsal and medial to the<br />

Extensor Hallucis Longus, measuring 6 cm in lenght. The incision<br />

is carried down to the 1st metatarsal periosteum and crossing the<br />

fist metatarsophallangeal joint into the base of the Halux proximal<br />

phallanx. The ful thickness of the periosteum and capsule is elevated<br />

surrounding the first metatarsal shaft, the metatarsal head , and the<br />

base of the proximal hallux phallanx. 2. A “full cheilectomy” procedure<br />

is performed around the joint, metatarsal head is completely<br />

denuded of any abnormal osseous growth, including osteophytes<br />

and deseased cartilagenous tissue surrounding the joint margins<br />

and any fibrotic tisuue is as well removed. 3. A modified transverse<br />

“V” osteotomy, (Austin or Chevron modified osteotomy), is produced<br />

to the metatarsal head and distal metatarsal shaft. The osteotomy<br />

addresses the “bunion deformity”, reducing the intermetatarsal<br />

angulation between the first and second metatarsals, as well as<br />

plantar-flexes the distal metatarsal head fragment, in order to<br />

decompresss the diseased arthritic joint, and adrresses the issue of<br />

a “first metatarsal elevatus” commonly seen in this pathology. This<br />

osteotomy is rigidly fixated with 2.0 cortical screws. In some cases<br />

whereas the bony tissue seemed soft / osteoporotic, a combination<br />

of Kishner wires and 2.0 cortical screws were utilised for proper<br />

fixation. 4. The deseased cartilage comprissing the metatarsal head<br />

and the base of the hallux proximal phallanx, was repetitively drilled<br />

with a .045” Kishner wire, so as to to create future vascular channels<br />

between the subchondral vascular bed and the internal aspect of the<br />

joint, where the “Acellular dermal graft” is to be applied. Copious<br />

irrigation of the surgical area with normal physiological saline<br />

solution is performed. 4. A Wright Medical “Graft Jacket” or a Stryker<br />

MMI Dermal Acellular Graft measuring 4cm x6cm long was cut or<br />

fashioned into a “ Cross fashion”. The 4 legs of this graft “Cross”<br />

were applied around the first metatarsal head, and tightly fixated to<br />

the first metatarsal head , proximal to the plantar head condyles or<br />

surgical neck), with 2-0 vicryl suture material, in a “tie wrap” fashion.<br />

The central aspect of the graft is well compressed against the internal<br />

metatarsal head cartilagenous area. At no time grafting material was<br />

left floating free in the joint. And no grafting material was applied to<br />

the Hallux proximal phallanx. After affixing the dermal acellular graft,<br />

the first metatarsophallangeal joint is re-articulated and the joint<br />

dorsal extension and plantarflexion range of motion is examined,<br />

making sure there is at least 70-80 degrees of dorsal extension of the<br />

proximal phallanx upon the first metatarsal head. Also, making sure<br />

there is available at least 30 degrees of plantarflexion of this joint. 5.<br />

Closure of tissues is realised in layers. The periosteum and capsular<br />

tissue is closed with 3-0 vicryl suture material. The superficial fascia<br />

underlying the skin is closed with 4-0 vicryl suture material. The skin<br />

is closed with 5-0 prolene suture material in a “horizontal matress”<br />

stitching technique. 6. The closed surgical wound is infiltrated with<br />

0.5% Marcaine plain local anesthetic, for post operative analgesia.<br />

References:<br />

1. Nicoladoni C. Uber zehenkontrakturen. Wien Klin Wochenschr<br />

51:1418, 1881.<br />

2. Cotterill JM: Condition o stiff gret toe in adolescents. Edinburgh<br />

Med J. 33:459, 1887.<br />

3. Hiss JM: Hallux Valgus: Its cause and simplified treatment. Am J.<br />

Surg 11:51, 1937.<br />

4. Nilsonne H.: Hallux rigidus and its treatment. Acta Orthop Scand<br />

1:295, 1930.<br />

6. Durrant MN, Siepert KK: Role of soft tissue structures as an<br />

etiology of hallux limitus. JAPMA 83: 173, 1993.<br />

7. Viladot A: Metatarsalgia due to biomechanical alterations of the<br />

forefoot. Orthop Clin North Am 4:165, 1973.<br />

8. Chang TJ: Stepwise approach to hallux limitus: a surgical<br />

perspective. Clin Podiatr Med Surg 13: 449, 1996.<br />

9. Hanft JR, Mason ET, Landsman AH, ET Al: A new radiographic<br />

classification for hallux limitus. J Foot Ankle Surg 32: 397, 1993.<br />

10. Kravitz SR, LA Porta GA, Lawton JH: KLL progressive staging<br />

classification of hallux limitus and hallux rigidus, Lower Extremity<br />

1:55,1994.<br />

11. Mahan KT: „Joint Preservations Techniques in Hallux Limitus/<br />

Rigidus <strong>Repair</strong>,“ in Musculoskeletal Deformities of the Lower<br />

Extremities, ed by LM Oloff, p529, WB Saunders, Philadelphia,<br />

1994.


12. Keogh P, Nagaria J, Stephens M: Cheilectomy for hallux rigidus. Ir<br />

J Med Sci 161:681, 1992.<br />

13. Sheriff MJ, Baumhauer JF: Hallux rigidus and osteoarthritis of the<br />

first metatarsophalangeal joint. J Bone Joint Surg AM 80: 898, 1998.<br />

14. O‘Driscoll SW: The healing and regeneration of articular cartilage.<br />

J Bone Joint Surg Am 80: 1795, 1998.<br />

15. Hamilton WG, O‘Malley MJ, Thompson FM, ET AL: Capsular<br />

interposition arthroplasty for severe hallux rigidus. Foot Ankle Int<br />

18: 68, 1997.<br />

16. Fuson SM: Modification of the Keller operation for increased<br />

functional capacity. J Foot Surg 21: 292, 1982.<br />

18. Roukis TS, Jacops PM, Dawson DM: A prospective comparison of<br />

clinical, radiographic, and intra-operative features of hallux limitus<br />

and hallux rigidus: short-term follow up and analysis. Paper presented<br />

at the American College of Foot and Ankle Surgeons Annual Meeting<br />

and Scientific Seminar, February 8, 2001, New Orleans.<br />

19. Poussa M, Rubak J, Ritsila V: Differentiation of the<br />

osteochondrogenic cells of the periosteum in chondrotrophic<br />

environment. Acta Orthop Scand 52: 235, 1981.<br />

20. O‘ Driscoll SW, Salter RB: The induction of neochondrogenesis<br />

in free intra-articular periosteal autografts under the influence of<br />

continous passive motion: an experimental investigation in the<br />

rabbit. J Bone Joint Surg AM 66: 1248, 1984.<br />

21. Cosentino GL: The Cosentino modification for tendon<br />

interpositional arthroplasty. J Foot Ankle Surg 34: 501, 1995.<br />

22. Roberts S, hollander AP, Caterson B, ET AL: Matrix turnover<br />

in human cartilage repair tissue in autologous chondrocyte<br />

implantation. Arthritis Rheum 44: 2586, 2001.<br />

23. Hanft JR, Merrill T, Marcinko DE, ET AL: First metatarsophallangeal<br />

joint replacement. J Foot ankle Surg 35: 78, 1996.<br />

24. Haddad SL: the use of osteotomies in the treatment of hallux<br />

limitus and rigidus. Foot ankle Clin 5: 629, 2000.<br />

25. Feltham GT, Hanks SE, Markus RE: age -based outcomes of<br />

cheilectomy for the treatment of hallux rigidus. Foot ankle Int 22:<br />

192,2001.<br />

26. Heller WA, Brage ME: the effects of cheilectomy on dorsiflexion<br />

of the first metatarsophallangeal joint. Foot Ankle Int 18: 803,1997.<br />

27. Grady JF, Axe TM: The modified Valenti procedure for the<br />

treatment of hallux limitus, a long term follow-up and analysis. J Foot<br />

Ankle Surg 38:123, 1999.<br />

28. Ganley JV, Lynch FR, Darrigan RD: Keller bunionectomy with<br />

fascia and tendon graft. JAPMA 76: 602, 1986.<br />

29. Lau JTC, Daniels TR: Outcomes following cheilectomy and<br />

interpositional arthroplasty in hallux rigidus. Foot Ankle Int 22:462,<br />

2001.<br />

30. Gusman DN, Messmer TE: Newell decompression procedure for<br />

hallux limitus; a preliminary report. JAPMA 85: 749, 1995.<br />

31. Toolan BC, Wright-Quinones VJ, Cunningham BJ, ET AL: an<br />

evaluation of the use of retrospectively adquired pre-operative<br />

AOFAS clinical rating scores to asses surgical outcome after elective<br />

foot and ankle surgery. Foot Ankle Int 22: 775,2001.<br />

32. Roukis TS, Landsman AS, Et AL: Distally based capsule-periosteum<br />

interpositional arthroplasty for hallux rigidus. Indications, operative<br />

tcnique, and short-term follow-up. JAPMA , Sept / Oct 2003, Vol 93,<br />

No 5.<br />

Acknowledgments:<br />

Provided at lecture time due to writting space limitations.<br />

Extended Abstracts 129<br />

8.2.2<br />

Expansion of chondrocyte populations on high extension culture<br />

surfaces for improved retention of phenotype<br />

T.M. Quinn 1 , B. Hinz 2 , M. Matmati 1 , D.H. Rosenzweig 1<br />

1 <strong>Montreal</strong>/<strong>Canada</strong>, 2 Toronto/<strong>Canada</strong><br />

Introduction: Cell-based cartilage therapies such as autologous<br />

chondrocyte implantation require isolation of chondrocytes<br />

from a healthy tissue biopsy, population expansion in vitro and<br />

implantation in a defect [1, 2]. During population expansion,<br />

moderate chondrocyte densities must be maintained for<br />

efficient cell growth [3]. However, contact inhibition can lead to<br />

loss of phenotype and reduced proliferation [3, 4]. This limited<br />

desirable range of cell densities means that in standard culture,<br />

repeated passaging and reseeding of chondrocytes is required.<br />

Passaging is associated with rapid changes in the chondrocyte<br />

phenotype [5, 6]. This dedifferentiation decreases the capacity for<br />

implanted chondrocytes to regenerate functional neocartilage and<br />

necessitates additional protocols for redifferentiation toward the<br />

desired phenotype. Improved understanding and control of factors<br />

promoting dedifferentiation may therefore lead to significant<br />

improvements in cell-based cartilage repair. To inhibit chondrocyte<br />

dedifferentiation during population expansion, we have developed<br />

a technique that facilitates more continuous growth of cells while<br />

limiting contact inhibition and reducing the necessity for passaging<br />

[7, 8]. Cells are grown at relatively high density on a continuously<br />

expanding elastic dish which provides increasing culture surface<br />

area as their population grows. This “continuous expansion” culture<br />

technique has been used previously to expand human mesenchymal<br />

stem cell populations more efficiently than by standard methods<br />

while maintaining pluripotent stem cell phenotype and inhibiting<br />

undesired fibrotic phenotypes [7, 8]. We hypothesized that<br />

continuous expansion culture could also be beneficial for primary<br />

chondrocytes.<br />

Content: Materials and Methods<br />

Knee joints from freshly slaughtered skeletally mature cows were<br />

obtained from a local slaughterhouse. Articular cartilage was cut<br />

from the femoropatellar groove with a scalpel, and chondrocytes<br />

were isolated according to established methods [9].<br />

Chondrocytes were cultured on several different surfaces. These<br />

included (1) high-extension silicone rubber (HESR) dishes which were<br />

continuously expanded, (2) standard polystyrene culture dishes, and<br />

(3) polystyrene culture dishes coated with approximately 1 mm of<br />

silicone rubber. All silicone rubber culture surfaces were chemically<br />

modified to coat them with covalently bound collagen type I to<br />

promote cell adhesion as previously described [7, 8].<br />

To initiate cultures, 10,000 chondrocytes per cm2 were seeded and<br />

subcultured in chondrocyte growth medium. Continuous expansion<br />

(CE) cultures were performed on high extension silicone rubber (HESR)<br />

dishes expanded from 12 cm2 to 76.8 cm2 over 10 days following a<br />

3-day initial attachment period. Surface expansion was performed<br />

using a motorized iris-like device. This 13-day period was defined<br />

as one generation, which corresponded to three conventional 1:2<br />

passages in standard (SD) culture on polystyrene. The final surface<br />

area in CE culture equaled the total surface area of the third passage<br />

in SD culture (8 wells at 9.6 cm2 each). Confluence at each passage<br />

in SD culture was ~80% and 0.25% Trypsin-EDTA solution was used.<br />

For experiments to control for silicone surface chemistry, standard<br />

cultures were performed on polystyrene dishes coated with silicone<br />

functionalized rubber (“static silicone” or SS cultures).<br />

At the end of each generation in CE, SD or SS culture, cells were<br />

trypsinized and counted. For a subsequent generation, 105 cells<br />

were then reseeded. 106 cells resulting from each generation were<br />

used for redifferentiation experiments (pellet cultures), and the<br />

remaining cells (1-2x106) were used for analyses of gene and protein<br />

expression. Cells were lysed in 1 mL of TRIzol reagent and RNA was<br />

isolated for quantitative real-time PCR analysis. PCR primers for<br />

collagen type II, aggrecan, COMP, Sox9, collagen type I, and GAPDH<br />

were generated exactly as described elsewhere [10]. Western blot<br />

analysis was also performed for alpha smooth muscle actin and<br />

α-tubulin.<br />

At the end of each generation, ~2×106 cells were centrifuged at<br />

500×g for 10 minutes in 1.5 mL microfuge tubes, and pellet cultures<br />

for assessment of de novo cartilage synthesis capacity were initiated<br />

as previously described [7, 11, 12]. TGFβ was intentionally excluded<br />

from redifferentiation medium in order to emphasize effects of CE<br />

versus SD culture without the concern of overwhelming growth


130<br />

Extended Abstracts<br />

factor stimulation. Pellets were incubated in centrifuge tubes for six<br />

days until they became firm and then transferred to a six-well plate<br />

(which helped maintain viability) and incubated for an additional<br />

six days to observe chondrocyte outgrowth. Medium was changed<br />

every two days. Pellets were then fixed with 4% paraformaldehyde<br />

and prepared for cryosectioning.<br />

For histological analysis, pellet cultures were fixed in 4%<br />

paraformaldehyde and embedded in tissue freezing medium. Frozen<br />

sections 10 μm thick were stained with Alcian Blue for proteoglycan<br />

and counterstained with Nuclear Fast Red. For immunofluorescence,<br />

sections were incubated with antibodies against phospho-histone<br />

H3, cleaved Caspase 3 and collagen type II, and visualized on an<br />

inverted fluorescence microscope.<br />

Results<br />

Chondrocyte attachment on both static silicone (SS) and standard<br />

polystyrene (SD) typically occurred by 3 days after seeding, at<br />

which point experiments began (Day 0). Cell morphology appeared<br />

similar for the two culture conditions during passaging. RNA-level<br />

expression of the cartilage-specific genes collagen type II, aggrecan,<br />

and cartilage oligomeric matrix protein (COMP) declined with each<br />

passage number for both culture conditions while collagen type I<br />

expression increased.<br />

Chondrocytes in continuous expansion (CE) culture had a more<br />

rounded and less spindle-like morphology than in SD culture. At the<br />

end of generation G1, real-time quantitative PCR revealed significantly<br />

higher expression of the cartilage-specific genes collagen type II,<br />

aggrecan and COMP in CE versus SD culture. These trends remained<br />

consistent through generations G2 and G3. In contrast, the fibrotic<br />

marker collagen type I was significantly downregulated in CE versus<br />

SD culture. Cell lysates from CE and SD cultures were subjected to<br />

SDS-PAGE and Western blot probing for the fibrotic marker α-SMA.<br />

SD culture lysates revealed a steady induction of α-SMA through the<br />

three generations which was inhibited in the CE culture lysates for<br />

generations G1 and G2. At the end of each generation, cell counting<br />

revealed fewer total chondrocytes in CE culture compared to SD<br />

culture.<br />

Pellets derived from SD cultures strongly adhered to the culture<br />

dish during the final 6 days of the 12-day pellet culture. In contrast,<br />

pellets derived from CE cultures only weakly adhered. Moreover,<br />

chondrocyte migration from adhered pellets was clearly evident for<br />

all three generations of SD culture but only for generation G3 of CE<br />

culture. CE pellets contained more sulphated glycosaminoglycans<br />

compared to SD pellets, as evident by Alcian blue staining. After<br />

generation G1, both CE and SD pellets stained strongly for collagen<br />

type II immunofluorescence. However, after generations G2 and<br />

G3, collagen type II immunofluorescence was only detectable in CE<br />

pellet cultures.<br />

Discussion<br />

Cell morphology, cartilage-specific gene expression, fibrotic<br />

gene expression, and cell numbers all indicated that continuous<br />

expansion (CE) culture preserves the chondrocyte phenotype during<br />

population expansion. Standard (SD) culture chondrocytes exhibited<br />

a spindle-like morphology characteristic of a more fibroblastlike<br />

phenotype compared to the more rounded chondrocyte-like<br />

appearance evident in CE culture. RNA-level expression of collagen<br />

type II, aggrecan and COMP were consistently upregulated in CE<br />

versus SD culture while RNA-level expression of collagen type I and<br />

protein-level expression of α-SMA were downregulated, indicative of<br />

less dedifferentiation. Consistent with improved preservation of the<br />

chondrocyte phenotype and associated reduced proliferation rates,<br />

fewer chondrocytes were obtained from CE culture at the end of<br />

each generation. Nevertheless, ample numbers of chondrocytes can<br />

still be generated in CE culture for clinical application in cell-based<br />

therapies.<br />

Chondrocytes from CE culture were superior to those from SD<br />

culture with respect to their ability to redifferentiate toward a mature<br />

chondrocyte phenotype. Pellets from SD culture readily attached to<br />

culture surfaces and exhibited dramatic outgrowth of chondrocytes,<br />

indicating they had transitioned to a more fibroblastic cell type.<br />

In contrast, pellets from CE culture were better able to generate<br />

neotissue with more nonadhesive, cartilage-like characteristics.<br />

Both SD and CE pellets initially were able to produce GAG and<br />

collagen type II, however only CE pellets maintained this production<br />

through three generations.<br />

The only differences between CE and static silicone (SS) cultures<br />

were the application of mechanical surface expansion in CE culture,<br />

compensated by more frequent enzymatic passaging in SS culture. In<br />

two generations, SS cultures were passaged five times compared to<br />

once in CE cultures, and CE cultures exhibited significantly superior<br />

retention of chondrogenic phenotype versus SS cultures, and also<br />

superior redifferentiation capacity for production of cartilage-like<br />

neotissue. The mechanical expansion aspect of CE culture and its<br />

reduction of the need for passaging therefore appears to specifically<br />

contribute to enhancement of the phenotype of chondrocytes<br />

cultured for cell-based therapies.<br />

A factor by which CE culture inhibits dedifferentiation may be<br />

associated with reduced passaging and limited exposure to<br />

degradative enzymes. Considering that passaging involves repeated<br />

nonspecific degradation of cell surface proteins and receptors and<br />

the frequent need for wholesale re-establishment of cell-surface<br />

attachments, this is perhaps not surprising. Minimization of the<br />

need for passaging also has important practical advantages: more<br />

easily automated methods reduce the need for human intervention<br />

and handling of cells. These features likely decrease the risk of error<br />

and bacterial contamination over long-term cultures.<br />

Physical factors contributing to chondrocyte phenotype preservation<br />

in CE culture may also include mechanical signalling. Dynamic strain<br />

or compression can have positive effects on the phenotype of<br />

cultured chondrocytes [13, 14], particularly regarding proteoglycan<br />

synthesis [15, 16]. In contrast to situations involving oscillatory<br />

dynamic strain, CE culture chondrocytes experience very slow<br />

but very high amplitude (over 600%) stretch applied steadily and<br />

monotonically over the course of several days. Further studies of<br />

the mechanotransduction pathways active during slow, continuous<br />

strain may therefore yield insights into how CE culture preserves<br />

chondrogenic phenotype compared to conventional passaging.<br />

In addition, opportunities remain for optimization of CE culture by<br />

superimposed dynamic stimulation of cells during growth.<br />

The cell signalling mechanisms involved in more advanced<br />

chondrocyte dedifferentiation downstream of increased enzymemediated<br />

passaging remain unclear, but several candidate<br />

mechanisms have been identified. Interleukin-1 (IL-1) production<br />

increases with chondrocyte passaging [5], and IL-1 signalling is<br />

directly involved in chondrocyte dedifferentiation [17]. Other studies<br />

have indicated that passaged and dedifferentiated chondrocytes<br />

display degradation of the receptor of hyaluronic acid, CD44 [18].<br />

Disruption of CD44 signalling in chondrocytes results in receptor<br />

cleavage, decreased expression of chondrogenic genes, and<br />

decreased production of sulphated-GAG [19]. IL-1 activity and CD44<br />

expression may therefore participate in the mechanisms by which CE<br />

culture improves maintenance of cartilage phenotype.<br />

Continuous expansion culture represents a significant departure<br />

from standard methods used for chondrocyte expansion. Given the<br />

economic and therapeutic importance of the final product, these<br />

fundamental changes to decades-old methods of cell culture may<br />

nevertheless be warranted.<br />

References:<br />

1. Batty L et al. ANZ J Surg 2011;81:18-25.<br />

2. Brittberg M et al. N Engl J Med 1994;331:889-95.<br />

3. Kolettas E et al. Rheumatology (Oxford) 2001;40:1146-56.<br />

4. Yu H et al. Biomaterials 1997;18:1425-31.<br />

5. Lin Z et al. J Orthop Res 2008;26:1230-7.<br />

6. Darling EM and Athanasiou KA. J Orthop Res 2005;23:425-32.<br />

7. Majd H et al. Stem Cells 2009;27:200-9.<br />

8. Majd H et al. Methods Mol Biol 2011;698:175-88.<br />

9. Barbero A and Martin I. Methods Mol Med 2007;140:237-47.<br />

10. Bosnakovski D et al. Biotechnol Bioeng 2006;93:1152-63.<br />

11. Bernstein P et al. Osteoarthritis <strong>Cartilage</strong> 2010;18:1596-607.<br />

12. Zhang Z et al. J Anat 2004;205:229-37.


13. Wong M et al. Bone 2003;33:685-93.<br />

14. Holmvall K et al. Exp Cell Res 1995;221:496-503.<br />

15. Chai DH et al. Osteoarthritis <strong>Cartilage</strong> 2010;18:249-56.<br />

16. Villanueva I et al. Acta Biomater 2009;5:2832-46.<br />

17. Hong EH et al. J Biol Chem 2011.<br />

18. Chow G et al. Arthritis Rheum 1998;41:1411-9.<br />

19. Takahashi N et al. Arthritis Rheum 2010;62:1338-48.<br />

Acknowledgments:<br />

Supported by the Collaborative Health Research Program (CIHR/<br />

NSERC) grant #1004005 to TMQ and BH, and Natural Sciences and<br />

Engineering Research Council (NSERC) Discovery Grant #342320-07<br />

to TMQ, a <strong>Canada</strong> Research Chair in Soft Tissue Biophysics to TMQ,<br />

and the Canadian Institutes of Health Research grant #210820 to BH.<br />

We also thank Ghazaleh Khayat and Sidharth Chaudhry for technical<br />

assistance.<br />

8.2.3<br />

2D and 3D cultures of chondrocytes<br />

T.J. Klein<br />

Brisbane/Australia<br />

Introduction: Chondrocytes normally reside in relatively low density in<br />

a dense network of extracellular matrix. The resident chondrocytes are<br />

responsible for growth, maturation, and remodelling of the articular<br />

cartilage to ensure its function in a mechanically challenging environment.<br />

Chondrocytes have a long lifespan and proliferation is normally not<br />

appreciable. Further, this thick tissue is not vascularised or innervated.<br />

As a consequence of these factors, cartilage does not regenerate when<br />

damaged, leading eventually to large-scale degeneration, in osteoarthritis.<br />

As osteoarthritis is one of the major causes of disability and loss of quality<br />

of life, understanding how the chondrocytes work, and whether they can<br />

be coaxed to form a more robust repair tissue has been and continues to<br />

be a major research interest.<br />

To probe the behaviour of chondrocytes in detail, a number of in vitro<br />

model systems have been developed. Pioneering work in chondrocyte<br />

culture was carried out by Holtzer, et al., in a series of publications starting<br />

in 1960 [1]. In these studies, chondrocytes were enzymatically removed<br />

from their matrix and plated onto two-dimensional surfaces or on fibrin<br />

clots as a differentiation model. Through these works, it became apparent<br />

that chondrocytes lose their differentiation status (de-differentiate) in 2D<br />

culture, and also their differentiation capacity after such culture. Further<br />

studies showed that once chondrocytes reach a high enough density,<br />

they can re-differentiation, marked by an increase in glycosaminoglycan<br />

accumulation and synthesis rates [2]. In a seminal paper by Benya and<br />

Shaffer, it was shown that 2D-cultured chondrocytes can revert back to<br />

the original phenotype when embedded in a 3D (agarose) matrix [3]. This<br />

has led to many advances in understanding of chondrocyte biology and<br />

also in the development of biomaterials for chondrocyte culture. One key<br />

aspect of the 3D culture system is the accumulation of extracellular matrix.<br />

This has led to the development of tissue engineering-based therapies<br />

for treatment of patients with cartilage defects [4 5], which is the ultimate<br />

utilitarian goal for chondrocyte research.<br />

It has become apparent through decades of research that the natural<br />

microenvironment, or niche, is critical for maintaining the native phenotype<br />

of cells, including stem cells and differentiated cells such as chondrocytes.<br />

Thus, recapitulating this niche in vitro is an important aim [6]. The<br />

chondrocyte niche includes a low oxygen tension, a range of physical<br />

stresses, growth factors, enzymes and cytokines, and a surrounding of<br />

extracellular matrix. Considering the complexity of the niche, it is unlikely<br />

that cell biologists, material scientists, tissue engineers, etc., will able to<br />

incorporate all of these factors. Therefore, simplified model systems that<br />

take into account the critical parts of the niche have been developed. This<br />

paper will review some of the current 2D and 3D chondrocyte culture<br />

systems.<br />

Content: 2-dimensional Cultures:<br />

2-dimensional (monolayer) cultures are the simplest, and most pervasive<br />

throughout the cell culture community. They provide excellent opportunities<br />

Extended Abstracts 131<br />

for imaging, controlling oxygen and other culture media conditions,<br />

and harvesting cells for biochemical analyses. They also allow for major<br />

expansion of cell numbers, which can be needed for clinical use and also<br />

for experimental studies. However, there are some severe limitations, most<br />

notably in the dedifferentiation of chondrocytes during their expansion [7].<br />

This phenomenon has been repeatedly documented on a gene and protein<br />

basis. Thus, studies involving 2D cultured chondrocytes must always keep<br />

in mind that the cells are no longer behaving as they would in the cartilage,<br />

and therefore the conclusions drawn by changing experimental conditions<br />

may not be valid in vivo.<br />

There are options for controlling the niche within a 2D system. In one<br />

instance, the material properties of the surface can be varied by either<br />

using another material or functionalizing the base material with synthetic<br />

or biological ligands. This has been shown to have some positive effects<br />

through changing the binding mechanisms [8]. Another option is to<br />

change the mechanical properties of the cell substrate. It is clear from<br />

work with MSCs, especially, that the stiffness of the substrate can affect<br />

the cell differentiation status [9]. A third option is to vary the cell seeding<br />

density. High density (e.g. >= 100,000 cells/cm^2) limits proliferation<br />

and results in a more chondrocyte-like cell. The microenvironment can<br />

also be modified by inclusion of additional cell types, which could include<br />

mesenchymal stem cells, or cells from a specific zone of cartilage [10].<br />

Despite advances in functionalisation of 2D surfaces, they are inherently<br />

limited by their 2D nature. Irrespective of surface treatments, only part of<br />

the cell will be in contact with the functionalized surface, leaving the rest in<br />

an unnatural state that does not favor a round chondrocyte shape.<br />

3-dimensional Cultures:<br />

3-dimensional cultures offer the most options for developing an appropriate<br />

chondrocyte niche, and are needed for advancing cartilage repair methods.<br />

The simplest and oldest method is the micromass pellet, where a large<br />

number of chondrocytes are centrifuged and allowed to interact and form<br />

a small neo-tissue. This method has proven useful for determining the<br />

chondrogenic potential of different cell types, as well as the influence of<br />

different media additives on cartilage formation. Taken one step further,<br />

cells can be seeded onto microporous membranes to form tissue disks or<br />

other shapes[11], which could be potentially useful in vivo. This has been<br />

shown to be useful in generating tissues with depth-dependent properties<br />

that are functionally important in the natural cartilage [12 13]. One asset of<br />

this method is that there is no exogenous biomaterial, so degradation and<br />

other common issues with scaffold-based approaches are avoided.<br />

The most versatile and rapid approach is the use of hydrogels. Hydrogels<br />

have been used to encapsulate cells for several decades [2 3 14], and<br />

more and more hydrogels incorporating different functionalities continue<br />

to emerge. Interestingly, still many of the best results in terms of matrix<br />

accumulation come from the use of simple polysaccharide gels (e.g.<br />

alginate), in comparison to more complex degradable synthetic gels<br />

[15]; it remains to be seen why this is. Alginate-agarose [4] and collagen<br />

gels are already in clinical use [5], and fibrin gel is commonly used as a<br />

sealant in clinical application, where it likely plays a larger role than a<br />

simple glue. Synthetic hydrogels, or those combining synthetic and<br />

natural components have great potential for both understanding the best<br />

conditions for chondrogenesis, but also the application in large clinical<br />

defects. Over the past several years, photo-sensitive hydrogels have been<br />

introduced as a simple, reproducible, and fast way to generate 3D neotissues<br />

and apply them in a minimally invasive manner [16]. Incorporation<br />

of extracellular matrix components [17 18], and/or selectively releasable<br />

adhesion motifs appears to be useful for improving the cartilage formation<br />

in these photocrosslinkable gels [19]. There currently is a large toolbox<br />

of techniques we can use to customize our hydrogel niche. Additive<br />

manufacturing techniques can further be used for fabrication of specific<br />

sizes and shapes to fit defects or interaction between specific cell types in<br />

defined geometries and biomaterials [20].<br />

What is the best model?<br />

As models become sophisticated, we must constantly question the value<br />

of the current and newly developed models. What is the benefit of each<br />

model? How will the new model advance our understanding of chondrocyte<br />

biology, cartilage formation, and/or cartilage repair? There can be different<br />

best models for different research questions, and our ever-increasing<br />

knowledge of cells, materials, and their interactions will facilitate model<br />

improvement. Finally, we must keep in mind the clinical aim to ensure that<br />

these models are translatable to the clinic.<br />

References:<br />

1. Holtzer H, Abbott J, Lash J, Holtzer S. The Loss of Phenotypic Traits by<br />

Differentiated Cells in Vitro, I. Dedifferentiation of <strong>Cartilage</strong> Cells. Proc


132<br />

Natl Acad Sci U S A 1960;46(12):1533-42.<br />

Extended Abstracts<br />

2. Abbott J, Holtzer H. The loss of phenotypic traits by differentiated<br />

cells.<br />

3. The reversible behavior of chondrocytes in primary cultures. J Cell<br />

Biol 1966;28(3):473-87. 3. Benya PD, Shaffer JD. Dedifferentiated<br />

chondrocytes reexpress the differentiated collagen phenotype when<br />

cultured in agarose gels. Cell 1982;30(1):215-24.<br />

4. Selmi TA, Verdonk P, Chambat P, Dubrana F, Potel JF, Barnouin L, et<br />

al. Autologous chondrocyte implantation in a novel alginate-agarose<br />

hydrogel: outcome at two years. J Bone Joint Surg Br 2008;90(5):597-<br />

604.<br />

5. Schneider U, Rackwitz L, Andereya S, Siebenlist S, Fensky F, Reichert J,<br />

et al. A prospective multicenter study on the outcome of type I collagen<br />

hydrogel-based autologous chondrocyte implantation (CaReS) for<br />

the repair of articular cartilage defects in the knee. Am J Sports Med<br />

2011;39(12):2558-65.<br />

6. Lutolf MP, Gilbert PM, Blau HM. Designing materials to direct stem-cell<br />

fate. Nature 2009;462(7272):433-41.<br />

7. Darling EM, Athanasiou KA. Rapid phenotypic changes in passaged<br />

articular chondrocyte subpopulations. J Orthop Res 2005;23(2):425-32.<br />

8. Mahmood TA, Miot S, Frank O, Martin I, Riesle J, Langer R, et al.<br />

Modulation of chondrocyte phenotype for tissue engineering by<br />

designing the biologic-polymer carrier interface. Biomacromolecules<br />

2006;7(11):3012-8.<br />

9. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem<br />

cell lineage specification. Cell 2006;126(4):677-89.<br />

10. Blewis ME, Schumacher BL, Klein TJ, Schmidt TA, Voegtline MS, Sah<br />

RL. Microenvironment regulation of PRG4 phenotype of chondrocytes. J<br />

Orthop Res 2007;25(5):685-95.<br />

11. Han E, Bae WC, Hsieh-Bonassera ND, Wong VW, Schumacher BL,<br />

Gortz S, et al. Shaped, stratified, scaffold-free grafts for articular cartilage<br />

defects. Clin Orthop Relat Res 2008;466(8):1912-20.<br />

12. Sun Y, Kandel R. Deep zone articular chondrocytes in vitro express<br />

genes that show specific changes with mineralization. J Bone Miner Res<br />

1999;14(11):1916-25.<br />

13. Klein TJ, Schumacher BL, Schmidt TA, Li KW, Voegtline MS, Masuda K,<br />

et al. Tissue engineering of stratified articular cartilage from chondrocyte<br />

subpopulations. Osteoarthritis <strong>Cartilage</strong> 2003;11(8):595-602.<br />

14. Guo JF, Jourdian GW, MacCallum DK. Culture and growth characteristics<br />

of chondrocytes encapsulated in alginate beads. Connect Tissue Res<br />

1989;19(2-4):277-97.<br />

15. Klein TJ, Rizzi SC, Schrobback K, Reichert JC, Jeon JE, Crawford RW,<br />

et al. Long-term effects of hydrogel properties on human chondrocyte<br />

behavior. Soft Matter 2010;6:5175-5183.<br />

16. Elisseeff J, Anseth K, Sims D, McIntosh W, Randolph M, Langer R.<br />

Transdermal photopolymerization for minimally invasive implantation.<br />

Proc Natl Acad Sci U S A 1999;96(6):3104-7.<br />

17. Bryant SJ, Arthur JA, Anseth KS. Incorporation of tissue-specific<br />

molecules alters chondrocyte metabolism and gene expression in<br />

photocrosslinked hydrogels. Acta Biomater 2005;1(2):243-52.<br />

18. Salinas CN, Anseth KS. Decorin moieties tethered into PEG networks<br />

induce chondrogenesis of human mesenchymal stem cells. J Biomed<br />

Mater Res A 2009;90(2):456-64.<br />

19. Kloxin AM, Kasko AM, Salinas CN, Anseth KS. Photodegradable<br />

hydrogels for dynamic tuning of physical and chemical properties.<br />

Science 2009;324(5923):59-63.<br />

20. Melchels FPW, Domingos MAN, Klein TJ, Malda J, Bartolo PJ,<br />

Hutmacher DW. Additive manufacturing of tissues and organs. Progress<br />

in Polymer Science 2011;Accepted 18/11/2011.<br />

Acknowledgments:<br />

I would like to acknowledge the Australian Research Council for funding,<br />

and members of the <strong>Cartilage</strong> Regeneration Laboratory for their hard<br />

work and support.<br />

8.3.1<br />

Intervertebral Disc Tissue Engineering: Is it ready for weight<br />

bearing?<br />

R. Kandel<br />

Toronto/<strong>Canada</strong><br />

Introduction: Degeneration of the intervertebral disc, which is<br />

composed of the annulus fibrosus (AF), nucleus pulposus (NP),<br />

and cartilage endplates, causes loss of disc function and can be<br />

associated with low back pain (1,2). Approximately 1 in 50 individuals<br />

become disabled by intervertebral disc degeneration; the annual<br />

total costs, which in 2004 in the United States alone, was estimated<br />

at over $100 billion. There is a growing consensus that the surgical<br />

treatments used currently for disc degeneration are not effective<br />

and cannot be further optimized; thus there is great interest in<br />

developing a biological therapy to treat the chronic pain that arises<br />

from disc degeneration (1,3).<br />

Content: Biological repair or replacement results in tissue that<br />

can remodel and respond to load, an outcome not achievable by<br />

current surgical therapies. Many different approaches to disc repair<br />

are being investigated. However a recent study raised concern<br />

about intradiscal injections and their potential to contribute to<br />

disc degeneration so a particular interest has developed in the use<br />

of regenerative medicine to generate an intervertebral disc that<br />

structurally and functionally resembles the in vivo disc (4). To date,<br />

we have advanced significantly towards accomplishing this goal.<br />

Studies have described the formation of the nucleus pulposus tissue.<br />

However we do not yet know whether we require the presence of<br />

both nucleus pulposus cells and notochordal cells in this tissue or<br />

whether generating tissue by just nucleus pulposus cells alone will<br />

be sufficient to form tissue of appropriate quality to withstand weight<br />

bearing over the long term. Investigation into the role of notochordal<br />

cells in nucleus tissue is a subject of intense investigation currently<br />

and much attention is directed towards trying to develop methods<br />

to obtain pure populations of these cells. In contrast engineering<br />

the annulus fibrosus has been more problematic given its complex<br />

structure. The annulus fibrosus surrounds the nucleus pulposus<br />

and has a cross-ply laminate structure consisting of between 10-<br />

25 lamellae, each composed of collagen fibres oriented parallel to<br />

each other and about 65o from the vertical, so every second lamella<br />

has the same orientation. Various approaches have been utilized to<br />

generate annulus fibrosus tissue ranging from the use of allograft<br />

tissue to in vitro generated annulus fibrosus tissue. (5,6). Annulus<br />

fibrosus cells are very responsive to their microenvironment. Our<br />

recent studies have shown that how a scaffold is pre-treated, such as<br />

coating it with fibronectin versus collagen or the amount of tension<br />

the cells experience can affect annulus cell and collagen alignment<br />

and thus tissue formation. Clearly creating annulus fibrosus tissue<br />

will require more study. An additional issue to be considered is<br />

ensuring that all the necessary components of the disc are replaced.<br />

It likely will not be sufficient to replace only the annulus fibrosus<br />

and nucleus pulposus as studies have shown that an intact cartilage<br />

endplate may be necessary for maintenance of the nucleus pulposus<br />

tissue. Co-culture of in vitro-formed nucleus pulposus tissue with in<br />

vitro-formed cartilage resulted in increased aggrecan and collagen<br />

gene expression compared with that in NP tissue grown alone. In<br />

addition there was reduced expression of degradative enzymes,<br />

MMP-3, MMP-13, and ADAMTS-5. Expression of genes for tumor<br />

necrosis factor α (TNFα) and TACE in nucleus pulposus cells was<br />

higher when grown in the absence of cartilage and corresponded<br />

with increased TNFα protein levels. This suggests that chondrocytes<br />

may secrete a factor(s) that inhibits TNFα production and positively<br />

enhances tissue maintenance. Loss of the cartilage endplate could<br />

be a potential mechanism explaining how changes in this tissue<br />

may contribute to the development of NP degenerative changes and<br />

suggesting that part of the disc replacement strategy must include<br />

repairing the cartilage endplate. Identifying a cell source to generate<br />

the different disc tissues is also another issue. Use of autogenous<br />

cells may be a problematic as studies in nucleus pulposus tissue<br />

engineering demonstrate that although nucleus pulposus tissue can<br />

be formed by the nucleus cells obtained from older animals (cows),<br />

they formed less tissue compared to cells obtained from younger<br />

animals (younger than adolescents). Interestingly the older cells had<br />

lower constitutive gene expression of collagen type II and aggrecan<br />

whereas collagen type I and link protein levels were similar to those<br />

of the younger cells. Metalloprotease (MMP) 13 gene and protein<br />

expression increased with age. There was no change in the levels of<br />

gene expression of MMP 2 and TIMP 1, 2, or 3 with age. Thus cells<br />

obtained from nucleus pulposus tissue harvested from younger or<br />

mature animals showed both genotypic and phenotypic differences<br />

in vitro. Although these changes may be circumvented by gene<br />

therapy the use of other sources of cells, such as stem cells, are


eing explored. Until we have markers that are definitively specific<br />

to the cells of the different disc tissues it will not be possible to use<br />

stem cells. Finally the method to implant the biological replacement<br />

will have to be developed. The surgical approach will have to be<br />

optimized and then it will be necessary to ensure that the implanted<br />

disc will integrate to bone in the presence of weight bearing. Clearly<br />

although a biological disc replacement is not yet available we appear<br />

to be making great progress, and in some ways moving faster than<br />

that of cartilage tissue engineering. Furthermore our attempts to<br />

create a disc have provided insights as to the biology of the disc<br />

and some of the requirements that may be critical to successful<br />

generation of an intervertebral disc. In the interim to ensure rapid<br />

translation into clinical practice once a biological disc replacement<br />

has been developed we should also be turning our attention to<br />

determining who would be a candidate for such a treatment, what<br />

is the optimal rehabilitation regimen, and how to define a successful<br />

outcome for those receiving a disc replacement.<br />

References:<br />

1) Raj PP. Pain Pract. 2008; 8(1):18- 24.<br />

2) Cheung K et al. The Spine J. 2010;10, 958-60.<br />

3) Kandel et al. Eur Spine J Suppl 2008; 4:480-91.<br />

4) Carragee EJ, Don AS, Hurwitz EL, Cuellar JM, Carrino JA, Herzog R.<br />

Spine (Phila Pa 1976). 2009;34:2338-45.<br />

5) Bowles RD et al. PNAS 2011;108:13106-11<br />

6) Luk KD and Ruan RK. Eur Spine J. 2008 Suppl 4:504-10.<br />

Acknowledgments:<br />

This work was supported by a grant from CIHR # MOP 114991.<br />

8.3.2<br />

Current clinical treatment strategies and future concepts<br />

S. Daisuke<br />

Kanagawa/Japan<br />

Introduction: The intervertebral disc (IVD) functions as an essential<br />

load absorber between all vertebrae by allowing bending, flexion, and<br />

torsion of the spine. IVD degeneration is a cell-mediated response to<br />

progressive structural failure and causes instability of the vertebral<br />

motion segments that are responsible for neural compressive<br />

manifestations and low back pain. Prolonged segmental instability<br />

eventually leads to deformity of the spine and many clinical<br />

problems. Current clinical concept, their limitations, and progress of<br />

future treatment investigations will be presented.<br />

Content: Treatment usually begins with non-operative modalities,<br />

such as physical therapy or methods for core strengthening;<br />

symptomatic medical treatment with non-steroidal anti-inflammatory<br />

medications is a further common method to reduce pain. Surgical<br />

methods are considered if conservative therapy fails. Although only<br />

a small percentage of patients with disc disorders finally will undergo<br />

surgery, spinal surgery has been one of the fastest growing<br />

disciplines in the musculoskeletal field in recent years. Nevertheless,<br />

current treatment options are still a matter of controversial<br />

discussion. The standard surgical intervention has been spinal<br />

arthrodesis with the aim to immobilize the spinal segment, preferably<br />

by bony fusion. The aim is to cease mechanical cues and inflammatory<br />

processes causing pain and disability. However, compared to<br />

conservative treatment only small benefits could be achieved, as<br />

assessed in several clinical studies. In addition, the occurrence of<br />

adjacent segment disease should not be underestimated. With the<br />

goal to better preserve the biomechanics of the spine, total disc<br />

replacement has been introduced and has become part of surgical<br />

routine in recent years. Since this technology has not been able to<br />

demonstrate any significant advantage to the standard spinal<br />

arthrodesis and in contrast has faced considerable complication<br />

rates, it has been critically debated. Other newer technologies<br />

include nucleus pulposus replacement or dynamic stabilization<br />

methods. Long-term clinical outcomes that may disclose any<br />

potential benefit of these new methods are not yet available;<br />

however the long-term success rates of all these procedures are<br />

Extended Abstracts 133<br />

estimated to be generally similar. Although in the main satisfying<br />

results can be achieved, all these treatment methods attempt to<br />

reduce pain but cannot repair the degenerated disc. In particular,<br />

they hardly can restore normal spine biomechanics and prevent<br />

degeneration of adjacent tissues. Therefore new treatments are<br />

under development with the aim to restore disc height and<br />

biomechanical function. The objective of such new regenerative<br />

strategies is to generate healthy disc tissue or functional<br />

replacements that decelerate or reverse painful degeneration<br />

processes. A number of biological approaches such as molecular,<br />

gene, and cell based therapies have been investigated and have<br />

shown promising results in both in vitro and in vivo studies. Recently,<br />

cell transplantation based on the supplementation of matrixproducing<br />

cells in an attempt to correct the decrease of matrix<br />

components, primarily proteoglycan and collagen, a major factor in<br />

disc degeneration has been under clinical trial. The concept goes<br />

back to 1996, when Mochida et al. demonstrated the importance of<br />

preserving NP elements for preventing the acceleration of disc<br />

degeneration following discectomy. This clinical study opened a new<br />

area of research into replacement of the cells lost by pathological<br />

manifestations or surgical intervention, potentially retarding the<br />

progression of disc degeneration. To test this hypothesis, an animal<br />

model study, performed by Nishimura and Mochida, demonstrated<br />

that reinsertion of autologous fresh or cryopreserved NP cells slowed<br />

degeneration in the rat IVD. Numerous subsequent studies have<br />

reported the efficacy of cell transplantation therapy using various<br />

animal models and donor cell types. The author’s lab has studied the<br />

potential of MSCs as an alternative cell source. They transplanted<br />

autologous MSCs tagged with the gene for green fluorescent protein<br />

(GFP) in rabbit disc degeneration model created by nucleus<br />

aspiration, and followed the GFP-labelled cells for a period of 48<br />

weeks, tracking the effects using MRI and radiography. They also<br />

used immunohistochemistry for chondroitin sulphate, keratin<br />

sulphate, collagen types I, II, and IV, HIF-1alpha and beta, HIF-2alpha<br />

and beta, glucose transporters GLUT-1 and GLUT-3, and MMP-2, and<br />

applied RT-PCR to assess expression of the genes for aggrecan,<br />

versican, collagen types I and II, IL-1b, IL-6, TNF-alpha, MMP-9, and<br />

MMP-13. MRI and radiographic results confirmed the regenerative<br />

effects of the procedure. GFP-positive cells were detected in the<br />

nucleus throughout the time course at proportions rising from 21% ±<br />

6% at 2 weeks to 55% ± 8% at 48 weeks, which demonstrated the<br />

survival and proliferation of MSCs. Immunohistochemistry showed<br />

positive staining for all proteoglycan epitopes and type II collagen in<br />

some of the GFP-positive cells. MSCs produced HIF-1alpha, MMP-2,<br />

and GLUT-3 with phenotypic activity comparable to NP cells. The RT-<br />

PCR demonstrated significant restoration of aggrecan, versican, and<br />

type II collagen gene expression, and significant suppression of TNFalpha<br />

and IL-1b expression in the transplantation group. Thus, MSCs<br />

transplanted into degenerating discs in vivo can survive, proliferate,<br />

and differentiate into cells expressing the phenotype of NP cells with<br />

suppression of inflammatory genes. In order to achieve a maximal<br />

effect in stem cell therapy for IVD disease, knowledge on cells,<br />

extracellular matrix and the microenvironment of the native disc<br />

must be extensively studied. Fully committed adult cells are cells<br />

that actively function as main cell population eventually going into<br />

apoptosis. Other than committed adult cells are the tissue specific<br />

somatic progenitor/stem cells which are an undifferentiated cells<br />

found among differentiated cells in tissue or organ that can renew<br />

itself. Stem cells are distributed around the body in various other<br />

‘niches’. Evidence of existence of the small stem cell population has<br />

not been well studied in the IVD. In order to identify the somatic stem<br />

cell population that reside in the IVD, “stem cell markers” were<br />

immunohistochemically analyzed in rat, beagle and human IVDs.<br />

The immunohistochemical analysis in rat IVD specimens revealed<br />

that most of these classical stem cell markers may not be useful in<br />

identifying endogenous stem cells in the disc. Especially, positivity<br />

of the hall mark markers CD90, CD105 and CD 166 was 85, 92 and 98<br />

percent (n=5). This was far different from the standard characteristics<br />

of stem cells which reside in small population with inactive cell<br />

viability at rest and after initiation, shows high self-renewal and<br />

proliferative ability with multi-potent differentiation. Regarding<br />

other markers, no significant expression (0-10percent) was detected<br />

for CD56, CD120a CD124 MHC class I, etc. in the rat disc, which may<br />

remain these markers as potential candidates. To note, negative<br />

expression in some of these markers may be a result of non-cross<br />

reactivity of the antibody to the rat. Result of human disc specimens<br />

showed that CD56, 90 105,166 was expressed in some young disc<br />

cells in sparse areas, such as NP cells forming clusters but in old<br />

aged disc where cells are isolated in single cells, merely none of<br />

these markers were positive in most discs. To overcome the lack of<br />

stem cell population indicated by the results, identification of<br />

progenitor cells by marker analysis was continued through FACS<br />

analysis. By plotting the positivity of these markers through serial<br />

culture periods, we are able to detect cell markers which correlate


134<br />

Extended Abstracts<br />

with cell proliferation. As mentioned, stem/progenitor cell<br />

populations show little or decreased cell number at primary culture<br />

and give rise to rapid proliferation several weeks after, meaning<br />

potential for high self-renewal. We also found that by culturing<br />

beagle NP cells in gels, several different colonies can be induced.<br />

These included sphere shaped colony and few adherent colonies.<br />

Number of adherent colonies increased with time with no change<br />

with culture period. On the other hand, change in number of sphere<br />

colonies formed showed correlation with proliferation curve pattern<br />

of progenitor cells. These findings provide useful information on the<br />

progenitor cell population in the IVD. Collectively, new techniques<br />

based on rigorous animal studies and clinical trials are on the way to<br />

be brought into future treatment of the intervertebral disc<br />

degeneration. Uncovering of cells and their microenvironment is<br />

needed to be investigated in parallel for obtaining maximal efficacy<br />

and safety.<br />

References:<br />

1. Alini M, Sakai D, Eglin D, et al. Cells and Biomaterials for<br />

Intervertebral Disc Rgeneration. Synthesis Lectures on Tissue<br />

Engineering. Morgan and Claypool Publishers. 2010; 1-104. 2. Sakai<br />

D. Stem cell regeneration of the intervertebral disk. Orthop Clin<br />

North Am. 2011;42(4):555-62.<br />

Acknowledgments:<br />

This work was supported in part by a Grant-in-Aid for Scientific<br />

Research and a Grant of The Science Frontier Program from the<br />

Ministry of Education, Culture, Sports, Science and Technology of<br />

Japan and a grant from AO Spine <strong>International</strong>.<br />

8.3.3<br />

Materials for intervertebral disc tissue engineering<br />

R. Mauck, M.B. Fisher, D.M. Elliott<br />

Philadelphia/United States of America<br />

Introduction: Intervertebral disc (IVD) degeneration is linked to<br />

multiple causal factors, including age and genetics, mechanical<br />

fatigue, injury through trauma, continued exposure to vibrational<br />

loading, and mechanical overload. Disc degeneration progresses<br />

over time and impacts a large percentage of the world population.<br />

Low back pain in the U.S. is widespread in the general population,<br />

and limits activity in more than 50% of the population over the age<br />

of 55 [1]. Lumbar disc degeneration has been strongly implicated<br />

as a causative factor in low back pain [2], and neither conservative<br />

treatments (stretching and exercise) nor surgical options (discectomy,<br />

fusion, and arthroplasty) restore disc structure or mechanical<br />

function. Indeed, fusion permanently locks adjacent vertebral bodies<br />

in place, while disc arthroplasty is relatively new to clinical practice<br />

and will likely suffer from the same problems as traditional implant<br />

materials <strong>–</strong> wear and the need for eventual replacement. As such,<br />

biomaterial and biologic interventions to regenerate a degenerate<br />

IVD would have a distinct competitive advantage over current clinical<br />

procedures, while also potentially reducing painful conditions and<br />

delaying or preventing the need for spinal fusion or replacement.<br />

This talk will focus on the critical structure-function relationships of<br />

the native disc, with a particular focus on the annulus fibrosus, and<br />

highlight emerging biomaterials and cell-based methods designed<br />

to foster regeneration and/or produce functional analogues for the<br />

repair and/or replacement of this unique load-bearing structure.<br />

Content: The IVD is composed of the nucleus pulposus (NP), a<br />

hydrated, gelatinous structure, and the annulus fibrosus (AF), a<br />

multi-lamellar fibrocartilage that surrounds the NP between the<br />

vertebral bodies. The NP is structurally and mechanically isotropic<br />

and contains a network of type II collagen interspersed with<br />

proteoglycans, resulting in high water content within the tissue.<br />

Conversely, the fibrocartilaginous AF has a high degree of structural<br />

organization over multiple length scales: aligned bundles of collagen<br />

fibers reside within each lamella and the direction of alignment<br />

alternates from one lamella to the next by 30° above and below the<br />

transverse plane [3, 4]. The resulting angle-ply laminate possesses<br />

pronounced mechanical anisotropy and nonlinearity, allowing the AF<br />

to support tension, shear, compression, and torsion [5]. The NP and<br />

AF regions work in concert; when the disc is compressed, hydrostatic<br />

pressure in the NP increases, resulting in stresses along the<br />

circumferential direction of the AF, which are in turn resisted by the<br />

organized lamellar AF structure. Injury or degeneration to either<br />

portion of this composite can compromise overall disc mechanical<br />

function, and as such, methods have been devised to address failure<br />

in either or both components. While disc degeneration can originate<br />

in multiple locations, tears within the AF region are especially<br />

pernicious as they can propagate to the NP region, eventually<br />

resulting in complete tears involving leakage of the NP (disc<br />

herniation). Along with herniation, there is often a loss of annulus<br />

tissue [6, 7]. In fact, annular tears are seen in more than half of the<br />

patients in early adulthood and are found in the majority of the<br />

elderly [8]. Yet, the most common surgical treatment for disc<br />

herniation, e.g. microdiscectomy, does nothing to repair the AF,<br />

which has a limited intrinsic healing potential [9]. As such,<br />

reherniation rates following these procedures can range from 5-15%<br />

[10-13], compromising long term disc health. Given the prevalence of<br />

low back pain associated with damage or degeneration of all or part<br />

of the IVD, a host of products and devices have been developed, with<br />

some reaching the commercial market. These include products that<br />

restore pressurization in the NP region or mechanically augment the<br />

torn AF (i.e., suture, meshes, and reinforcing barriers) [7, 14]. While<br />

such approaches have shown promise in cadaveric studies, those<br />

focused on the AF do not recapitulate the structure or function of the<br />

native tissue, and thus, abnormal loading and continued degeneration<br />

may occur. Furthermore, the majority of these devices are inert,<br />

preventing integration with the native disc. Given these limitations<br />

in current commercial devices and clinical methods, a number of<br />

tissue engineering and regenerative medicine approaches have<br />

been pursued (for review, see [5]). These approaches have focused<br />

on the AF, the NP, or a combination of the two. In most cases, the<br />

disc is separated into an NP region containing chondrocyte-like cells<br />

and an AF region containing fibroblast-like cells. These cellular subpopulations<br />

have been seeded in a variety of tissue engineering<br />

scaffolds or hydrogels to evaluate their ability to reconstitute the<br />

histological structure and composition of native tissue [15-19].<br />

Scaffolds have included alginate and agarose hydrogels, collagen<br />

gels, collagen/glycosaminoglycan gels, collagen/hyaluronic acid<br />

scaffolds and collagen sponges, to name but a few [20-25]. These<br />

and other studies have established that disc cells can be maintained<br />

in three dimensional culture, that they produce plentiful ECM with<br />

time, and that certain mechanical, biological, and structural cues<br />

may foster construct maturation (i.e., a hydrogel for the NP and a<br />

porous/fibrous scaffold for the AF). More recently, multi-potential<br />

mesenchymal stem cells (MSCs) have been employed in a number of<br />

these scaffolding systems, with evidence of disc-like matrix formation<br />

in these 3D biomaterial contexts [26]. While these studies highlight<br />

the potential to generate disc-like neo-tissue by combining cells and<br />

scaffolds, for many years, little consideration was given to the<br />

mechanical properties and function of these engineered materials<br />

within a load bearing environment. The mechanical requirements for<br />

an engineered disc construct will depend largely on its intended<br />

location and use. An AF ‘patch’ would be expected to integrate across<br />

an AF fissure, resist further NP herniation, and transfer tensile forces<br />

with disc compression. An NP construct would be expected to fill the<br />

central region of the disc and pressurize and engage the AF with<br />

loading. A total biologic disc replacement would be expected to carry<br />

out both of these critical functions, while also integrating seamlessly<br />

with the surrounding vertebral bodies. Depending on application,<br />

these materials or constructs might be required to match native<br />

tissue at the time of implantation (if early remobilization is expected)<br />

or could conceivably mature in place to match these properties (if a<br />

period of protection from loading were possible). In either format or<br />

application, these engineered materials would eventually be<br />

expected to withstand dynamic mechanical loading (at multiple body<br />

weights) in multiple directions, consistent with the complex and<br />

demanding loading environment of the disc with normal activities.<br />

While these requirements may seem insurmountable, the last<br />

decade has borne witness to marked progress in the area of disc<br />

tissue engineering. For example, in seminal work in the field,<br />

Bonassar and colleagues created a composite disc by encapsulating<br />

NP cells in an alginate hydrogel surrounded by an AF cell-seeded<br />

random fibrous scaffold of polylactic acid reinforced polyglycolic<br />

acid [27, 28]. After 16 weeks of subcutaneous implantation,<br />

biochemical content of these disc-like constructs approached native<br />

levels, and, importantly, compressive mechanical properties<br />

increased with time [27]. More recently, using a collagen-gel annular<br />

region along with an alginate NP region, both seeded with disc cells,<br />

this same group reported on the long term (>6 month) maintenance<br />

of disc height, reproduction of native tissue properties, and<br />

physiologic remodeling and integration of an engineered construct<br />

in a rat tail disc replacement model [29]. These promising studies<br />

highlight the potential for the engineering of a mechanically viable<br />

NP, AF, or whole disc structure. As an alternative to implantation of<br />

an immature construct (with in vivo maturation), new materials can<br />

be used to direct the formation of an organized tissue matching


native disc structure and function. For example, using aligned<br />

nanofibrous scaffolds as a starting template, we demonstrated that<br />

a single annulus layer, with physiologic fiber organization, could be<br />

produced [30]. When these oriented nanofibrous constructs were<br />

seeded with AF cells [31], constructs increased in tensile modulus<br />

with time and the orientation of collagen coincided with the scaffold<br />

fiber direction. Extending this work to consider the multi-lamellar<br />

hierarchy of the native AF, we next assembled MSC-seeded angle-ply<br />

bi-lamellar structures with a fiber orientations of ±30° in each layer<br />

[32]. By 10 weeks, angle-ply bi-layers reached a modulus very close<br />

to the modulus of the native AF (~18 MPa). When sections across<br />

two fiber planes were viewed using polarized light microscopy, a<br />

multi-scale collagen architecture mirroring the cross-ply organization<br />

of the native AF was observed: two layers of aligned, opposing<br />

collagen. Such studies provided insight into the fundamental<br />

reinforcing effect of alternating directions in angle ply laminates,<br />

and may serve as a promising tissue engineering template for AF<br />

repair. Further, these angle-ply laminate structures can be formed<br />

into disc-like angle-ply structure (DAPS) that replicate the multiscale<br />

architecture of the intervertebral disc, inclusive of a laminate<br />

AF region and a gelatinous NP region [33]. In vitro culture of such<br />

constructs showed that the mechanical characteristics of the disc<br />

under compression and torsion were qualitatively similar to native<br />

tissue, although lesser in magnitude, and increased with time in<br />

culture. Cells seeded into both AF and NP regions adopted<br />

morphologies that mirror those seen in native tissue, with rounded<br />

cells in the NP and more elongated cells in the AF regions. In the AF<br />

region, the collagenous matrix deposited followed the angle-ply<br />

configuration of the scaffold. While considerable advances have<br />

been made in disc tissue engineering, most notably the transplant of<br />

a mechanically functional disc-like analogue in vivo in a rodent<br />

model, there remain a number of hurdles to overcome before biologic<br />

disc replacement becomes a clinical reality. Advances in disc<br />

replacement in a small animal model will need to be evaluated in<br />

larger species, where diffusional and mechanical demands will be<br />

greater. Engineering for an inflamed environment, likely present in<br />

degenerating discs, will further complicate this transition, and novel<br />

materials that can address this scenario may be required. Despite<br />

these limitations, lessons learned in the pursuit of a full biologic disc<br />

replacement will likely provide nearer term solutions for instances of<br />

NP degeneration or AF fracture. Along with these applications, new<br />

and minimally surgical approaches will need to be developed to ease<br />

clinical application. These advances will provide early stage<br />

treatment options that may slow disc degeneration, until such time<br />

as biologic substitutes become a clinical reality. Despite the<br />

remaining hurdles, progress in disc tissue engineering has been<br />

rapid and is already making an impact. These advances will continue<br />

to accelerate and will one day provide a paradigm shift in the<br />

treatment of disc degeneration, providing long term and functional<br />

solutions for what is today an intractable, debilitating, and<br />

widespread condition.<br />

References:<br />

1. Katz, R.T., Impairment and disability rating in low back pain. Clin<br />

Occup Environ Med, 2006. 5(3): p. 719-40, viii.<br />

2. Albert, H.B., et al., Modic changes, possible causes and relation to<br />

low back pain. Med Hypotheses, 2008. 70(2): p. 361-8.<br />

3. Cassidy, J.J., A. Hiltner, and E. Baer, Hierarchical structure of the<br />

intervertebral disc. Connect Tissue Res, 1989. 23(1): p. 75-88.<br />

4. Marchand, F. and A.M. Ahmed, Investigation of the laminate<br />

structure of lumbar disc anulus fibrosus. Spine (Phila Pa 1976), 1990.<br />

15(5): p. 402-10.<br />

5. Nerurkar, N.L., D.M. Elliott, and R.L. Mauck, Mechanical design<br />

criteria for intervertebral disc tissue engineering. J Biomech, 2010.<br />

43(6): p. 1017-30.<br />

6. Ahlgren, B.D., et al., Effect of anular repair on the healing strength<br />

of the intervertebral disc: a sheep model. Spine (Phila Pa 1976),<br />

2000. 25(17): p. 2165-70.<br />

7. Heuer, F., et al., Biomechanical evaluation of conventional<br />

anulus fibrosus closure methods required for nucleus replacement.<br />

Laboratory investigation. J Neurosurg Spine, 2008. 9(3): p. 307-13.<br />

8. Videman, T. and M. Nurminen, The occurrence of anular tears and<br />

their relation to lifetime back pain history: a cadaveric study using<br />

barium sulfate discography. Spine (Phila Pa 1976), 2004. 29(23): p.<br />

2668-76.<br />

Extended Abstracts 135<br />

9. Fazzalari, N.L., et al., Mechanical and pathologic consequences of<br />

induced concentric anular tears in an ovine model. Spine (Phila Pa<br />

1976), 2001. 26(23): p. 2575-81.<br />

10. Veresciagina, K., B. Spakauskas, and K.V. Ambrozaitis, Clinical<br />

outcomes of patients with lumbar disc herniation, selected for onelevel<br />

open-discectomy and microdiscectomy. Eur Spine J, 2010.<br />

19(9): p. 1450-8.<br />

11. Yeung, A.T. and P.M. Tsou, Posterolateral endoscopic excision<br />

for lumbar disc herniation: Surgical technique, outcome, and<br />

complications in 307 consecutive cases. Spine (Phila Pa 1976), 2002.<br />

27(7): p. 722-31.<br />

12. Thome, C., et al., Outcome after lumbar sequestrectomy compared<br />

with microdiscectomy: a prospective randomized study. J Neurosurg<br />

Spine, 2005. 2(3): p. 271-8.<br />

13. Fakouri, B., et al., Lumbar microdiscectomy versus<br />

sequesterectomy/free fragmentectomy: a long-term (>2 y)<br />

retrospective study of the clinical outcome. J Spinal Disord Tech,<br />

2011. 24(1): p. 6-10.<br />

14. Bron, J.L., et al., <strong>Repair</strong>, regenerative and supportive therapies<br />

of the annulus fibrosus: achievements and challenges. Eur Spine J,<br />

2009. 18(3): p. 301-13.<br />

15. Chiba, K., et al., Metabolism of the extracellular matrix formed by<br />

intervertebral disc cells cultured in alginate. Spine (Phila Pa 1976),<br />

1997. 22(24): p. 2885-93.<br />

16. Gruber, H.E., et al., Three-dimensional culture of human disc cells<br />

within agarose or a collagen sponge: assessment of proteoglycan<br />

production. Biomaterials, 2006. 27(3): p. 371-6.<br />

17. Gruber, H.E., et al., Cell shape and gene expression in human<br />

intervertebral disc cells: in vitro tissue engineering studies. Biotech<br />

Histochem, 2003. 78(2): p. 109-17.<br />

18. Wang, J.Y., et al., Intervertebral disc cells exhibit differences in<br />

gene expression in alginate and monolayer culture. Spine (Phila Pa<br />

1976), 2001. 26(16): p. 1747-51; discussion 1752.<br />

19. Baer, A.E., et al., Collagen gene expression and mechanical<br />

properties of intervertebral disc cell-alginate cultures. J Orthop Res,<br />

2001. 19(1): p. 2-10.<br />

20. Sato, M., et al., An atelocollagen honeycomb-shaped scaffold<br />

with a membrane seal (ACHMS-scaffold) for the culture of annulus<br />

fibrosus cells from an intervertebral disc. J Biomed Mater Res A,<br />

2003. 64(2): p. 248-56.<br />

21. Rong, Y., et al., Proteoglycans synthesized by canine intervertebral<br />

disc cells grown in a type I collagen-glycosaminoglycan matrix.<br />

Tissue Eng, 2002. 8(6): p. 1037-47.<br />

22. Saad, L. and M. Spector, Effects of collagen type on the behavior<br />

of adult canine annulus fibrosus cells in collagen-glycosaminoglycan<br />

scaffolds. J Biomed Mater Res A, 2004. 71(2): p. 233-41.<br />

23. Alini, M., et al., The potential and limitations of a cell-seeded<br />

collagen/hyaluronan scaffold to engineer an intervertebral disc-like<br />

matrix. Spine (Phila Pa 1976), 2003. 28(5): p. 446-54; discussion<br />

453.<br />

24. Shao, X. and C.J. Hunter, Developing an alginate/chitosan hybrid<br />

fiber scaffold for annulus fibrosus cells. J Biomed Mater Res A, 2007.<br />

82(3): p. 701-10.<br />

25. Smith, L.J., et al., Nucleus pulposus cells synthesize a functional<br />

extracellular matrix and respond to inflammatory cytokine challenge<br />

following long-term agarose culture. Eur Cell Mater, 2011. 22: p. 291-<br />

301.<br />

26. Gupta, M.S., E.S. Cooper, and S.B. Nicoll, Transforming growth<br />

factor-beta 3 stimulates cartilage matrix elaboration by human<br />

marrow-derived stromal cells encapsulated in photocrosslinked<br />

carboxymethylcellulose hydrogels: potential for nucleus pulposus<br />

replacement. Tissue Eng Part A, 2011. 17(23-24): p. 2903-10.<br />

27. Mizuno, H., et al., Biomechanical and biochemical characterization<br />

of composite tissue-engineered intervertebral discs. Biomaterials,<br />

2006. 27(3): p. 362-70.


136<br />

Extended Abstracts<br />

28. Mizuno, H., et al., Tissue-engineered composites of anulus<br />

fibrosus and nucleus pulposus for intervertebral disc replacement.<br />

Spine (Phila Pa 1976), 2004. 29(12): p. 1290-7; discussion 1297-8.<br />

29. Bowles, R.D., et al., Tissue-engineered intervertebral discs<br />

produce new matrix, maintain disc height, and restore biomechanical<br />

function to the rodent spine. Proc Natl Acad Sci U S A, 2011. 108(32):<br />

p. 13106-11.<br />

30. Nerurkar, N.L., D.M. Elliott, and R.L. Mauck, Mechanics of<br />

oriented electrospun nanofibrous scaffolds for annulus fibrosus<br />

tissue engineering. J Orthop Res, 2007. 25(8): p. 1018-28.<br />

31. Nerurkar, N.L., R.L. Mauck, and D.M. Elliott, ISSLS prize winner:<br />

Integrating theoretical and experimental methods for functional<br />

tissue engineering of the annulus fibrosus. Spine, 2008. 33(25): p.<br />

2691-701. 32. Nerurkar, N.L., et al., Nanofibrous biologic laminates<br />

replicate the form and function of the annulus fibrosus. Nat Mater,<br />

2009. 8(12): p. 986-92. 33. Nerurkar, N.L., et al., Engineered disc-like<br />

angle-ply structures for intervertebral disc replacement. Spine (Phila<br />

Pa 1976), 2011. 35(8): p. 867-73.<br />

Acknowledgments:<br />

This work was supported by the National Institutes of Health, the<br />

Department of Veterans’ Affairs, the US Department of Defense, and<br />

the Penn Center for Musculoskeletal Disorders.<br />

9.1.1<br />

<strong>Cartilage</strong> repair to prevent osteoarthritis. -Fact or fiction?-<br />

T.S. De Windt, D.B.F. Saris<br />

Utrecht/Netherlands<br />

Introduction: We should remain modest and realistic in our<br />

perception of outcomes and discussion with patients of cell based<br />

treatment for osteoarthritis (OA) and end stage cartilage damage.<br />

As best we know the challenge still remains to show prevention of<br />

OA after cell therapy as well as the successful implementation of cell<br />

therapy in the treatment of OA. However many important steps have<br />

been made and some relevant boundaries and advances have been<br />

described. Time to reflect and take score since, OA is a common<br />

condition affecting many adults through pain and decreased joint<br />

function which has a significant impact on quality of life.1 The<br />

general increase in activity level of the past decades and the high<br />

demands in terms of diagnostic and treatment modalities for fast<br />

return to the pre-injury level create one of the biggest challenges<br />

in our field. As such, early OA is increasingly being recognized in<br />

younger active patients, raising the bar. Early OA is considered<br />

more difficult to diagnose than ‘obvious full blown’ OA as signs<br />

and symptoms may still be limited, often becoming manifest after<br />

higher strains such as sport activities.2 In contrast, the diagnosis in<br />

OA is easily made based on the history, physical examination and<br />

radiologic investigation in patients older than 50. In early OA, the<br />

articular cartilage shows fibrillation and vertical fissures that extend<br />

into the mid-zone of the cartilage. The articular surface becomes<br />

discontinuous and there is progressive increase in subchondral bone<br />

plate and subarticular spongiosa.3 Other structures such as the<br />

menisci and ligaments are frequently affected simultaneously, thus<br />

disturbing the joint homeostasis. 4, 5 As early OA generally presents<br />

in younger more active patients with high demands, traditional<br />

treatment strategies for OA such as, anti-inflammatory drugs and<br />

physical therapy will provide temporary relief while endoprosthetic<br />

joint replacement limits function and is prone to future revision<br />

surgery.6 As such, articular cartilage repair is hoped to hold promise<br />

in the treatment of (early) OA patients, providing symptom relief and<br />

potentially delaying or altering osteoarthritic changes and the need<br />

for joint replacement.<br />

Content: <strong>Cartilage</strong> repair procedures for (early OA) Marrow<br />

stimulation Microfracture, is a surgically fast and cost-effective bone<br />

marrow stimulating procedure indicated in cartilage lesions up to 2<br />

cm2 or sometimes even 4 cm2 which provides clinical improvement<br />

for at least two to five years.7, 8 Bae et al. evaluated 44 patients<br />

with an average lesion size of 3.9 cm2 (range 1-6 cm2, Outerbridge<br />

grade IV) with moderate osteoarthritic changes who underwent<br />

microfracture.9 After a mean of 2.3 years, significant improvement<br />

in pain and daily living was seen. In addition, using second-look<br />

arthroscopy, defect filling was determined which was confirmed with<br />

histologic evaluation and collagen type II staining. Miller et al.10<br />

and Steadman et al.11 evaluated microfracture for degenerative<br />

lesions and high impact athletics, respectively with satisfying<br />

clinical outcome and return to high impact sports for more than<br />

five seasons. However, these studies were not aimed specifically at<br />

(early) OA. Brittberg et al. used drilling and subsequent carbon fiber<br />

scaffold implantation for treatment of early osteoarthritic defects in<br />

two separate cohorts with a short-term success rate of over 80% in<br />

terms of pain and clinical outcome.12, 13 Autologous chondrocyte<br />

implantation Autologous chondrocyte implantation (ACI) has found<br />

its place in our treatment algorithm for larger lesions 4 cm2, or on<br />

special indication between 2-4 cm2, demonstrating efficacy for over<br />

ten years follow- op.14 We know that the disturbed joint homeostasis<br />

in early osteoarthritis creates extra difficulty in local cellular<br />

regeneration.6 However, increasing interest in cartilage repair for<br />

(early) osteoarthritis has shown promising results. Indeed In vitro<br />

studies have demonstrated good proliferation and re-differentiation<br />

potential of osteoarthritic chondrocytes, with capability to produce<br />

cartilage specific proteins.15, 16 Furthermore, Kuroda et al.<br />

demonstrated effective cartilage repair after ACI in anterior cruciate<br />

ligament transacted rats.17 Minas et al. reported on a large cohort<br />

consisting of 153 patients (mean age 38 years) with early OA treated<br />

with ACI and followed for up to 11 years. At final follow-up eight<br />

percent of joints were considered failures while 50-75% experienced<br />

significant improvement. At five years, 92 % of patients were<br />

functioning well, delaying the need for joint replacement.18 Filardo<br />

et al. also demonstrated significant improvement after second<br />

generation ACI in patients with degenerative lesions.19 However, it<br />

must be stated that compared to the general population, a higher<br />

failure rate (18.5% at 6 years) was found. Several other studies<br />

have reported improvement in clinical outcome and MRI determined<br />

defect filling after ACI in degenerative and focal osteoarthritis.20,<br />

21 As of yet, no clinical data is available for cell-based therapy in<br />

generalized (early) OA. Osteochondral autologous and allograft<br />

transfer Although limited clinical data is available, osteochondral<br />

autograft transfer (OAT) has been implemented by Hangody et al. in<br />

82 professional athletes with signs of OA. In this 17 year prospective<br />

study, similar success rates were found to that of less athletic patients,<br />

although high motivation resulted in better subjective evaluation.22<br />

In addition, Jakob et al. found good results in ten patients with<br />

patellofemoral OA treated with OAT.23 However concomitant<br />

procedures for patellofemoral maltracking may be an important<br />

confounder. Könst et al. used autologous bone grafting combined<br />

with gel-type ACI (GACI) to treat 9 patients with severe osteochondral<br />

defects. At one year follow-up, statistically significant improvement<br />

was demonstrated in eight patients with only one patient needing<br />

conversion to total knee arhtroplasty. (TKA)24 Osteochondral<br />

allograft tranfer has susccesfully been applied in patients (mean<br />

age 24.3, range 16-44) with steroid-induced osteonecrosis with 90%<br />

graft survival at six years.25 At their last follow-up, TKA was avoided<br />

in 27 out of 28 knees. In contrast, Beaver et al. found a higher failure<br />

rate for post-traumatic osteoarticular bipolar lesions treated with<br />

fresh allografts.26 Furthermore, primary OA has been reported to<br />

reduce clinical outcome after allograft transfer.27 Discussion: fact<br />

from fiction As cartilage repair procedures for treatment of (early)<br />

OA is still a challenge, careful treatment selection is warranted,<br />

specifically in more advanced OA and younger patients. Concomitant<br />

injuries to the whole joint and surroundings are important to treat<br />

as these disturb the joint homeostasis. For instance, anterior<br />

cruciate ligament tears are known to be a risk factor for OA.28-32<br />

Furthermore, correction of malalignment in unicompartemental OA<br />

through osteotomy has proven to reduce pain and improve knee<br />

function.33 Numerous studies have demonstrated the importance of<br />

early treatment in cartilage repair, as reduced clinical outcome was<br />

correlated with defect age ie longer time since symptom onset.7,<br />

35-38 It remains unclear however, if cartilage repair can prevent<br />

OA from developing and the long-term effect on OA has yet to be<br />

determined. Current evidence does suggest however, that TKA can<br />

be delayed with cartilage repair, but there are others means to do<br />

this as well or better. Furthermore, the short-term results (up to five<br />

years) in patients with early osteoarthritic defects are promising.39<br />

New (surgical) strategies may provide symptom relief in active<br />

patients with more severe OA as well. This is an extra challenging<br />

population in which randomized controlled trials have shown that<br />

arthroscopic debridement has no advantage over optimal physical<br />

and medical care.40, 41 However, optimizing tissue regeneration<br />

with functioning cartilage and use of mesenchymal stem cell therapy<br />

are promising strategies which could be implemented for (early) OA.<br />

Conclusion We should remain modest and realistic in our perception<br />

of outcomes and discussion with patients of cell based treatment<br />

for osteoarthritis (OA) and end stage cartilage damage. At best the<br />

challenge still remains to show prevention of OA after cell therapy<br />

as well as the successful implementation of cell therapy in the<br />

treatment of OA. However many important steps have been made<br />

and some relevant boundaries and advances have been described.


Promising results of cartilage repair in early OA encourage us to<br />

continue aiming at long-term effectiveness of preventive and curative<br />

treatment modalities for OA. While not yet a fact; cartilage repair is<br />

a feasible new treatment for (early) OA demonstrating preliminary<br />

short-term effectiveness.<br />

References:<br />

1. Woolf AD, Pfleger B. Burden of major musculoskeletal conditions.<br />

Bull World Health Organ 2003; 81(9):646-656.<br />

2. Luyten FP, Denti M, Filardo G, Kon E, Engebretsen L. Definition and<br />

classification of early osteoarthritis of the knee. Knee Surg Sports<br />

Traumatol Arthrosc 2012; 20(3):401-406.<br />

3. Madry H, Luyten FP, Facchini A. Biological aspects of early<br />

osteoarthritis. Knee Surg Sports Traumatol Arthrosc 2012; 20(3):407-<br />

422.<br />

4. Madry H, Luyten FP, Facchini A. Biological aspects of early<br />

osteoarthritis. Knee Surg Sports Traumatol Arthrosc 2012; 20(3):407-<br />

422.<br />

5. Saris DB, Dhert WJ, Verbout AJ. Joint homeostasis. The discrepancy<br />

between old and fresh defects in cartilage repair. J Bone Joint Surg Br<br />

2003; 85(7):1067-1076.<br />

6. Gomoll AH, Filardo G, de GL et al. Surgical treatment for early<br />

osteoarthritis. Part I: cartilage repair procedures. Knee Surg Sports<br />

Traumatol Arthrosc 2012; 20(3):450-466.<br />

7. Vanlauwe J, Saris DB, Victor J, Almqvist KF, Bellemans J, Luyten FP.<br />

Five-year outcome of characterized chondrocyte implantation versus<br />

microfracture for symptomatic cartilage defects of the knee: early<br />

treatment matters. Am J Sports Med 2011; 39(12):2566-2574.<br />

8. Mithoefer K, McAdams T, Williams RJ, Kreuz PC, Mandelbaum BR.<br />

Clinical efficacy of the microfracture technique for articular cartilage<br />

repair in the knee: an evidence-based systematic analysis. Am J<br />

Sports Med 2009; 37(10):2053-2063.<br />

9. Bae DK, Yoon KH, Song SJ. <strong>Cartilage</strong> healing after microfracture in<br />

osteoarthritic knees. Arthroscopy 2006; 22(4):367-374.<br />

10. Miller BS, Steadman JR, Briggs KK, Rodrigo JJ, Rodkey WG. Patient<br />

satisfaction and outcome after microfracture of the degenerative<br />

knee. J Knee Surg 2004; 17(1):13-17.<br />

11. Steadman JR, Ramappa AJ, Maxwell RB, Briggs KK. An arthroscopic<br />

treatment regimen for osteoarthritis of the knee. Arthroscopy 2007;<br />

23(9):948-955.<br />

12. de Windt TS, Concaro S, Lindahl A, Saris DB, Brittberg M.<br />

Strategies for patient profiling in articular cartilage repair of the<br />

knee: a prospective cohort of patients treated by one experienced<br />

cartilage surgeon. Knee Surg Sports Traumatol Arthrosc 2012.<br />

13. Brittberg M, Faxen E, Peterson L. Carbon fiber scaffolds in the<br />

treatment of early knee osteoarthritis. A prospective 4-year followup<br />

of 37 patients. Clin Orthop Relat Res 1994;(307):155-164.<br />

14. Peterson L, Vasiliadis HS, Brittberg M, Lindahl A. Autologous<br />

chondrocyte implantation: a long-term follow-up. Am J Sports Med<br />

2010; 38(6):1117-1124.<br />

15. Tallheden T, Bengtsson C, Brantsing C et al. Proliferation and<br />

differentiation potential of chondrocytes from osteoarthritic patients.<br />

Arthritis Res Ther 2005; 7(3):R560-R568.<br />

16. Jiang YZ, Zhang SF, Qi YY, Wang LL, Ouyang HW. Cell transplantation<br />

for articular cartilage defects: principles of past, present, and future<br />

practice. Cell Transplant 2011; 20(5):593-607.<br />

17. Kuroda T, Matsumoto T, Mifune Y et al. Therapeutic strategy<br />

of third-generation autologous chondrocyte implantation for<br />

osteoarthritis. Ups J Med Sci 2011; 116(2):107-114.<br />

18. Minas T, Gomoll AH, Solhpour S, Rosenberger R, Probst C, Bryant<br />

T. Autologous chondrocyte implantation for joint preservation<br />

in patients with early osteoarthritis. Clin Orthop Relat Res 2010;<br />

468(1):147-157.<br />

19. Filardo G, Kon E, Di MA et al. Second-generation arthroscopic<br />

Extended Abstracts 137<br />

autologous chondrocyte implantation for the treatment of<br />

degenerative cartilage lesions. Knee Surg Sports Traumatol Arthrosc<br />

2011.<br />

20. Kreuz PC, Muller S, Ossendorf C, Kaps C, Erggelet C. Treatment of<br />

focal degenerative cartilage defects with polymer-based autologous<br />

chondrocyte grafts: four-year clinical results. Arthritis Res Ther 2009;<br />

11(2):R33.<br />

21. Ossendorf C, Kaps C, Kreuz PC, Burmester GR, Sittinger M,<br />

Erggelet C. Treatment of posttraumatic and focal osteoarthritic<br />

cartilage defects of the knee with autologous polymer-based threedimensional<br />

chondrocyte grafts: 2-year clinical results. Arthritis Res<br />

Ther 2007; 9(2):R41.<br />

22. Hangody L, Dobos J, Balo E, Panics G, Hangody LR, Berkes I.<br />

Clinical experiences with autologous osteochondral mosaicplasty in<br />

an athletic population: a 17-year prospective multicenter study. Am J<br />

Sports Med 2010; 38(6):1125-1133.<br />

23. Jakob RP, Franz T, Gautier E, Mainil-Varlet P. Autologous<br />

osteochondral grafting in the knee: indication, results, and<br />

reflections. Clin Orthop Relat Res 2002;(401):170-184.<br />

24. Konst YE, Benink RJ, Veldstra R, van der Krieke TJ, Helder MN,<br />

van Royen BJ. Treatment of severe osteochondral defects of the knee<br />

by combined autologous bone grafting and autologous chondrocyte<br />

implantation using fibrin gel. Knee Surg Sports Traumatol Arthrosc<br />

2012.<br />

25. Gortz S, De Young AJ, Bugbee WD. Fresh osteochondral<br />

allografting for steroid-associated osteonecrosis of the femoral<br />

condyles. Clin Orthop Relat Res 2010; 468(5):1269-1278.<br />

26. Beaver RJ, Schemitsch EH, Gross AE. Disassembly of a one-piece<br />

metal-backed acetabular component. A case report. J Bone Joint<br />

Surg Br 1991; 73(6):908-910.<br />

27. Gomoll AH, Filardo G, Almqvist FK et al. Surgical treatment for<br />

early osteoarthritis. Part II: allografts and concurrent procedures.<br />

Knee Surg Sports Traumatol Arthrosc 2012; 20(3):468-486.<br />

28. Daniel DM, Stone ML, Dobson BE, Fithian DC, Rossman DJ,<br />

Kaufman KR. Fate of the ACL-injured patient. A prospective outcome<br />

study. Am J Sports Med 1994; 22(5):632-644.<br />

29. Kannus P, Jarvinen M. Posttraumatic anterior cruciate ligament<br />

insufficiency as a cause of osteoarthritis in a knee joint. Clin<br />

Rheumatol 1989; 8(2):251-260.<br />

30. Maletius W, Messner K. Eighteen- to twenty-four-year follow-up<br />

after complete rupture of the anterior cruciate ligament. Am J Sports<br />

Med 1999; 27(6):711-717.<br />

31. Von PA, Roos EM, Roos H. High prevalence of osteoarthritis 14<br />

years after an anterior cruciate ligament tear in male soccer players:<br />

a study of radiographic and patient relevant outcomes. Ann Rheum<br />

Dis 2004; 63(3):269-273.<br />

32. Stein V, Li L, Lo G et al. Pattern of joint damage in persons with<br />

knee osteoarthritis and concomitant ACL tears. Rheumatol Int 2011.<br />

33. Brouwer RW, van-Raaij TM, Bierma-Zeinstra-Sita MA, Verhagen<br />

AP, Jakma-Tijs TSC, Verhaar-Jan AN. Osteotomy for treating knee<br />

osteoarthritis. Cochrane Database of Systematic Reviews 2007.<br />

34. Bauer S, Khan RJ, Ebert JR et al. Knee joint preservation with<br />

combined neutralising High Tibial Osteotomy (HTO) and Matrixinduced<br />

Autologous Chondrocyte Implantation (MACI) in younger<br />

patients with medial knee osteoarthritis: A case series with<br />

prospective clinical and MRI follow-up over 5years. Knee 2011.<br />

35. de Windt TS, Bekkers JE, Creemers LB, Dhert WJ, Saris DB. Patient<br />

profiling in cartilage regeneration: prognostic factors determining<br />

success of treatment for cartilage defects. Am J Sports Med 2009;<br />

37 Suppl 1:58S-62S.<br />

36. Krishnan SP, Skinner JA, Bartlett W et al. Who is the ideal<br />

candidate for autologous chondrocyte implantation? J Bone Joint<br />

Surg Br 2006; 88(1):61-64.<br />

37. Mithoefer K, Williams RJ, III, Warren RF, Wickiewicz TL, Marx<br />

RG. High-impact athletics after knee articular cartilage repair: a<br />

prospective evaluation of the microfracture technique. Am J Sports<br />

Med 2006; 34(9):1413-1418.


138<br />

Extended Abstracts<br />

38. Saris DB, Vanlauwe J, Victor J et al. Treatment of symptomatic<br />

cartilage defects of the knee: characterized chondrocyte implantation<br />

results in better clinical outcome at 36 months in a randomized<br />

trial compared to microfracture. Am J Sports Med 2009; 37 Suppl<br />

1:10S-19S.<br />

39. Luyten FP, Vanlauwe J. Tissue engineering approaches for<br />

osteoarthritis. Bone 2011.<br />

40. Kirkley A, Birmingham TB, Litchfield RB et al. A randomized trial<br />

of arthroscopic surgery for osteoarthritis of the knee. N Engl J Med<br />

2008; 359(11):1097-1107.<br />

41. Moseley JB, O‘Malley K, Petersen NJ et al. A controlled trial of<br />

arthroscopic surgery for osteoarthritis of the knee. N Engl J Med<br />

2002; 347(2):81-88.<br />

42. Kaul G, Cucchiarini M, Remberger K, Kohn D, Madry H. Failed<br />

cartilage repair for early osteoarthritis defects: a biochemical,<br />

histological and immunohistochemical analysis of the repair tissue<br />

after treatment with marrow-stimulation techniques. Knee Surg<br />

Sports Traumatol Arthrosc 2012.<br />

9.1.2<br />

What is the key pathway to prevent post-traumatic arthritis for<br />

future molecule-based therapy?<br />

S. Chubinskaya<br />

Chicago/United States of America<br />

Introduction: Joint injuries are becoming increasingly common, with<br />

young adults between the ages of 18-44 seeking medical attention<br />

for joint sprains, dislocation, fractures, anterior cruciate ligament and<br />

meniscal tears, and others. The cascade of events that follow these<br />

joint injuries have been shown to increase the risk (20-50%) of posttraumatic<br />

osteoarthritis (PTOA). Understanding biological responses<br />

that predispose the onset of PTOA will help in determining treatment<br />

strategies in order to delay and/or prevent the progression of the<br />

disease.<br />

Content: Recent research on the events that follow joint trauma<br />

have shown chondrocyte death/apoptosis, inflammation (elevation<br />

of caspases, selected pro-inflammatory cytokines, matrix fragments,<br />

nitric oxide, reactive oxygen species, etc.) and matrix damage<br />

(activation of aggrecanases and matrix metalloproteinases that<br />

lead to the cleavage of cartilage matrix constituents) to be early<br />

phase responses to injury. Together they lead to the development<br />

of OA-like focal cartilage lesions characterized by the loss of matrix<br />

molecules, surface fibrillation, and fissures that if untreated have<br />

a tendency to expand and progress to fully-blown disease. One of<br />

the unanswered PTOA questions is when and which therapies that<br />

have been developed as disease-modifying OA drugs are indicated<br />

for patients with post-traumatic OA and whether a different set of<br />

treatments and molecular targets has to be considered. Through<br />

basic and clinical research an impressive progress has been made<br />

towards elucidation of pathogenesis of PTOA and understanding<br />

the mechanisms that govern immediate cellular responses to<br />

injury. However, this still requires further validation in a large<br />

cohort of patients with various types of joint injuries. The ideal<br />

therapy to arrest and prevent the development and progression<br />

of PTOA must be multi-varied and include anabolic effects on<br />

chondrocyte metabolism characterized by elevated intrinsic repair,<br />

while protecting integrity of cell membrane and inhibiting catabolic<br />

pathways that lead to chondrocyte death and matrix loss. The<br />

following are the key mechanisms that should constitute the basis<br />

for the design of intervention therapies: 1) Chondroprotection; 2)<br />

Anti-inflammatory; 3) Matrix protection; and 4) Pro-anabolic, stimuli<br />

of cartilage remodeling and regeneration. Chondroprotective factors<br />

prevent the damage of chondrocyte membrane, inhibit apoptosis, or<br />

antagonize pathways that lead to cell death. Among those available<br />

for clinical or experimental use are P188 surfactant, various specific<br />

and broad inhibitors of caspases, anti-oxidants (inhibitors of nitric<br />

oxide synthase, rotenone, N-acetylcysteine) and other. As antiinflammatory,<br />

anti-tumor necrosis factor and interleukin-1 receptor<br />

antagonist received the most attention due to their availability for<br />

clinical use in rheumatoid arthritis or in various experimental animal<br />

models of OA or PTOA. To protect cartilage matrix integrity various<br />

inhibitors of aggrecanases and matrix metalloproteinases have been<br />

considered. One of the very important directions in the development<br />

of pharmacological interventions in PTOA is the ability to stimulate<br />

production of new cartilage extracellular matrix. The best candidates<br />

are growth factors including members of the Transforming Growth<br />

Factor (TGF)-β superfamily, bone morphogenetic proteins (BMP),<br />

Fibroblast Growth Factors, and Insulin like Growth Factor (IGF)-<br />

1. The most studied and the most promising for cartilage repair<br />

is osteogenic protein-1 or BMP-7 (OP-1/BMP-7). At this stage of<br />

our cumulative knowledge, BMP-7 appears to be one of the best<br />

candidate therapeutic agents for cartilage treatment after injury,<br />

since it affects both catabolic and anabolic responses. We believe that<br />

the most beneficial agents for the treatment of PTOA are those that<br />

target multiple pathways and mechanisms. A number of molecular<br />

targets have been identified and many of the existing therapeutic<br />

agents have been already tested in vitro and in vivo. However, the<br />

biggest remaining challenge is the translation of this knowledge into<br />

the clinic and the development of appropriate effective therapy/<br />

therapies administered within the window of opportunity. Currently,<br />

the most suitable route for administering such therapy appears to<br />

be intra-articular injections that allow accumulation of critical doses<br />

of the drug within the damaged area and also reduce the risk of<br />

systemic side effects. To monitor the efficacy of the PTOA therapy,<br />

an appropriate set of bio- and imaging markers is needed that could<br />

predict and correlate with the progression of the disease, since it<br />

takes years and decades for the disease to develop.<br />

References:<br />

1. Anderson, DD; Chubinskaya, S; Guilak, F; Martin, JA; Oegema, TR;<br />

Olson, SA and Buckwalter, JA. (2011). Post-traumatic osteoarthritis:<br />

improved understanding and opportunities for early intervention. J<br />

Orthop Res, 29(6):802-9.<br />

2. Bajaj, S; Shoemaker, T; Hakimiyan, AA; Rappoport, L; Pascual-<br />

Garrido, C; Oegema, TR; Wimmer, MA and Chubinskaya, S. (2010).<br />

Protective effect of P188 in the model of acute trauma to human ankle<br />

cartilage: the mechanism of action. J Orthop Trauma, 24(9):571-6.<br />

3. Buckwalter, JA and Brown, TD. (2004). Joint injury, repair, and<br />

remodeling: roles in posttraumatic osteoarthritis. Clin Orthop Relat<br />

Res, 423):7-16.<br />

4. Chubinskaya, S; Hurtig, M and Rueger, DC. (2007). OP-1/BMP-7 in<br />

cartilage repair. Int Orthop, 31(6):773-81.<br />

5. Chubinskaya, S; Otten, L; Soeder, S; Borgia, JA; Aigner, T; Rueger,<br />

DC and Loeser, RF.(2011). Regulation of chondrocyte gene expression<br />

by osteogenic protein-1. Arthritis Res Ther, 13(2):R55.<br />

6. Ellsworth, JL; Berry, J; Bukowski, T; Claus, J; Feldhaus, A;<br />

Holderman, S; Holdren, MS; Lum, KD; Moore, EE; Raymond, F; Ren,<br />

H; Shea, P; Sprecher, C; Storey, H; Thompson, DL; Waggie, K; Yao,<br />

L; Fernandes, RJ; Eyre, DR and Hughes, SD. (2002). Fibroblast Post-<br />

Traumatic Osteoarthritis: Biologic Approaches to Treatment growth<br />

factor-18 is a trophic factor for mature chondrocytes and their<br />

progenitors. Osteoarthritis <strong>Cartilage</strong>, 10(4):308-20.<br />

7. Elsaid, KA; Machan, JT; Waller, K; Fleming, BC and Jay, GD.<br />

(2009). The impact of anterior cruciate ligament injury on lubricin<br />

metabolism and the effect of inhibiting tumor necrosis factor<br />

alpha on chondroprotection in an animal model. Arthritis Rheum,<br />

60(10):2997-3006.<br />

8. Lotz, MK and Kraus, VB. (2010). New developments in osteoarthritis.<br />

Posttraumatic osteoarthritis: pathogenesis and pharmacological<br />

treatment options. Arthritis Res Ther, 12(3):211.<br />

Acknowledgments:<br />

This work was supported by the National Football League Charities,<br />

Ciba-Geigy Endowed Chair and Department of Biochemistry, Rush<br />

University Medical Center. The author would like to acknowledge<br />

Drs. Markus A. Wimmer, Theodore R Oegema, and Jeffrey A. Borgia<br />

for their important contributions to this work. The authors would<br />

like to acknowledge Dr. Arkady Margulis for tissue procurement and<br />

Dr. Lev Rappoport and Mrs. Arnavaz Hakimiyan for their technical<br />

assistance. The author also would like to acknowledge the Gift of<br />

Hope Organ & Tissue Donor Network and donor’s families.


9.1.3<br />

ACL reconstruction and osteoarthritis: Evidence from long-term<br />

follow-up and potential solutions<br />

R.A. Magnussen 1 , V. Duthon 2 , E. Servien 3 , P. Neyret 3<br />

1 Columbus/United States of America, 2 Geneva/Switzerland, 3 Lyon/<br />

France<br />

Introduction: Injury to the anterior cruciate ligament frequently<br />

results in symptomatic knee instability that significantly limits<br />

knee function. Modern surgical and rehabilitation techniques are<br />

frequently able to restore knee stability and allow return to high<br />

function; however, a major long-term concern remains the high risk<br />

of subsequent development of arthritis in this young, active patient<br />

population. The goal of this presentation is to explore several<br />

questions, the answers to which are key to our understanding and<br />

eventually to the prevention of this frequent source of morbidity.<br />

These questions include:<br />

- What is the natural history of ACL deficiency?<br />

- How important is the status of the meniscus at the time of<br />

reconstruction?<br />

- Does ACL reconstruction prevent the development of osteoarthritis<br />

in the long term?<br />

- Can we predict which patients will develop osteoarthritis?<br />

- What can be done???<br />

Content: What is the natural history of ACL deficiency?<br />

Review of the literature reveals osteoarthritis rates of about 40%<br />

following fifteen years of ACL deficiency. Longer-term follow-up<br />

demonstrates these rates to increase to near 90% by 25 to 35 years,<br />

with up to 50% of patients undergoing TKA in some series. Meniscal<br />

status does not appear to be the major predictor of outcome in<br />

this population. Meniscal loss likely contributes to more rapid<br />

development of osteoarthritis in this patient population.<br />

How important is the status of the meniscus at the time of<br />

reconstruction?<br />

Meniscus status appears to be a much strong predictor of subsequent<br />

development of osteoarthritis in cases of ACL reconstruction.<br />

Review of the literature reveals numerous comparative studies<br />

with 4 to 12 year follow-up comparing rates of osteoarthritis in ACLreconstructed<br />

patients who underwent partial meniscectomy with<br />

normal menisci versus those with normal menisci. These studies<br />

demonstrate 2- to 10-fold increased risk of osteoarthritis following<br />

partial meniscectomy.<br />

Our series of patients who underwent ACL reconstruction in Lyon<br />

between 1978 and 1983 that have been followed for a mean of 24.5<br />

years demonstrate similar findings. Patients were twice as likely to<br />

develop IKDC grade C or D osteoarthritis if they underwent partial<br />

meniscectomy at the time of ACL reconstruction. Articular cartilage<br />

lesions noted at the time of ACL reconstruction also appear to be<br />

significant predictors of future development of osteoarthritis. In this<br />

same series, the presence of medial compartment cartilage defects<br />

at reconstruction was associated with a 5-times increased risk of<br />

osteoarthritis at final follow-up.<br />

Does ACL reconstruction prevent the development of osteoarthritis<br />

in the long term?<br />

Unfortunately, long-term comparative studies evaluating osteoarthritis<br />

rates in similar ACL-injured populations treated with or without ACL<br />

reconstruction are rare. One can compare case series of patients<br />

treated with and without surgery to estimate expected rates of<br />

osteoarthritis. In patients with normal menisci at the time of surgery,<br />

rates of osteoarthritis appear to be higher 25 to 35 years later in patients<br />

that did not undergo ACL reconstruction (40 to 90%) relative to those<br />

who did undergo reconstruction (35%). Similar results are found in<br />

patients with abnormal menisci at the time of reconstruction, with nonreconstructed<br />

patients exhibiting higher rates of osteoarthritis (90%)<br />

than those who underwent ACL reconstruction (40 to 50%). One must<br />

remember that such comparisons are subject to numerous sources of<br />

bias and represent low-level evidence.<br />

Several comparative studies evaluating these same questions 11 to<br />

19 years after ACL injury has recently been published. The results are<br />

variable with osteoarthritis rates between 28 and 65% in all groups.<br />

No clear change in osteoarthritis rates with ACL reconstruction is<br />

consistently evident.<br />

Extended Abstracts 139<br />

Can we predict which patients will develop osteoarthritis?<br />

Following ACL reconstruction, it would be ideal to be able to predict<br />

which patients are at highest risk for subsequent osteoarthritis<br />

development and then intervene in some way to minimize this risk.<br />

As above, undergoing partial meniscectomy at the time of surgery<br />

does appear to increase this risk. Further, our data from Lyon indicate<br />

that patients with no evidence of degenerative change on plain films<br />

11 years after surgery are at very low risk to develop osteoarthritis<br />

over the next 15 years. Similarly, if early evidence of degenerative<br />

change is visible on radiographs 11 years following surgery, the risk<br />

of significant progression of osteoarthritis over the next 15 years is<br />

quite high.<br />

What can be done???<br />

Strategies to reduce osteoarthritis risk begin at the time of the initial<br />

ACL reconstruction. Meniscal repair and preservation has been<br />

shown in several series to reduce subsequent risk of osteoarthritis.<br />

Meniscus tissue can also potentially be maximized by operating<br />

earlier on ACL-deficient knees and preventing the occurrence of<br />

some meniscal tears that can develop with persistent instability.<br />

Similarly, when evidence of early degenerative change is seen,<br />

one can consider intervention at the time through either activity<br />

modification (limiting high-impact activities) or altering joint loading<br />

forces through high tibial osteotomy. There is also great potential<br />

exhibited by many new techniques that may aid in meniscal<br />

preservation, cartilage restoration, and joint protection. Further<br />

research and longer follow-up are necessary to accurately evaluate<br />

the potential of such techniques.<br />

References:<br />

Pernin J, Verdonk P, Aïtsiselmi T, Marsin P, Neyret P, Long-term<br />

Follow-up of 24.5 years After Intra-Articular AnteriorCruciate<br />

Ligament Reconstruction With Lateral Extra articular Augmentation,<br />

Am J Sports Med 2012, June 38(6) 1094-102<br />

Rotterud J, Risberg MA, Engelretsen L, Aroen A, Patients with focal<br />

full-thickness cartilage lesions benefit less from ACL reconstruction<br />

at 2-5 years follow-up Knee Surg Sports Traumatol Arthrosc; DOI<br />

10.1007/S00167-011-1739-y; E. Ruh 2011 Nov 8<br />

Hjermundrud V, Bune T, Risberg M, Engelretsen L, Aroen A; Full<br />

Thickness cartilage lesion do not offer knee function in patients with<br />

ACL injury; Knee Surg Sports Traumatol Arthrosc 2010, March 18<br />

(3)298-303; DOI 10.1007/S00167.009-0894-x<br />

Widuschowski W, Widuschowski J, Koczy B, Szyluk K; Untreated<br />

asymptomatic deep cartilage lesions associated with anterior<br />

cruciate ligament injury : results at 10 and 15 years follow-up; Am J<br />

Sports Med 2009, Apr ; 37(4) 688-92 Epub 2009 Feb 3;<br />

Bonin N, Aïtsiselmi T, Donell St, Dejour H, Neyret P; Anterior cruciate<br />

reconstruction combined with valgus upper tibial osteotomy : 12<br />

years follow-up; The Knee 2004, 11(6) : 431-37<br />

Neyret P, Walch G, Dejour H; Rev Chir Orthop 1998; Intra-nural<br />

medical menisectomy by the Trillat technique Long Term follow-up<br />

of 258 operations; French J. Orthop Sur 1988, 4, 520-34<br />

Acknowledgments:<br />

- TBF (cartipatch)<br />

- Smith and Nephew<br />

- Tornier


140<br />

Extended Abstracts<br />

9.1.4<br />

Anatomic ACL Reconstruction -Current concept and future<br />

perspective<br />

F.H. Fu<br />

Pittsburgh/United States of America<br />

Introduction: Rationale for Anatomic DBACL Reconstruction<br />

- Anatomy is the basis of orthopedic surgery. The goals of anatomic<br />

ACL reconstruction are to restore 60-80% of the native ACL anatomy,<br />

and to maintain a long term knee health.<br />

- Traditional ACL-R has been successful in returning patients to sports<br />

activities. However, radiographic evidence of degenerative changes has<br />

been observed in up to 90% of patients at mid-term follow-up study after<br />

traditional SBACL reconstruction.1-2<br />

- Critical review of the literature from the last ten years reveals that<br />

between 10% and 30% of patients complain of pain and residual<br />

instability following traditional SB (SB) ACL reconstruction.3 Metaanalysis<br />

showed that no more than 60% of the patients will make a full<br />

recovery after their ACL reconstruction.4<br />

- The PL bundle, which is not traditionally reconstructed, plays a significant<br />

role in rotatory stability in the knee. Numerous clinical and basic science<br />

studies have demonstrated that: 1) traditional SBACL reconstruction<br />

does not adequately restore normal knee kinematics, particularly tibial<br />

rotation5, and 2) anatomic DB (DB) reconstruction more closely restores<br />

normal knee kinematics when compared to SB reconstruction.6<br />

- Recent biodynamic research has shown evidence that anatomic DB<br />

ACL-R restores knee kinematics during daily activities back to normal<br />

and might help prevent the early onset of knee osteoarthritis.7<br />

- A recently performed RCT, with 85% follow-up at 3-5 years, showed<br />

clinical superiority for anatomic placement of the graft and small but<br />

significant differences between anatomic DB and SB ACL-R.8<br />

Content: The principle of anatomic ACL DB reconstruction<br />

- Reproducing the two bundle anatomy of ACL<br />

- The ACL is composed of two functional bundles, the anteromedial (AM)<br />

bundle and the posterolateral (PL) bundle.9<br />

- Reproducing the insertion sites of ACL<br />

- The insertion sites of the AM and PL bundle should be identified and<br />

marked for anatomic tunnel placement. The femoral insertion sites of<br />

the AM and PL bundle are oriented vertically with the knee in extension<br />

and become horizontal in 90º of knee flexion (surgical position for ACL<br />

reconstruction surgery). In extension the two bundles are parallel and in<br />

flexion they become crossed.9<br />

- Reproducing the tension pattern of ACL<br />

- The AM bundle has its highest tension at 45o of knee flexion, and was<br />

taut throughout the range of motion. The PL bundle has its highest<br />

tension at full extension, and becomes lax as the knee flexes. The AM<br />

and PL graft should be fixed at these angles of knee flexion to closely<br />

reproduce the native tension pattern.10<br />

- Individualized surgery<br />

- The insertion sites of each bundle should be identified and marked, and<br />

the size of the insertion sites should be measured to tailor the surgery<br />

for each individual. The concept of anatomic ACL reconstruction can be<br />

applied to all ACL surgeries (SB, DB, revision, one-bundle augmentation).<br />

The decision of whether to perform a single or DB ACL reconstruction<br />

should be dictated by the unique anatomy of the patient.11<br />

Pitfalls in Traditional ACL reconstruction<br />

- Femoral insertion sites orientation changes with knee flexion: The<br />

femoral AM and PL insertion sites are horizonally oriented when the<br />

knee is close to 90 degrees of flexion, while they are vertically oriented<br />

in knee extension. The important concept is often neglected in ACL<br />

reconstruction.<br />

- The use of clock face reference: The knee is a 3 dimensional structure.<br />

The clock concept is easy to use. However, it is inaccurate in describing<br />

the location of femoral tunnel placement and lead to non-anatomic<br />

tunnel position.<br />

- Inability to observe the femoral insertion site well by using the<br />

anteromedial portal: The anteromedial portal provides a superior<br />

view of the lateral wall of the notch and the femoral insertion site of<br />

ACL than the anterolateral portal, which is sufficient in observing the<br />

tibial insertion site.<br />

- Graft impingement: It is a concept created by us because of nonanatomic<br />

tunnel placement. The native ACL does NOT impinge with<br />

notch and PCL. As long as the tunnels were placed in an anatomic<br />

fashion, there will be no impingement. However, if the tunnel is<br />

placed non-anatomically, impingement may occur.<br />

- Mismatch tunnels: With fear of impingement, we traditionally mismatch<br />

our tunnel placement by placing the tibial tunnel more posteriorly<br />

(close to the PL insertion site), and placing the femoral tunnel at the<br />

native AM or high AM position.11-13 Non-anatomic ACL reconstruction<br />

leads to inferior biomechanical properties and biological healing due<br />

to non-physiological biomechanical stress to the graft.<br />

- DB ACL-R does not necessarily mean anatomic reconstruction, if the<br />

native anatomy was not followed as a guideline for placement of the<br />

tunnels.<br />

Anatomic DB ACL Reconstruction<br />

- Pre-operatively, the ACL insertion site and ACL length can be measured<br />

on the sagittal MRI. The ACL inclination angle can also be measured.<br />

- The MRI can also be used to measure the size of the certain autografts.<br />

Both the patellar tendon and the quadriceps tendon size can be<br />

measured on the sagittal MRI sequence. The quadriceps tendon is<br />

often much larger than the patellar tendon and can offer more autograft<br />

substance.<br />

- Anatomic DB ACL-R is an “Insertion Site Surgery”. We utilize three<br />

portals: Lateral Portal (LP), Medial Portal (MP), and Accessory Medial<br />

Portal (AMP).<br />

- We routinely place the arthroscope in the MP and work through<br />

the AMP. In doing so, visualization of the femoral insertion of the<br />

ACL is greatly enhanced and the need for notchplasty is virtually<br />

eliminated.14<br />

- The anatomic insertion sites of each native ACL bundle are marked on<br />

the femur and tibia with a thermal device, with care taken to preserve<br />

the border of the bundles for later reference. This is a critical step in<br />

identifying the correct placement of the tunnels, and is performed<br />

prior to resection of any residual ACL tissue. In addition, the length<br />

and width of the AM and PL bundle insertion site are measured as<br />

references to decide tunnel diameters. The surgery is individualized for<br />

each patient.<br />

- A “lateral bifurcate ridge” is often seen on the femoral insertion<br />

between the AM and PL bundles, where as a “lateral intercondylar<br />

ridge” is often seen on the upper limit of both the AM and PL bundles.<br />

These are useful surgical landmarks in addition to the native insertion<br />

fibers.15-16<br />

- Notchplasty destroys the femoral anatomy of the ACL and is<br />

unnecessary if medial and accessory medial portals are used.<br />

- The tibial and femoral tunnels are placed at their native insertion site,<br />

which were previously marked by thermal device.<br />

- The PL femoral tunnel is always drilled through the anteromedial<br />

portal. The primary advantage of drilling trans-tibially for the AM<br />

femoral tunnel is the creation of a longer tunnel which diverges from<br />

the PL femoral tunnel, and we routinely attempt this approach first<br />

before using the accessory medial portal. However, sometimes it can’t<br />

reach the anatomic insertion site. In that case, the tunnel will be drill<br />

through the anteromedial portal.<br />

- The PL graft is then passed first, followed by the AM graft. Femoral<br />

fixation is typically performed with an EndoButton.<br />

- Post-operatively, the MRI can be used to compare the pre- and postop<br />

insertion site size to measure how much of the insertion site is<br />

restored. To assess proper tunnel and graft placement the pre- and<br />

post-operative inclination angle and tunnel angle can be compared on<br />

MRI and AP radiographs respectively.17 Additionally 3D CT scan can be<br />

used to evaluate tunnel position.<br />

Anatomic SB ACL Reconstruction


- Except for an one bundle augmentation (performed when only one of<br />

the two native bundles are torn), there are a few other scenarios where<br />

we prefer to perform SB surgery (30%):18<br />

- Small native ACL insertion site (< 14mm)<br />

- Open growth plates<br />

- Severe arthritic changes<br />

- Multiple knee ligament injuries<br />

- Severe bone bruising<br />

- Narrow or shallow intercondylar notch<br />

- SB surgery is performed with attention to soft tissue and bony<br />

landmarks, while identifying rupture pattern and native ACL insertion<br />

sites (similar to DB ACL surgery). Then, the tibial tunnel is placed at<br />

midway between the native insertion sites of the AM bundle and PL<br />

bundles.<br />

- The distance from anterior margin of ACL footprint to center of tibial<br />

tunnel should be measured, and the femoral tunnel should be placed<br />

at the same distance from the posterior margin (knee in 90º flexion) of<br />

the femoral ACL footprint.<br />

One Bundle Augmentation<br />

- In some cases only the AM or the PL bundle is torn, then the intact<br />

bundle is saved and “augmented” with a SB reconstruction <strong>–</strong> either the<br />

AM or PL, whichever one is torn.<br />

Clinical Outcomes<br />

- Clinical improvements have been demonstrated in recent prospective<br />

and randomized level I and level II studies. These studies have shown<br />

superior outcomes for anatomic DB reconstruction rather than<br />

conventional SB reconstruction.8,19<br />

- While preliminary results are encouraging, additional work is needed<br />

to critically evaluate the outcomes of DB ACL reconstruction in terms<br />

of joint kinematics, degenerative joint changes, and patient-reported<br />

outcomes. Better (true objective) methods for rotational laxity<br />

measurement, intermediate and long-term outcomes are needed in<br />

the future.<br />

- A meta-analysis by Lubowitz et al. summarized the results of pivot<br />

shift consistent with the convention of IKDC. The normal and nearly<br />

abnormal data were pooled together for pivot shift and therefore,<br />

SB and DB achieved 94.6% and 97.5% of good pivot shift results<br />

respectively with no difference between the two groups. This is how we<br />

reported clinical outcome for many years. However, if we want to review<br />

the data more critically by only comparing the “normal” category, DB<br />

provided significantly better results (83.1% DB vs. 67.9% SB).20,21<br />

- Complications such as graft failure, hardware failure, and infections<br />

are also seen with DB ACL reconstruction. The double tunnels in femur<br />

and tibia may increase the difficulty in revising failed grafts.<br />

To fully assess the outcome of ACL reconstruction, we need to improve<br />

our outcome measures. Outcome measures should be objective,<br />

accurate, precise and reliable. Examples are in vivo kinematics with<br />

dynamic stereo x-ray, high resolution/ 3D MRI and 3D CT scan. Only<br />

then we can improve our surgical technique and protect the long-term<br />

knee health of our patients.<br />

Conclusion<br />

- The goals of anatomic ACL reconstruction are to restore 60-80% of<br />

the native ACL anatomy, and to maintain a long term knee health.<br />

- The DB anatomy, insertion sites, and tension pattern need to be<br />

reproduced to restore native ACL anatomy and knee kinematics.<br />

- Surgery should be individualized to the patient.<br />

- Better restoration of ACL anatomy leads to better restoration of the<br />

knee joint function.<br />

- Anatomic DBACL Reconstruction is a principle that can be applied to<br />

SB, one-bundle augmentation and revision ACL surgeries<br />

- We need improved, more objective outcomes measures, including<br />

biology, kinematics and imaging.<br />

Extended Abstracts 141<br />

References:<br />

1) Fithian DC, Paxton EW, Stone ML, Luetzow WF, Csintalan RP,<br />

Phelan D, Daniel DM. Prospective trial of a treatment algorithm for<br />

the management of the anterior cruciate ligament-injured knee. Am<br />

J Sports Med 2005;33-3:335-46.<br />

2) Keays SL, Newcombe PA, Bullock-Saxton JE, Bullock MI, Keays AC.<br />

Factors involved in the development of osteoarthritis after anterior<br />

cruciate ligament surgery. Am J Sports Med 2010;38-3:455-63.<br />

3) Yunes M, Richmond JC, Engels EA, Pinczewski LA. Patellar versus<br />

hamstring tendons in anterior cruciate ligament reconstruction: A<br />

meta-analysis. Arthroscopy 2001;17-3:248-57.<br />

4) Biau DJ, Tournoux C, Katsahian S, Schranz P, Nizard R. ACL<br />

reconstruction: a meta-analysis of functional scores. Clin Orthop<br />

Relat Res 2007;458:180-7.<br />

5) Tashman S, Collon D, Anderson K, Kolowich P, Anderst W.<br />

Abnormal rotational knee motion during running after anterior<br />

cruciate ligament reconstruction. Am J Sports Med 2004;32-4:975-<br />

83.<br />

6) Yagi M, Kuroda R, Nagamune K, Yoshiya S, Kurosaka M. Doublebundle<br />

ACL reconstruction can improve rotational stability. Clinical<br />

Orthopaedics and Related Research 2007-454:100-7.<br />

7) Kopf, S; Moloney, G; Freismuth, K; Fu, FH; Tashman, S. Anatomic<br />

DB Anterior Cruciate Ligament Reconstruction Restores Normal<br />

Dynamic In Vivo Knee Kinematics. Presented as podium presentation<br />

during the Orthopaedic Research <strong>Society</strong> Meeting in San Francisco,<br />

February, 2012.<br />

8) Hussein M, van Eck CF, Cretnik A, Dinevski D, Fu FH. Prospective<br />

randomized clinical evaluation of conventional single-bundle,<br />

anatomic single-bundle, and anatomic DBanterior cruciate ligament<br />

reconstruction: 281 cases with 3- to 5-year follow-up. 2011 November<br />

15. [Epub ahead of print]<br />

9) Chhabra A, Starman JS, Ferretti M, Vidal AF, Zantop T, Fu FH.<br />

Anatomic, radiographic, biomechanical, and kinematic evaluation of<br />

the anterior cruciate ligament and its two functional bundles. J Bone<br />

Joint Surg Am 2006;88 Suppl 4:2-10.<br />

10) Gabriel MT, Wong EK, Woo SL, Yagi M, Debski RE. Distribution of<br />

in situ forces in the anterior cruciate ligament in response to rotatory<br />

loads. J Orthop Res 2004;22-1:85-9.<br />

11) van Eck CF, Schreiber VM, Liu TT, Fu FH. The anatomic approach<br />

to primary, revision and augmentation anterior cruciate ligament<br />

reconstruction. Knee Surg Sports Traumatol Arthrosc 2010;DOI<br />

10.1007/s00167-010-1191-4<br />

12) Kopf S, Forsythe B, Wong AK, Tashman S, Anderst W, Irrgang JJ,<br />

Fu FH. Nonanatomic tunnel position in traditional transtibial singlebundle<br />

anterior cruciate ligament reconstruction evaluated by threedimensional<br />

computed tomography. J Bone Joint Surg Am 2010;92-<br />

6:1427-31.<br />

13) Forsythe B, Kopf S, Wong AK, Martins CA, Anderst W, Tashman S,<br />

Fu FH. The location of femoral and tibial tunnels in anatomic doublebundle<br />

anterior cruciate ligament reconstruction analyzed by threedimensional<br />

computed tomography models. J Bone Joint Surg Am<br />

2010;92-6:1418-26.<br />

14) Cohen SB, Fu FH. Three-portal technique for anterior cruciate<br />

ligament reconstruction: use of a central medial portal. Arthroscopy<br />

2007;23-3:325 e1-5.<br />

15) van Eck CF, Morse KR, Lesniak BP, Kropf EJ, Tranovich MJ, van<br />

Dijk CN, Fu FH. Does the lateral intercondylar ridge disappear in ACL<br />

deficient patients? Knee Surg Sports Traumatol Arthrosc 2010;DOI<br />

10.1007/s00167-009-1038-z.<br />

16) Fu FH, Jordan SS. The lateral intercondylar ridge--a key to<br />

anatomic anterior cruciate ligament reconstruction. J Bone Joint<br />

Surg Am 2007;89-10:2103-4.<br />

17) Illingworth KD, Hensler D, Working ZM, Macalena JA, Tashman<br />

S, Fu FH. A simple evaluation of anterior cruciate ligament femoral<br />

tunnel position: the inclination angle and femoral tunnel angle. Am J<br />

Sports Med. 2011 Dec;39(12):2611-8. Epub 2011 Sep 9.


142<br />

Extended Abstracts<br />

18) van Eck CF, Lesniak BP, Schreiber VM, Fu FH. Anatomic Single-<br />

and Double-Bundle Anterior Cruciate Ligament Reconstruction<br />

Flowchart. Arthroscopy 2010;26-2:258-68<br />

19) Kondo E, Yasuda K, Azuma H, Tanabe Y, Yagi T. Prospective<br />

clinical comparisons of anatomic double-bundle versus singlebundle<br />

anterior cruciate ligament reconstruction procedures in 328<br />

consecutive patients. American Journal of Sports Medicine.36(9)()<br />

(pp 1675-1687), 2008.Date of Publication: Sep 2008. 2008-9:1675-<br />

87.<br />

20) Meredick RB, Vance KJ, Appleby D, Lubowitz JH. Outcome of<br />

single-bundle versus double-bundle reconstruction of the anterior<br />

cruciate ligament: a meta-analysis. Am J Sports Med 2008;36-<br />

7:1414-21.<br />

21) Irrgang JJ, Bost JE, Fu FH. Re: Outcome of single-bundle versus<br />

double-bundle reconstruction of the anterior cruciate ligament: a<br />

meta-analysis. Am J Sports Med 2009;37-2:421-2; author reply 2.<br />

Acknowledgments:<br />

University of Pittsburgh Medical Center Department of Orthopaedic<br />

Surgery<br />

14.2<br />

The current prospects for gene therapy as a non-cellular therapy<br />

L. Goodrich 1 , W. Mcilwraith 1 , J. Samulski 2<br />

1 Fort Collins/United States of America, 2 Chapel Hill/United States<br />

of America<br />

Introduction: The following is an overview of what will be presented<br />

in the presentation of “the current prospects for gene therapy as a<br />

non-cellular therapy.<br />

Content: Gene therapy for joint disease has been in existence for 2<br />

decades. Its applications have included therapy for osteoarthritis,<br />

cartilage repair, rheumatoid arthritis and other, less common joint<br />

abnormalities. There is a large body of evidence in both animal studies<br />

and clinical trials that suggest the therapy is safe and effective[1-4]. So<br />

where are we now and what does the future hold for gene therapy? Will<br />

it be a clinical reality and if so when? This presentation will give a brief<br />

overview of gene therapy, a background on what has been done and<br />

where it is going as a clinical modality to treat joint disease. Intra-articular<br />

gene therapy is typically administered by one of two approaches, 1)<br />

direct or otherwise known as in-vivo or 2) indirect or otherwise known<br />

as ex-vivo[3]. Direct gene therapy is most often administered into the<br />

joint by intra-articular injection and indirect is commonly performed by<br />

delivering cells that have been genetically modified to the joint. Many<br />

different types of cells have been utilized for ex-vivo gene therapy<br />

including synoviocytes, chondrocytes, mesenchymal stem cells, or<br />

antigen presenting cells. For this presentation, gene therapy as a noncellular<br />

therapy will be discussed and therefore, ex-vivo approaches<br />

will not be covered. The concept of direct in-vivo injection of gene<br />

therapy vectors is straightforward. Therapeutic complementary DNA<br />

(cDNA), is placed into a vector backbone (usually derived from a virus)<br />

and the gene therapeutic vector is then injected into the joint. The<br />

injection of these vector particles usually results in transduction of the<br />

tissues within the joint to result in production of the desired therapeutic<br />

protein intra-articularly. Tissues within the joint that are targets most<br />

commonly include synovium, ligaments, cartilage and perhaps joint<br />

capsule[5]. For many gene therapeutic vectors, chondrocytes within<br />

the cartilage are not efficiently transduced due to the dense matrix<br />

components of cartilage although one viral vector, adenoassociated<br />

viral vector (AAV) appears to perhaps overcome that barrier. The<br />

overarching goal for the gene therapeutic approach is to get long-term<br />

protein production in the diseased joint to both reduce inflammation<br />

and enhance catabolism to result in a reduction or a reversal of<br />

disease progression. Many different transgenes have been used intraarticularly<br />

to treat joint disease, the most common including interleukin<br />

receptor antagonist protein (IL-1ra), insulin-like growth factor-I (IGF-I),<br />

transforming growth factor beta (TGF-ß), and human tumor necrosis<br />

factor receptor-immunoglobulin Fc (TNFR:Fc). Viral gene therapy has<br />

been most commonly utilized due the fact that nonviral gene delivery<br />

has not been efficacious in animal models[6]. Viral vectors that have<br />

been successfully utilized in-vivo in animal models include adenovirus,<br />

AAV, retrovirus, herpes simplex virus (HSV) and lentivirus[1, 7, 8]. As<br />

one would expect, all viral vectors have advantages and disadvantages.<br />

Adenovirus and HSV initiate inflammation and cytotoxicity within the<br />

joint and, most importantly, do not result in long-term expression of<br />

transgenes. Retroviruses require host-cell division and therefore are<br />

generally only utilized for ex-vivo transfer since cell division is low in the<br />

intra-articular environment[9]. Lentivirus, also considered a retrovirus<br />

but a nononcoretrovirus, can stably transduce joint tissues but also<br />

inserts the cDNA into the cellular genome which has been associated<br />

with disease from insertional mutagenesis[10]. While lentivirus can<br />

result in long-term expression, the complexity of their biology and<br />

safety concerns have lessened the enthusiasm for their use. Much<br />

excitement has been generated for use of AAV vectors in intra-articular<br />

gene therapy in the last decade. The viral vector appears to safely and<br />

efficiently transduce joint tissues and while it does not incorporate<br />

into the cellular genome, it stays episomally and results in long-term<br />

production of protein[1, 4, 11]. Advances in the biology of AAV have<br />

also made the vector more efficient as in its wild type state it exists<br />

as a single stranded DNA virus that counts on the cellular machinery<br />

to produce protein. Advances in self-complementary AAV viral vectors<br />

(scAAV) have overcome this barrier and resulted in much more efficient<br />

intra-articular vectors[12, 13]. To date, there have been six clinical trials<br />

of in-vivo gene therapy, 4 of which are published[1]. Two clinical trials<br />

have been performed using retrovirus to deliver IL-1ra[9, 14], two<br />

clinical trials have been performed using AAV to deliver TNFR-Fc[4,<br />

11], one has used plasmid HSV-tk (unpublished) and one using NFkB<br />

oligonucleotide (unpublished). While initial results of clinical trials<br />

have shown promise, the field has been troubled by FDA mandates<br />

of gene therapy trials to be either terminated or put on hold due to<br />

complications of patients in trials until they have been successfully<br />

proven to be gene therapy related or not. In one trial in which a subject<br />

died that was injected into the circulation of the liver proved to be a<br />

large barrier to getting gene therapy back on track[15]. Another trial in<br />

which a subject died after a high dose intra-articular injection of AAV<br />

again put progress on hold until the death was concluded to be non<br />

treatment related[16]. Further clinical trials both in humans and animals<br />

are on the forefront of advancement of gene therapy. Most likely the<br />

few clinical trials published along with the large body of pre-clinical<br />

data that exist on the utility of gene therapy for nonfatal diseases such<br />

as osteoarthritis, cartilage degeneration and rheumatoid arthritis will<br />

prove that this therapy will most likely conquer most concerns related<br />

to safety and efficacy. Since no highly effective drugs exist for the<br />

treatment of OA and over 40 million people suffer from this disease, an<br />

efficacious treatment would be a welcomed clinical entity. Currently,<br />

gene therapy is experiencing resurgence, especially in the field of<br />

arthritis. Many animal models reveal efficacy and enthusiasm exists<br />

for continued testing to treat both humans and animals that suffer<br />

from cartilage damage and osteoarthritis[7, 8, 12, 17, 18]. The field will<br />

continue to experience significant hurdles such as the need to prove<br />

safety and efficacy, as well as adequate funding. This will most likely<br />

slow progress but the current data indicate great promise and most<br />

certainly gene therapy will overcome significant hurdles and achieve<br />

success in the face of various challenges.<br />

References:<br />

1. Evans CH, Ghivizzani SC, Robbins PD (2011). Getting arthritis gene<br />

therapy into the clinic. Nat Rev Rheumatol 7: 244-249.<br />

2. Gelse K, von der Mark K, Schneider H (2003). <strong>Cartilage</strong> regeneration<br />

by gene therapy. Current gene therapy 3: 305-317.<br />

3. Robbins PD, Evans CH, Chernajovsky Y (2003). Gene therapy for<br />

arthritis. Gene Ther 10: 902-911.<br />

4. Mease PJ, et al. (2010). Safety, tolerability, and clinical outcomes<br />

after intraarticular injection of a recombinant adeno-associated vector<br />

containing a tumor necrosis factor antagonist gene: results of a phase<br />

1/2 Study. The Journal of rheumatology 37: 692-703.<br />

5. Gouze E, Gouze JN, Palmer GD, Pilapil C, Evans CH, Ghivizzani SC<br />

(2007). Transgene persistence and cell turnover in the diarthrodial<br />

joint: implications for gene therapy of chronic joint diseases. Mol Ther<br />

15: 1114-1120.<br />

6. Ghivizzani SC, et al. (2008). Perspectives on the use of gene therapy<br />

for chronic joint diseases. Current gene therapy 8: 273-286.<br />

7. Goodrich LR, Brower-Toland BD, Warnick L, Robbins PD, Evans CH,<br />

Nixon AJ (2006). Direct adenovirus-mediated IGF-I gene transduction<br />

of synovium induces persisting synovial fluid IGF-I ligand elevations.<br />

Gene Ther 13: 1253-1262.<br />

8. Nixon AJ, et al. (2007). Gene therapy in musculoskeletal repair. Ann<br />

N Y Acad Sci 1117: 310-327.


9. Evans CH, et al. (2005). Gene transfer to human joints: progress<br />

toward a gene therapy of arthritis. Proceedings of the National Academy<br />

of Sciences of the United States of America 102: 8698-8703.<br />

10. Kohn DB, Sadelain M, Glorioso JC (2003). Occurrence of leukaemia<br />

following gene therapy of X-linked SCID. Nat Rev Cancer 3: 477-488.<br />

11. Mease PJ, et al. (2010). Efficacy and safety of retreatment in patients<br />

with rheumatoid arthritis with previous inadequate response to tumor<br />

necrosis factor inhibitors: results from the SUNRISE trial. The Journal of<br />

rheumatology 37: 917-927.<br />

12. Kay JD, et al. (2009). Intra-articular gene delivery and expression<br />

of interleukin-1Ra mediated by self-complementary adeno-associated<br />

virus. The journal of gene medicine 11: 605-614.<br />

13. Ishihara A, Bartlett JS, Bertone AL (2012). Inflammation and<br />

immune response of intra-articular serotype 2 adeno-associated virus<br />

or adenovirus vectors in a large animal model. Arthritis 2012: 735472.<br />

14. Wehling P, et al. (2009). Clinical responses to gene therapy in joints<br />

of two subjects with rheumatoid arthritis. Human gene therapy 20: 97-<br />

101.<br />

15. Raper SE, et al. (2003). Fatal systemic inflammatory response<br />

syndrome in a ornithine transcarbamylase deficient patient following<br />

adenoviral gene transfer. Molecular genetics and metabolism 80: 148-<br />

158.<br />

16. Frank KM, et al. (2009). Investigation of the cause of death in a<br />

gene-therapy trial. The New England journal of medicine 361: 161-169.<br />

17. Morisset S, Frisbie DD, Robbins PD, Nixon AJ, McIlwraith CW (2007).<br />

IL-1ra/IGF-1 gene therapy modulates repair of microfractured chondral<br />

defects. Clin Orthop Relat Res 462: 221-228.<br />

18. Frisbie DD, Ghivizzani SC, Robbins PD, Evans CH, McIlwraith CW<br />

(2002). Treatment of experimental equine osteoarthritis by in vivo<br />

delivery of the equine interleukin-1 receptor antagonist gene. Gene<br />

Ther 9: 12-20.<br />

Acknowledgments:<br />

K08AR054903-01A2 and Grayson Jockey Club Foundation<br />

15.1.1<br />

Platelet rich plasma: Overview of current knowledge: hope, hype<br />

and reality.<br />

L.A. Fortier 1 , T. Mccarrell 2 , B.J. Cole 2 , S. Boswell 1 , L.V. Schnabel 1<br />

1 Ithaca/United States of America, 2 Chicago/United States of<br />

America<br />

Introduction: Platelet concentrates such as platelet-rich plasma<br />

(PRP) have gained popularity in sports medicine and orthopedics to<br />

promote accelerated physiological healing and return to function.<br />

The concept that PRP can improve joint or tendon disease is based on<br />

the physiologic role of platelets and their contained growth factors<br />

in wound healing. However, PRP is comprised of all substances in<br />

blood and components in this milieu have bioactive functions that<br />

positively and negatively affect musculoskeletal tissue regeneration<br />

and healing. Mixed reports of success have been reported after<br />

use of PRP in sports medicine, but with the field in its infancy,<br />

there are sufficiently positive outcome data to continue use of and<br />

investigations into PRP.<br />

Content: In sports medicine there are numerous clinical objectives<br />

motivating the use of PRP including promotion of tissue regeneration<br />

in both bony and soft tissues, prevention and treatment of infection,<br />

and restoration of function. Reviews on the basic science and clinical<br />

indications of PRP are common. Originally, PRP was considered as a<br />

method to deliver platelets and therefore growth factors. This lead to<br />

the common thought that more is better, leading to a race amongst<br />

manufacturers to develop systems that would increase platelet<br />

concentrations to a greater level compared to their competitors.<br />

However, concerns were raised about the increase in leukocytes<br />

in some preparations leading to the concept that PRP is a milieu<br />

of all blood components and not simply a means of growth factor<br />

delivery. Ex vivo studies indicated that concentrations of leukocytes<br />

in PRP were directly correlated to loss of normal tendon matrix<br />

Extended Abstracts 143<br />

and an increase in inflammatory molecules. It is unclear what the<br />

clinical ramifications of increased leukocytes are in clinical patients<br />

because the majority of studies do not report leukocyte or platelet<br />

concentration in PRP being administered to the patient. This raises<br />

another significant concern because PRP is not always generated<br />

from a patient’s blood. There are numerous factors that affect a<br />

patient’s platelet count including nutrition, hydration, smoking, and<br />

natural diurnal variation. This makes it important that at a minimum,<br />

PRP be tested for platelet counts when performing clinical studies so<br />

that outcome can be related to the PRP product delivered. Ideally,<br />

leukocyte counts in the PRP product are determined as well.<br />

The hype over PRP in North America began in early 2009 when two<br />

famous athletes received PRP injections and successfully returned<br />

to professional athletics earlier than anticipated if they were treated<br />

with conventional therapy. PRP was being used and reported on<br />

in small case series prior to this time, but this is when the real<br />

media blitz began, and PRP became of interest to a broad range of<br />

physicians and patients. There are few level 1 studies and several<br />

level 2 or 3 studies that have mixed results regarding the efficacy of<br />

PRP for treatment of musculoskeletal ailments including joint pain,<br />

patellar tendonitis, Achilles tendonosis, and epicondylitis. Clinical<br />

observation and opinion suggests that pain relief and restoration<br />

of function occur more rapidly than expected for some orthopedic<br />

problems with the use of PRP. This has led to investigations of antinociceptive<br />

and anti-inflammatory properties of PRP in our laboratory<br />

and others. Our data indicates that in patients with osteoarthritis,<br />

PRP does decrease the production of pro-inflammatory markers of<br />

pain such as tumor necrosis factor which supports the concept that<br />

PRP functions to decrease pain and inflammation. A by-product of<br />

decreasing inflammation would be joint preservation, but there is<br />

no clinical data to suggest that PRP increases production of cartilage<br />

extracellular matrix proteins such as aggrecan or type II collagen.<br />

In summary, PRP is a useful regenerative therapy to address many<br />

musculoskeletal injuries. It is important to understand that PRP is<br />

more than just platelets and that it contains many bioactive factors that<br />

act in anabolic, catabolic, pro-inflammatory, and anti-inflammatory<br />

pathways. The precise combination and concentration of platelets,<br />

leukocytes, and other plasma components best for musculoskeletal<br />

healing is not presently known, and clinicians should be aware that the<br />

effects of PRP are not solely based on platelet concentration. Finally,<br />

it is imperative for those involved in clinical study design to take into<br />

consideration diurnal variation in platelet count and to understand that<br />

some patients in some instances will simply fail to generate PRP. This<br />

information is important for assessment of clinical outcome.<br />

References:<br />

1. Rodeo SA, Delos D, Weber A, et al. What’s new in orthopaedic<br />

research. J Bone Joint Surg Am 2010;92:2491-501.<br />

2. Schnabel LV, Mohammed HO, Miller BJ, et al. Platelet rich plasma<br />

(PRP) enhances anabolic gene expression patterns in flexor digitorum<br />

superficialis tendons. J Orthop Res 2007;25:230-40.<br />

3. Paoloni J, De Vos RJ, Hamilton B, Murrell GA, Orchard J. Plateletrich<br />

plasma treatment for ligament and tendon injuries. Clin J Sport<br />

Med 2011;21:37-45.<br />

4. Peerbooms JC, Sluimer J, Bruijn DJ, Gosens T. Positive effect of an<br />

autologous platelet concentrate in lateral epicondylitis in a doubleblind<br />

randomized controlled trial: platelet-rich plasma versus<br />

corticosteroid injection with a 1-year follow-up. Am J Sports Med<br />

2010;38:255-62.<br />

5. Lopez-Vidriero E, Goulding KA, Simon DA, Sanchez M, Johnson DH.<br />

The use of platelet-rich plasma in arthroscopy and sports medicine:<br />

optimizing the healing environment. Arthroscopy 2010;26:269-78.<br />

6. McCarrel T, Fortier L. Temporal growth factor release from plateletrich<br />

plasma, trehalose lyophilized platelets, and bone marrow<br />

aspirate and their effect on tendon and ligament gene expression. J<br />

Orthop Res 2009;27:1033-42.<br />

7. Dohan Ehrenfest DM, Bielecki T, Corso MD, Inchingolo F,<br />

Sammartino G. Shedding light in the controversial terminology<br />

for platelet-rich products: Platelet-rich plasma (PRP), platelet-rich<br />

fibrin (PRF), platelet-leukocyte gel (PLG), preparation rich in growth<br />

factors (PRGF), classification and commercialism. J Biomed Mater<br />

Res A 2010;.<br />

8. Senzel L, Gnatenko DV, Bahou WF. The platelet proteome. Curr<br />

Opin Hematol 2009;16:329-33.<br />

9. Foster TE, Puskas BL, Mandelbaum BR, Gerhardt MB, Rodeo SA.<br />

Platelet-rich plasma: from basic science to clinical applications. Am J<br />

Sports Med 2009;37:2259-72.


144<br />

Extended Abstracts<br />

15.1.2<br />

Platelet rich plasma and joint tissue repair<br />

E. Kon, G. Filardo, B. Di Matteo, A. Di Martino, M. Marcacci<br />

Bologna/Italy<br />

Introduction: Definition and biological rational Orthopaedic and<br />

rheumatologic physicians must everyday face the complex issue of<br />

cartilage pathology, whose prevalence (1,2,3) is rapidly increasing due<br />

to the massive involvement in sport activity by the entire population,<br />

from the youngs to the middle-aged and even elder individuals, all<br />

moved by the awareness of the importance of physical activity as<br />

a preventive medical approach. Despite the positive aspects of<br />

this widespread life-style, new medical problems have emerged: in<br />

particular, cartilage lesions are becoming one of the most important<br />

challenge for both basic researchers and clinicians. In fact, although<br />

being able to sustain huge mechanical stresses, cartilage has limited<br />

healing potential due to several reasons: first of all, the relative<br />

isolation from systemic regulation caused by the lack of nerves and<br />

vessels when compared to other tissues. Furthermore, its complex<br />

histological structure, consisting of chondrocytes surrounded by<br />

matrix made of a specialized framework of collagen, aggrecans<br />

and fluid, determines an intrinsic vulnerability that, starting from<br />

small and focal lesions, can evolve in an accelerated degenerative<br />

process whose final stage is osteoarthritis (OA), a chronic condition<br />

difficult to treat by conservative means and often requiring a surgical<br />

approach such as arthroplasty. Several treatment, both conservative<br />

and surgical, could address cartilage pathology (4,5,6) and the<br />

therapeutic decision has to be taken according to the specific<br />

case and the eventual presence of other comorbidities (meniscal<br />

status, axial alignment, etc..). Besides traditional pharmacological<br />

approaches (paracetamol, NSAIDs, opioids), physiotherapy and<br />

recognized injective treatments (i.e.,viscosupplementation or<br />

corticosteroids), some new trends have emerged in the last years,<br />

opening a large and sometimes controversial research topic: the<br />

application of blood-derivatives for joint degenerative pathology.<br />

Recently, a blood-derived product, platelet-rich plasma (PRP), has<br />

gained increasing attention as a promising procedure to stimulate<br />

cartilage repair. PRP is an autologous concentrate of plateletderived<br />

growth factors (GFs) and other molecules, obtained directly<br />

from the peripheral venous blood of the patient. GFs are a group<br />

of polypeptides playing a fundamental role in the regulation of<br />

growth and development of several tissues, including cartilage. The<br />

biological rational of PRP is that platelets contain storage pools of GFs<br />

including: platelet-derived growth factor (PDGF); transforming growth<br />

factor (TGF-β); platelet-derived epidermal growth factor (PDEGF);<br />

vascular endothelial growth factor (VEGF); insulin-like growth factor<br />

1 (IGF-1); fibroblastic growth factor (FGF); and epidermal growth<br />

factor (EGF) (7, 8). Besides GFs, alpha granules are also a source of<br />

cytokines, chemokines and many other mediators (1) all involved<br />

in complex biological mechanisms stimulating chemotaxis, cell<br />

proliferation and maturation, modulating inflammatory molecules<br />

and attracting leukocytes (1). Platelets also store dense granules,<br />

rich of ADP, ATP, calcium ions, histamine, serotonin and dopamine,<br />

which play a complex role in tissue modulation and regeneration<br />

(9). Finally, platelets contain lisosomal granules which can secrete<br />

acid hydrolases, cathepsin D and E, elastases and lysozyme<br />

(10,11), and most likely other not yet well characterized molecules,<br />

the role of which in tissue healing should not be underestimated.<br />

Several in vitro and in vivo animal studies showed the potential<br />

beneficial effect of PRP in promoting cellular anabolism and tissue<br />

regeneration (12, 13). This fascinating regenerative approach has led<br />

to some urging controversies among the scientific community. First<br />

of all, the lack of universal definition: in general, PRP is regarded as<br />

a blood derivative generated by differential centrifugation of whole<br />

blood, with a higher concentration of platelets compared to basal<br />

level, commonly about 400% of the peripheral blood count (14,15).<br />

However, in the literature PRP concentrations have been reported to<br />

range widely, from 4 to 8 times than those found in whole blood (18),<br />

and good results have been reported also with lower concentrations<br />

(16,17). Furthermore, several different procedures have been<br />

described to obtain PRP, thus implying the existence of qualitative<br />

and quantitative differences among substances used in various preclinical<br />

and clinical studies. In particular, the presence of leukocytes<br />

in the final product has been considered a crucial point, due to their<br />

potential negative pro-inflammatory effect. Further differences lie<br />

in the storage modalities and the activation methods, thus adding<br />

other variables to consider when comparing clinical results reported<br />

in literature (18).<br />

Content: Clinical application Besides the application in cartilage<br />

degenerative pathology, platelet derived growth factors have been<br />

applied in a large number of clinical conditions both as a mere<br />

conservative treatment or as a “biological augmentation” during<br />

surgical procedures. The range of pathologies treated with PRP<br />

comprehends a remarkable variety of tendinopathies (19, 20, 21),<br />

muscle lesions and even anterior cruciate ligament (ACL)<br />

reconstruction (14). For what concerns cartilage pathology, PRP<br />

application has been investigated in several studies. Also in this<br />

case conservative and surgical approaches have been attempted.<br />

With respect to the surgical approach the first paper, a case report,<br />

was published in 2003 by Sanchez et al. (22) concerning a 12 years<br />

old football player with a large (> 2 cm2) articular cartilage avulsion<br />

in the knee and treated arthroscopically with re-attachment of the<br />

chondral fragment and platelet concentrate injection at the fragment<br />

center and all around the interface with the healthy surrounding<br />

tissue. Clinical results were excellent with full functional recovery<br />

and resumption of the sport practice after 18 weeks. The patient<br />

returned to the same pre-injury level just a few weeks later. Another<br />

surgical application of PRP was explored by Dhollander et al. (23)<br />

who treated 5 osteochondral defects of the patella with a combined<br />

collagen I/III scaffold membrane implant on the prepared lesion site<br />

and PRP injection underneath. Clinical results at 24 months of followup<br />

were satisfying and even MRI evaluation showed good quality of<br />

the repair tissue, but a clear role of the platelet concentrate couldn’t<br />

be demonstrated. A further study of Siclari et al. (24) proved the<br />

efficacy of a polyglycolic acid/hyaluronan scaffold immersed in PRP<br />

for treating full-thickness chondral defects of the knee: 52 patients<br />

were treated arthroscopically and evaluated at 1 year follow-up<br />

obtaining a significant clinical improvement. Five biopsies were also<br />

performed revealing an homogeneous, well integrated repair tissue.<br />

Giannini et al. (25) applied an innovative technique in treating focal<br />

osteochondral talar lesions, using an autologous preparation<br />

consisting of bone marrow concentrate and PRP applied on a<br />

hyaluronan scaffold or mixed with collagen powder. They treated 48<br />

patients through an arthroscopic single-step procedure and reported<br />

their results at 24 months of follow-up: clinical score registered a<br />

sensible increase with most of the patients coming back to sport<br />

activity. In light of these studies, PRP seems an effective biological<br />

enhancer for acellular scaffolds used in treating chondral defects.<br />

Another study (26), however, pointed out that, at least in the animal<br />

model, PRP could even decrease the regenerative potential of an<br />

osteochondral biomimetic scaffold. PRP intra-articular injective<br />

treatment has been the subject of nine studies to date, mostly<br />

focusing on knee application. In 2008 Sanchez et al. published a<br />

retrospective observational study on 60 patients (27) comparing<br />

platelet concentrate and HA. Patients from both groups underwent<br />

three injections one week apart and were evaluated basally and at 5<br />

weeks of follow up. Results were encouraging, with a better pain<br />

control in PRP group, even though the short follow-up is an important<br />

limitation. In 2010 Sampson et al. published a study (28) on 14<br />

patients with primary or secondary OA of the knee and previous<br />

unsuccessful treatments. The patients received 3 PRP injections one<br />

month apart and were evaluated at basal level and up to one year of<br />

follow-up. A statistically significant clinical improvement was<br />

documented , with a reduction of the pain both at rest and during<br />

physical activity. In 2010 Wang-Saegusa et al. (29) reported their<br />

results after treating 261 patients, affected by uni- or bilateral knee<br />

OA, with three injections of platelet concentrate two weeks apart.<br />

Prospective evaluation up to 6 months was performed and also in<br />

this case statistical analysis revealed significant results with increase<br />

in all the scores applied. Kon et al. published in 2009 a prospective<br />

study (30) on 91 patients (for a total of 115 knees) treated with three<br />

injections of PRP (one every three weeks). Inclusion criteria were:<br />

clinical history of knee pain or articular swelling lasting more than 4<br />

months, radiographic or MRI signs of OA or cartilage degenerative<br />

lesions. Patients underwent clinical evaluation at basal level and at<br />

2, 6, and 12 months of follow-up. Eighty percent of the patients<br />

treated expressed satisfaction for the treatment received. Clinical<br />

outcome registered a statistically relevant improvement in all the<br />

variables considered just after 2 months from the end of the<br />

treatment. These results were later confirmed at 6 months of followup,<br />

whereas a tendency of worsening was reported from 6 to 12<br />

months of follow-up. Despite the decrease reported after one year,<br />

the clinical scores at that time were still higher than the basal level.<br />

Some influencing factors were detected: in particular it was observed<br />

that young male patients are the best responding group, especially<br />

in case of simple chondropathy without signs of OA. A later study by<br />

the same authors (31) evaluating the patients at 24 months of followup<br />

confirmed this trend with a further decrease in the clinical<br />

outcome thus concluding that intra-articular therapy with plateletderived<br />

GFs is time dependent with an average duration of 9 months<br />

and better and long lasting results in younger patients with lower<br />

level of joint degeneration. In another multi-center study carried on<br />

by Kon et al. (32), the clinical effectiveness of PRP was compared to<br />

low molecular weight HA (LWHA) and high molecular weight HA<br />

(HWHA). Three homogeneous groups of patients were respectively<br />

treated with 3 weekly injections of PRP, LWHA, or HWHA. The results


evidenced a better performance for PRP group at 6 months of followup.<br />

In particular, subgroups analysis (chondropathy vs early vs<br />

severe OA) revealed again that, in the chondropathy group, PRP<br />

gave markedly higher results than HA at 6 months of follow-up,<br />

whereas in the early OA group the gap in favor of PRP is reduced and<br />

in the severe OA subgroup no difference in clinical outcome was<br />

observed between treatments. Furthermore, patients under 50 years<br />

old have greater chance to benefit from this biological approach<br />

with GFs supplementation. Finally, a comparative study between<br />

PRP with or without leukocytes used to treat 144 patients affected<br />

by knee cartilage pathology has recently been published by Filardo<br />

et al. showing comparable positive clinical effects with both<br />

treatments, with PRP-leukocyte group suffering from more swelling<br />

and pain reaction after the injections (33). Injective application of<br />

PRP has been tested also in case of hip osteoarthritis and<br />

osteochondral lesions of the talus. A pilot study led by Battaglia et<br />

al. (34) reported the results of PRP ultra-sound guided injective<br />

treatment in 20 patients affected by hip OA. At short term evaluation,<br />

clinical outcome was positive but a gradual worsening occurred up<br />

to one year of follow-up. Lastly, a prospective study of Mei-Dan et al.<br />

(35) compared the efficacy of HA and PRP in 30 patients (15 per<br />

group) affected by talar osteochondral lesions and evaluated up to<br />

28 weeks of follow-up. Results were statistically significant and PRP<br />

proved to be more effective in controlling pain and re-establishing<br />

function. Conclusions Many studies have been published on PRP<br />

application for joint tissues repair and there is evidence about the<br />

safety of this method either through injections or during surgical<br />

procedures. The majority of these studies reveals interesting<br />

preliminary results for cartilage degenerative pathology and also for<br />

tendinopathies, but too many are still the questions to clarify,<br />

concerning the real biological potential, the indications, and the<br />

application methods. Many aspects have still to be understood also<br />

in terms of basic science. A “miracle aura” has arisen in the last<br />

years toward PRP application but the literature is still controversial<br />

and further high quality, randomised control trials are needed to<br />

endorse this therapy as a real valid option among joint regenerative<br />

procedures.<br />

References:<br />

1. Curl WW, Krome J, Gordon ES, Rushing J, Smith BP, Poehling<br />

GG. <strong>Cartilage</strong> injuries: a review of 31,516 knee arthroscopies.<br />

Arthroscopy. 1997 Aug;13(4):456-60<br />

2. Widuchowski W, Widuchowski J, Trzaska T (2007) Articular<br />

cartilage defects: study of 25,124 knee arthroscopies. Knee 14:177-<br />

182<br />

3. Peers KH, Lysens RJ: Patellar tendinopathy in athletes: current<br />

diagnostic and therapeutic recommendations. Sports Med.<br />

2005;35:71-87.<br />

4. Kon E, Filardo G, Drobnic M, Madry H, Jelic M, van Dijk N, Della<br />

Villa S. Non-surgical management of early knee osteoarthritis. Knee<br />

Surg Sports Traumatol Arthrosc. 2011 Oct 25. (Epub ahead of print)<br />

5. Gomoll AH, Filardo G, de Girolamo L, Esprequeira-Mendes J,<br />

Marcacci M, Rodkey WG, Steadman RJ, Zaffagnini S, Kon E. Surgical<br />

treatment for early osteoarthritis. Part I: cartilage repair procedures.<br />

. Knee Surg Sports Traumatol Arthrosc. 2011 Nov 24. (Epub ahead<br />

of print)<br />

6. Gomoll AH, Filardo G, Almqvist FK, Bugbee WD, Jelic M, Monllau JC,<br />

Puddu G, Rodkey WG, Verdonk P, Verdonk R, Zaffagnini S, Marcacci<br />

M Surgical treatment for early osteoarthritis. Part II: allografts and<br />

concurrent procedures. Knee Surg Sports Traumatol Arthrosc. 2011<br />

Nov 9. (Epub ahead of print)<br />

7.Foster TE, Puskas BL, Mandelbaum BR, Gerhardt MB, Rodeo SA.<br />

Platelet-rich plasma: from basic science to clinical applications. Am J<br />

Sports Med. 2009 Nov;37(11):2259-72.<br />

8.Sanchez AR, Sheridan PJ, Kupp LI. Is platelet-rich plasma the<br />

perfect enhancement factor? A current review. Int J Oral Maxillofac<br />

Implants 2003;18:93<strong>–</strong>103.<br />

9. Mishra A, Woodall J Jr, Vieira A. Treatment of tendon and muscle<br />

using platelet-rich plasma. Clin Sports Med. 2009 Jan;28(1):113-25.<br />

10. Anitua E, Andia I, Ardanza B, Nurden P, Nurden AT. Autologous<br />

platelets as a source of proteins for healing and tissue regeneration.<br />

Tromb Haemost 2004;91:4-15.<br />

Extended Abstracts 145<br />

11. Senet P, Bon FX, Benbunan M, Bussel A, Traineau R, Calvo F,<br />

Dubertret L, Dosquet C. Randomized trial and local biological effect<br />

of autologous platelets used as adjuvant therapy for chronic venous<br />

leg ulcers. J Vasc Surg 2003;38:1342-8<br />

12. Kon E, Filardo G, Di Martino A, Marcacci M. Platelet-rich plasma<br />

(PRP) to treat sports injuries: evidence to support its use Knee Surg<br />

Sports Traumatol Arthrosc. 2011 Apr;19(4):516-27. Epub 2010 Nov<br />

17.<br />

13. Torricelli P, Fini M, Filardo G, Tschon M, Pischedda M, Pacorini<br />

A, Kon E, Giardino R. Regenerative medicine for the treatment of<br />

musculoskeletal overuse injuries in competition horses. Int Orthop.<br />

2011 Oct;35(10):1569-76. doi: 10.1007/s00264-011-1237-3. Epub<br />

2011 Mar<br />

14. Marx R. Platelet rich plasma (PRP): what is PRP and what is not<br />

PRP? Implant Dent 2001;10:225<strong>–</strong>8.<br />

15. Marx RE. Platelet-rich plasma: evidence to support its use. J Oral<br />

Maxillofac Surgery 2004;62:489<strong>–</strong>96.<br />

16. Creaney L, Hamilton B.Growth factor delivery methods in the<br />

management of sports injuries: the state of play. Br J Sports Med.<br />

2008 May;42(5):314-20.<br />

17. Sánchez M, Anitua E, Azofra J, Andía I, Padilla S, Mujika I.<br />

Comparison of surgically repaired Achilles tendon tears using plateletrich<br />

fibrin matrices. Am J Sports Med. 2007 Feb;35(2):245-51.<br />

18. Tschon M, Fini M, Giardino R, Filardo G, Dallari D, Torricelli P,<br />

Martini L, Giavaresi G, Kon E, Maltarello MC, Nicolini A, Carpi A. Lights<br />

and shadows concerning platelet products for musculoskeletal<br />

regeneration. Front Biosci 2011 Jan 1;3:96107.Review<br />

19. Kon E, Filardo G, Delcogliano M, Presti ML, Russo A, Bondi A,<br />

Di Martino A, Cenacchi A, Fornasari PM, Marcacci M. Platelet-rich<br />

plasma: new clinical application: a pilot study for treatment of<br />

jumper’s knee. Injury. 2009 Jun;40(6):598-603. Epub 2009 Apr 19.<br />

20. Filardo G, Kon E, Della Villa S, Vincentelli F, Fornasari PM, Marcacci<br />

M. Use of platelet-rich plasma for the treatment of refractory jumper’s<br />

knee. Int Orthop. 2010 Aug; 34(6):909-15. Epub 2009 Jul 31.<br />

21. Filardo G, Presti ML, Kon E, Marcacci M. Nonoperative biological<br />

treatment approach for partial Achilles tendon lesion. Orthopedics.<br />

2010 Feb;33(2):120-3. doi: 10.3928/01477447-20100104-31.<br />

22. Sánchez M, Azofra J, Anitua E, Andía I, Padilla S, Santisteban J,<br />

Mujika I. Plasma rich in growth fac tors to treat an articular cartilage<br />

avulsion: a case report. Med Sci Sports Exerc. 2003 Oct;35(10):1648-<br />

52.<br />

23. Dhollander AA, De Neve F, Almqvist KF, Verdonk R, Lambrecht<br />

S, Elewaut D, Verbruggen G, Verdonk PC Autologous matrix-induced<br />

chondrogenesis combined with platelet-rich plasma gel: technical<br />

description and a five pilot patients report. Knee Surg Sports<br />

Traumatol Arthrosc. 2011 Apr;19(4):536-42. Epub 2010 Dec 11.<br />

24. Siclari A, Mascaro G, Gentili C, Cancedda R, Boux E.A Cell-free<br />

Scaffold-based <strong>Cartilage</strong> <strong>Repair</strong> Provides Improved Function Hyalinelike<br />

<strong>Repair</strong> at One year. Clin Orthop Relat Res. 2012 Mar;470(3):910-<br />

9. Epub 2011 Oct 1.<br />

25. Clin Orthop Relat Res. 2009 Dec;467(12):3307-20. Epub 2009<br />

May 16.One-step bone marrow-derived cell transplantation in talar<br />

osteochondral lesions.Giannini S, Buda R, Vannini F, Cavallo M,<br />

Grigolo B.<br />

26. Kon E, Filardo G, Delcogliano M, Fini M, Salamanna F, Giavaresi G,<br />

Martin I, Marcacci M. Platelet autologous growth factors decrease the<br />

osteochondral regeneration capability of a collagen-hydroxyapatite<br />

scaffold in a sheep model. BMC Musculoskelet Disord. 2010 Sep<br />

27;11:220.<br />

27. Sánchez M, Anitua E, Azofra J, Aguirre JJ, Andia I . Intra-articular<br />

injection of an autolo gous preparation rich in growth factors for<br />

the treatment of knee OA: a retrospective cohort study. Clin Exp<br />

Rheumatol. 2008 Sep-Oct;26(5):910-3.<br />

28. Sampson S, Reed M, Silvers H, Meng M, Mandelbaum B. Injection<br />

of platelet-rich plasma in patients with primary and secondary<br />

knee osteoarthritis: a pilot study. Am J Phys Med Rehabil. 2010<br />

Dec;89(12):961-9.


146<br />

Extended Abstracts<br />

29. Wang-Saegusa A, Cugat R, Ares O, Seijas R, Cuscó X, Garcia-<br />

Balletbó M. Infiltration of plasma rich in growth factors for<br />

osteoarthritis of the knee short-term effects on function and quality<br />

of life. Arch Orthop Trauma Surg. 2011 Mar;131(3):311-7. Epub 2010<br />

Aug 17.<br />

30. Kon E, Buda R, Filardo G, Di Martino A, Timoncini A, Cenacchi<br />

A, Fornasari PM, Giannini S, Marcacci M. Platelet-rich plasma: intraarticular<br />

knee injections produced favorable results on degenerative<br />

cartilage lesions. Knee Surg Sports Traumatol Arthrosc. 2010<br />

Apr;18(4):472-9. Epub 2009 Oct 17.<br />

31. Filardo G, Kon E, Buda R, Timoncini A, Di Martino A, Cenacchi<br />

A, Fornasari PM, Giannini S, Marcacci M. Platelet-rich plasma intraarticular<br />

knee injections for the treatment of degenerative cartilage<br />

lesions and osteoarthritis. Knee Surg Sports Traumatol Arthrosc.<br />

2011 Apr;19(4):528-35. Epub 2010 Aug 26.<br />

32. Kon E, Mandelbaum B, Buda R, Filardo G, Delcogliano M, Timoncini<br />

A, Fornasari PM, Giannini S, Marcacci M. Platelet-Rich Plasma Intra-<br />

Articular Injection Versus HyaluronicAcid Viscosupplementation<br />

as Treatments for <strong>Cartilage</strong> Pathology: From Early Degeneration to<br />

Osteoarthritis. Arthroscopy. 2011 Aug 8. (Epub ahead of print)<br />

33. Filardo G, Kon E, Pereira Ruiz MT, Vaccaro F, Guitaldi R, Di<br />

Martino A, Cenacchi A, Fornasari PM, Marcacci M.Platelet-rich<br />

plasma intra-articular injections for cartilage degeneration and<br />

osteoarthritis: single- versus double-spinning approach. Knee Surg<br />

Sports Traumatol Arthrosc. 2011 Dec 28. (Epub ahead of print)<br />

34.Battaglia M, Guaraldi F, Vannini F, Buscio T, Buda R, Galletti S,<br />

Giannini S. Platelet-rich plasma (PRP) intra-articular ultrasoundguided<br />

injections as a possible treatment for hip osteoarthritis: a<br />

pilot study. Clin Exp Rheumatol. 2011 Jul-Aug;29(4):754.<br />

35. Mei-Dan O, Carmont MR, Laver L, Mann G, Maffulli N, Nyska<br />

MPlatelet-Rich Plasma or Hyaluronate in the Management of<br />

Osteochondral Lesions of the Talus. Am J Sports Med. 2012 Jan 17.<br />

(Epub ahead of print)<br />

Acknowledgments:<br />

We would like to thank Letizia Merli, MD; Francesco Perdisa, MD;<br />

Luca Andriolo, MD; Silvio Patella, MD; Federica Balboni, MS; Giulio<br />

Altadonna, MS; Angela Montaperto, JD, all from Rizzoli Orthopaedic<br />

Institute, Bologna, Italy.<br />

15.1.3<br />

Clinical experiences with Platelet-Rich Plasma<br />

S. Rodeo<br />

New York/United States of America<br />

Introduction: 10th World Congress of the <strong>International</strong> <strong>Cartilage</strong><br />

<strong>Repair</strong> <strong>Society</strong> Clinical Experience with Platelet Rich Plasma Scott<br />

A. Rodeo, MD Co-Chief, Sports Medicine and Shoulder Service<br />

Professor, Orthopaedic Surgery, Weill Medical College of Cornell<br />

University Attending Orthopaedic Surgeon, The Hospital for Special<br />

Surgery Associate Team Physician, New York Giants Football<br />

This talk will review recent clinical data on the use of PRP for tissues<br />

in and around the synovial joint, including hyaline cartilage, ligament,<br />

tendon, and meniscus. Cytokines are known to have important and<br />

fundamental roles in connective tissue biology Cytokines affect: -<br />

cell proliferation - matrix synthesis - angiogenesis - chemotaxis<br />

Connective tissue healing requires a complex timing and sequence<br />

of cytokine expression à thus, single factor therapy has distinct<br />

limitations The rationale and attraction of PRP is the ability to deliver<br />

numerous cytokines in physiologically-relevant proportions Despite<br />

vast basic science and laboratory data demonstrating a positive<br />

effect of various PRP formulations on basic cell biology, this has not<br />

translated into a consistently positive clinical effect. A fundamental<br />

limitation in studying PRP is the fact that there is tremendous<br />

variability in various commercially-available PRP formulations.<br />

There are variations in: • Platelet recovery • Inclusion of WBCs •<br />

Platelet activation (thrombin, Ca+) • Kinetics of cytokine release<br />

from PRP/PRFM • Preservation/function of the platelets and WBCs<br />

• Ratio between fibrinogen and thrombin concentration • Formation<br />

of a fibrin matrix (fibrin polymerization) • Microstructure of the final<br />

fibrin network (ability to trap cytokines and other bioactive factors)<br />

There is also variability with an individual:<br />

• Platelet counts vary markedly between patients<br />

• Platelet counts vary day to day within an individual<br />

• Growth factor content per platelet<br />

• Other proteins/factors in the plasma<br />

• Another important limitation in the use of PRP:<br />

Exogenous growth factor therapy often only improves the structural<br />

properties of tissues by inducing more (scar) tissue formation, while<br />

NOT improving the material properties<br />

A large body of data shows that cytokines can increase production of<br />

matrix proteins BUT a critical deficiency is that tissue microstructure<br />

is not reformed Thus, it appears that growth factors (PRP) still do<br />

not provide the proper cellular and molecular signals to drive<br />

regenerative healing I hypothesize that both pluripotent cells AND<br />

appropriate signals (cytokines) are needed to reconstitute tissue<br />

composition and structure Summary of major points for each<br />

different tissue type: Articular cartilage/osteoarthritis A number<br />

of studies have demonstrated clinical improvement in symptoms<br />

following PRP injections in the knee. There is very little data available<br />

for joints other than the knee. Most studies report better results in<br />

younger patients with lesser degrees of degeneration.<br />

The clinical effect typically wears off after 6-12 months. There is very<br />

little (virtually zero) data that has demonstrated a positive structural<br />

effect (actual regeneration of cartilage tissue). Very few studies that<br />

have compared to intra-articular steroid or hyaluronic acid. Kon et<br />

al (Arthroscopy 2011) reported PRP superior to hyaluronic acid in<br />

younger patients and with earlier grade of OA.<br />

PRP releasate may inhibit the adverse effects of IL-1β and other<br />

negative factors in the inflammatory environment.<br />

Further studies required in this area: 1. Further define best way to<br />

isolate and concentrate IL-1 receptor antagonist (IL-1Ra). 2. Define<br />

the role (positive or negative) of white blood cells when treating<br />

cartilage lesions. 3. Arthritis is very heterogeneous condition: the<br />

effect of a specific PRP formulation may differ significantly based<br />

on the underlying biologic/inflammatory milieu. Patellar Tendon:<br />

There is very little data available on the effect of PRP for patellar<br />

tendonopathy There is some positive data for lateral epicondylitis,<br />

suggesting that PRP may be effective for extra-articular tendons<br />

(such as patellar tendon). One study reports positive results and<br />

demonstrates potential for improvement in MRI appearance of<br />

degenerative patellar tendon (Filardo et al, Int Orthop 2010)<br />

Meniscus:<br />

There is very little data available on the effect of PRP on meniscus<br />

healing. Placement of an exogenous fibrin clot has been used in<br />

conjunction with meniscus repair, and thus the concept of using PRP<br />

during meniscus repair makes sense. Ishida et al showed a positive<br />

effect in both in vitro and in vivo studies (rabbit model). However,<br />

there is very little clinical data available at this time.<br />

ACL:<br />

A number of studies have examined the effect of PRP applied to an<br />

ACL graft and/or the graft-bone interface in the bone tunnel. MRI<br />

has been used to examine signal in the graft, as a reflection of graft<br />

maturation and remodeling. Some studies have shown a positive<br />

effect on graft remodeling. Further studies are required to better<br />

define the potential for PRP to affect either graft-bone healing and<br />

remodeling/maturation of the intra-articular portion of the graft.<br />

Furthermore, the relationship between morphologic changes in the<br />

graft and graft function/knee stability is still unclear.<br />

Content: 10th World Congress of the <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong><br />

<strong>Society</strong> Clinical Experience with Platelet Rich Plasma Scott A. Rodeo,<br />

MD Co-Chief, Sports Medicine and Shoulder Service Professor,<br />

Orthopaedic Surgery, Weill Medical College of Cornell University<br />

Attending Orthopaedic Surgeon, The Hospital for Special Surgery<br />

Associate Team Physician, New York Giants Football<br />

This talk will review recent clinical data on the use of PRP for tissues


in and around the synovial joint, including hyaline cartilage, ligament,<br />

tendon, and meniscus. Cytokines are known to have important and<br />

fundamental roles in connective tissue biology Cytokines affect:<br />

- cell proliferation - matrix synthesis - angiogenesis - chemotaxis<br />

Connective tissue healing requires a complex timing and sequence<br />

of cytokine expression à thus, single factor therapy has distinct<br />

limitations The rationale and attraction of PRP is the ability to deliver<br />

numerous cytokines in physiologically-relevant proportions Despite<br />

vast basic science and laboratory data demonstrating a positive<br />

effect of various PRP formulations on basic cell biology, this has not<br />

translated into a consistently positive clinical effect. A fundamental<br />

limitation in studying PRP is the fact that there is tremendous<br />

variability in various commercially-available PRP formulations.<br />

There are variations in: • Platelet recovery • Inclusion of WBCs •<br />

Platelet activation (thrombin, Ca+) • Kinetics of cytokine release<br />

from PRP/PRFM • Preservation/function of the platelets and WBCs<br />

• Ratio between fibrinogen and thrombin concentration • Formation<br />

of a fibrin matrix (fibrin polymerization) • Microstructure of the final<br />

fibrin network (ability to trap cytokines and other bioactive factors)<br />

There is also variability with an individual:<br />

• Platelet counts vary markedly between patients<br />

• Platelet counts vary day to day within an individual<br />

• Growth factor content per platelet<br />

• Other proteins/factors in the plasma<br />

Another important limitation in the use of PRP:<br />

Exogenous growth factor therapy often only improves the structural<br />

properties of tissues by inducing more (scar) tissue formation, while<br />

NOT improving the material properties<br />

A large body of data shows that cytokines can increase production of<br />

matrix proteins BUT a critical deficiency is that tissue microstructure<br />

is not reformed Thus, it appears that growth factors (PRP) still do<br />

not provide the proper cellular and molecular signals to drive<br />

regenerative healing I hypothesize that both pluripotent cells AND<br />

appropriate signals (cytokines) are needed to reconstitute tissue<br />

composition and structure Summary of major points for each<br />

different tissue type: Articular cartilage/osteoarthritis A number<br />

of studies have demonstrated clinical improvement in symptoms<br />

following PRP injections in the knee. There is very little data available<br />

for joints other than the knee. Most studies report better results in<br />

younger patients with lesser degrees of degeneration. The clinical<br />

effect typically wears off after 6-12 months.<br />

There is very little (virtually zero) data that has demonstrated a<br />

positive structural effect (actual regeneration of cartilage tissue).<br />

Very few studies that have compared to intra-articular steroid or<br />

hyaluronic acid. Kon et al (Arthroscopy 2011) reported PRP superior<br />

to hyaluronic acid in younger patients and with earlier grade of OA.<br />

PRP releasate may inhibit the adverse effects of IL-1β and other<br />

negative factors in the inflammatory environment. Further studies<br />

required in this area: 1. Further define best way to isolate and<br />

concentrate IL-1 receptor antagonist (IL-1Ra). 2. Define the role<br />

(positive or negative) of white blood cells when treating cartilage<br />

lesions. 3. Arthritis is very heterogeneous condition: the effect of<br />

a specific PRP formulation may differ significantly based on the<br />

underlying biologic/inflammatory milieu. Patellar Tendon:<br />

There is very little data available on the effect of PRP for patellar<br />

tendonopathy There is some positive data for lateral epicondylitis,<br />

suggesting that PRP may be effective for extra-articular tendons<br />

(such as patellar tendon). One study reports positive results and<br />

demonstrates potential for improvement in MRI appearance of<br />

degenerative patellar tendon (Filardo et al, Int Orthop 2010)<br />

Meniscus:<br />

There is very little data available on the effect of PRP on meniscus<br />

healing. Placement of an exogenous fibrin clot has been used in<br />

conjunction with meniscus repair, and thus the concept of using PRP<br />

during meniscus repair makes sense. Ishida et al showed a positive<br />

effect in both in vitro and in vivo studies (rabbit model). However,<br />

there is very little clinical data available at this time.<br />

ACL:<br />

A number of studies have examined the effect of PRP applied to an<br />

ACL graft and/or the graft-bone interface in the bone tunnel. MRI<br />

has been used to examine signal in the graft, as a reflection of graft<br />

Extended Abstracts 147<br />

maturation and remodeling. Some studies have shown a positive<br />

effect on graft remodeling. Further studies are required to better<br />

define the potential for PRP to affect either graft-bone healing and<br />

remodeling/maturation of the intra-articular portion of the graft.<br />

Furthermore, the relationship between morphologic changes in the<br />

graft and graft function/knee stability is still unclear.<br />

15.2.2<br />

Hydrogels in cartilage repair<br />

J.D. Kisiday, D.D. Frisbie, L. Goodrich, W. Mcilwraith<br />

Fort Collins/United States of America<br />

Introduction: Hydrogels are a class of polymer materials consisting<br />

of a hydrophilic network that is capable of absorbing water up to<br />

hundreds of times the dry weight of the polymer. The high water<br />

content of hydrogels is a favorable environment for cells as the<br />

polymer network readily allows for diffusion of oxygen, nutrients,<br />

and waste products. As a result, many biocompatible synthetic and<br />

natural polymers that assemble into hydrogels have been explored<br />

as scaffolds for cartilage tissue engineering (1).<br />

Content: Enthusiasm for hydrogels for cartilage repair has been<br />

generated from a long history of the use of hydrogels to study<br />

chondrocyte biology. For example, agarose, a polysaccharide<br />

derived from algae, has been used for nearly 40 years to create<br />

three dimensional suspension cultures that holds chondrocytes<br />

in a round morphology as they exist in cartilage. Chondrocytes<br />

encapsulated in agarose have been use to study a variety of cellular<br />

behaviors, including redifferentiation following dedifferentation to<br />

a fibroblastic phenotype that occurs during monolayer expansion<br />

(2), the response to physical forces (3), and the influence of growth<br />

factors and pro-inflammatory cytokines. In addition, in recent<br />

years agarose has been shown to support chondrogenesis of<br />

various types of progenitor cells in the presence of a chondrogenic<br />

cytokine (4-6) and/or resulting from mechanical loading (7). In both<br />

chondrocyte and progenitor cell studies, the encapsulated cells<br />

have secreted a robust cartilaginous neo-tissue that is retained<br />

within the hydrogel, which lends support to the potential of<br />

hydrogels to host repair in cartilage defects. Given these findings,<br />

the exploration of novel scaffolds for tissue engineering are often<br />

initiated in vitro by designing experiments that are based on<br />

multiweek cultures and cartilaginous extracellular matrix synthesis<br />

as used to study chondrocyte biology. In our lab, we continue to use<br />

agarose hydrogels as control cultures for the development of novel<br />

scaffolds or for characterizing chondrogenic priming technologies for<br />

progenitor cells. Hydrogels lend themselves to ongoing optimization<br />

for cartilage repair by the potential to introduce modifications to the<br />

polymer network. For example, researchers have explored whether<br />

the incorporation of biomolecules enhances cartilaginous neotissue<br />

synthesis. One such approach has been to incorporate the cell<br />

adhesion RGD peptide sequence, which has been added to alginate<br />

(8) and polyethylene glycol (PEG) (9) hydrogels as it is known that<br />

cells do not attach to these polymer networks. Another approach<br />

involved chemically crosslinking chondroitin sulfate moieties to<br />

PEG hydrogel to stimulate bone marrow-derived mesenchymal<br />

stem cell chondrogenesis (10). A second modification is to provide a<br />

concentrated, local supply of growth factors attached to the polymer<br />

network, which can influence cell behaviors after implantation. This<br />

approach may prove to be particularly important for progenitor<br />

cell therapies as the majority of cartilage repair animal studies<br />

have implanted undifferentiated cells, with the expectation that<br />

the joint environment stimulates chondrogenesis and subsequent<br />

accumulation of repair tissue. One method for incorporating growth<br />

factors into hydrogels is to tether growth factors to the hydrogel<br />

network (11). Or, the polymer network of certain hydrogels have been<br />

found to adsorb growth factors when they are added to the solution<br />

of monomers prior to assembly (11, 12). Given the importance of<br />

inducing progenitor cell chondrogenesis, many of these studies have<br />

focused on the tethering or adsorption of the chondrogenic cytokine<br />

transforming growth factor beta. Modifications of the hydrogel<br />

network may also be used to better control degradation in an effort<br />

to achieve the ideal tissue engineering goal of coordinating neotissue<br />

accumulation with resorption of the scaffold. For example,<br />

matrix metalloproteinase cleavage sites have been engineered into<br />

PEG hydrogels (13) to increase the rate of degradation in the joint,<br />

allowing for the maturation of a continuous repair tissue over time<br />

that may otherwise be impeded by undegraded scaffold. These<br />

examples illustrate the potential to tune hydrogels to meet the needs<br />

of a cartilage tissue engineering scaffold. Hydrogels have also been


148<br />

Extended Abstracts<br />

used to prepare cells for implantation, with the most established<br />

method being the alginate-recovered chondrocyte technique (14).<br />

In this method, chondrocytes are encapsulated in alginate and<br />

cultured in vitro to allow for the accumulation of extracellular matrix.<br />

The alginate network is readily dissociate without harming the<br />

encapsulated cells, resulting in a population of chondrocytes that<br />

are surrounded by a cell-associated neo-cartilage which may be<br />

used for treating cartilage defects within a scaffold or as a scaffoldfree<br />

implant. In our lab, we have investigated the use of selfassembling<br />

peptide hydrogel as a temporary in vitro environment<br />

for chondrogenic priming of bone marrow mesenchymal stem cells.<br />

Similar to the motivation for tethering or adsorbing growth factors,<br />

this work is intended to induce chondrogenesis so that successful<br />

cartilage repair is not entirely dependent on chondrogenic cues<br />

from the joint environment. Our preliminary data has demonstrated<br />

that this technique results in an individual cell suspension of<br />

chondrogenic mesenchymal stem cells that may be seeded into<br />

hydrogels using conventional techniques. From a clinical perspective,<br />

a favorable aspect of many hydrogels is the potential to cast a cellseeded<br />

hydrogel directly into a defect, obviating the need for ex vivo<br />

handling or a fixation strategy. This may be achieved when cells<br />

may be resuspended in a solution of unassembled polymers prior<br />

to gelation and subjected to a crosslinking step without causing<br />

harm to the cells. All of the hydrogels discussed in this abstract<br />

meet these requirements for injectability, with crosslinking achieved<br />

through innocuous processes such as changes in temperature,<br />

pH, or exposure to ions or light. The ease of application of such<br />

hydrogels can be illustrated with fibrin hydrogels, which have<br />

been used as scaffolds or sealants in human clinics for many years<br />

(15). Fibrin hydrogels may be cast using an arthroscopic approach<br />

in which cells suspended in fibrinogen solution are mixed with<br />

thrombin during injection (16). We have used this technique in our<br />

research center to deliver cells to cartilage defects. While the ease<br />

of administration is favorable for clinical translation, most hydrogels<br />

share a property that is common among tissue engineering scaffolds<br />

in that the mechanical stiffness of the hydrogel is much lower than<br />

those of articular cartilage; therefore, hydrogels are typically unable<br />

to support normal joint loading immediately after implantation.<br />

This limitation must be considered for restrictions on joint loading<br />

until functional neo-cartilage is synthesized and assembled by<br />

the implanted cells over time. However, the ability of hydrogels to<br />

support the chondrocyte phenotype, potential for optimization, and<br />

ease of application merit the ongoing investigation of hydrogels for<br />

cartilage repair.<br />

References:<br />

1. Spiller KL, Maher SA, Lowman AM. Hydrogels for the repair of<br />

articular cartilage defects. Tissue Eng Part B Rev. 2011;17(4):281-99<br />

2. Benya PD, Shaffer JD. Dedifferentiated chondrocytes reexpress<br />

the differentiated collagen phenotype when cultured in agarose<br />

gels. Cell. 1982;30(1):215-24.<br />

3. Grodzinsky AJ, Levenston ME, Jin M, Frank EH. <strong>Cartilage</strong> tissue<br />

remodeling in response to mechanical forces. Annu Rev Biomed Eng.<br />

2000;2:691-713.<br />

4. Huang CY, Reuben PM, D’Ippolito G, Schiller PC, Cheung HS.<br />

Chondrogenesis of human bone marrow-derived mesenchymal<br />

stem cells in agarose culture. Anat Rec A Discov Mol Cell Evol Biol.<br />

2004;278(1):428-36.<br />

5. Kisiday JD, Kopesky PW, Evans CH, Grodzinsky AJ, McIlwraith CW,<br />

Frisbie DD. Evaluation of adult equine bone marrow- and adiposederived<br />

progenitor cell chondrogenesis in hydrogel cultures. J Orthop<br />

Res. 2008;26(3):322-31.<br />

6. Mauck RL, Yuan X, Tuan RS. Chondrogenic differentiation and<br />

functional maturation of bovine mesenchymal stem cells in longterm<br />

agarose culture. Osteoarthritis <strong>Cartilage</strong>. 2006;14(2):179-89.<br />

7. Kisiday JD, Frisbie DD, McIlwraith CW, Grodzinsky AJ. Dynamic<br />

compression stimulates proteoglycan synthesis by mesenchymal<br />

stem cells in the absence of chondrogenic cytokines. Tissue Eng Part<br />

A. 2009;15(10):2817-24.<br />

8. Genes NG, Rowley JA, Mooney DJ, Bonassar LJ. Effect of substrate<br />

mechanics on chondrocyte adhesion to modified alginate surfaces.<br />

Arch Biochem Biophys. 2004;422(2):161-7.<br />

9. Salinas CN, Anseth KS. The influence of the RGD peptide motif<br />

and its contextual presentation in PEG gels on human mesenchymal<br />

stem cell viability. J Tissue Eng Regen Med. 2008;2(5):296-304.<br />

10. Varghese S, Hwang NS, Canver AC, Theprungsirikul P, Lin DW,<br />

Elisseeff J. Chondroitin sulfate based niches for chondrogenic<br />

differentiation of mesenchymal stem cells. Matrix Biol. 2008;27(1):12-<br />

21.<br />

11. Miller RE, Kopesky PW, Grodzinsky AJ. Growth factor delivery<br />

through self-assembling peptide scaffolds. Clin Orthop Relat Res.<br />

2011;469(10):2716-24.<br />

12. Shah RN, Shah NA, Del Rosario Lim MM, Hsieh C, Nuber G, Stupp<br />

SI. Supramolecular design of self-assembling nanofibers for cartilage<br />

regeneration. Proc Natl Acad Sci U S A. 2010;107(8):3293-8.<br />

13. Park Y, Lutolf MP, Hubbell JA, Hunziker EB, Wong M. Bovine<br />

primary chondrocyte culture in synthetic matrix metalloproteinasesensitive<br />

poly(ethylene glycol)-based hydrogels as a scaffold for<br />

cartilage repair. Tissue Eng. 2004;10(3-4):515-22.<br />

14. Masuda K, Sah RL, Hejna MJ, Thonar EJ. A novel two-step method<br />

for the formation of tissue-engineered cartilage by mature bovine<br />

chondrocytes: the alginate-recovered-chondrocyte (ARC) method. J<br />

Orthop Res. 2003;21(1):139-48.<br />

15. Ahmed TA, Dare EV, Hincke M. Fibrin: a versatile scaffold for tissue<br />

engineering applications. Tissue Eng Part B Rev. 2008;14(2):199-<br />

215.<br />

16. Wilke MM, Nydam DV, Nixon AJ. Enhanced early chondrogenesis<br />

in articular defects following arthroscopic mesenchymal stem cell<br />

implantation in an equine model. J Orthop Res. 2007;25(7):913-25.<br />

15.2.3<br />

Clinical application of scaffolds for cartilage repair<br />

S. Nehrer<br />

Krems/Austria<br />

Introduction: Cell based therapies in cartilage repair Autologous<br />

Chondrocyte Transplantation with the periosteal flap has changed<br />

the paradigm of the treatment of cartilage defects from repair to<br />

regeneration. This has been demonstrated in randomized trials proving<br />

the concept of regenerating tissue in a cell based therapy approach.<br />

However, the limitations of the periosteal flap concerning size and<br />

thickness and surgical demands with suturing and the variability of the<br />

biological reaction including hypertrophy, as well as the uncontrolled<br />

celltransplatation in a cell suspension has supported the introduction<br />

of biomaterials. The first attempt was to replace the periosteal flap<br />

by membranes which were sutured or glued to the defect combined<br />

with the cellsuspension injected or seeded onto the membrane during<br />

the surgery before implantation. Concerns about the phenotype of<br />

the cultured cells led to the use biomaterial assisted technologies in<br />

sponges or gels to maintain the chondrocyte function of the cells and<br />

allow a more controlled dispersion of the chondrocytes throughout the<br />

defect. The matrix characteristics concerning biochemical composition,<br />

biophysical appearance in gel, foams or sponges, degradation<br />

dynamics and products, toxicity, immunological reactions and general<br />

biocompatibility are important parameters of biomaterial development.<br />

The cell-biomaterial interaction in a biological environment is lastly the<br />

decisive process of successful cartilage regeneration.<br />

Content: Clinical application of scaffolds for cartilage repair Stefan<br />

Nehrer, Florian Halbwirth Center for Regenerative Medicine Danube<br />

University Krems Biomaterials in clinical use Various materials have<br />

been tested in in-vitro laboratory studies and in-vivo animal<br />

experiment to evaluate the best material for human use. Most of the<br />

materials are able to support the chondrocyte phenotype, allow<br />

proliferation and cell migration and facilitate production of cartilage<br />

specific substances. Animal models show in most cases that cell<br />

loaded constructs show superior results than unloaded matrices.<br />

Based on this results clinical studies were employed to prove<br />

feasibility and safety of these technologies. Most of the reported<br />

results are based on case studies only few randomized controlled<br />

trials were performed. There is still discussion about the appropriate<br />

control group in the study design, since microfracture is performed<br />

arthroscopically and basically a different approach, only very few<br />

studies compare matrix techniques with other cell transplantation<br />

methods, however the benchmark is still autologous chondrocyte<br />

transplantation with the periosteum patch. Collagen membrane The<br />

collagen membrane was the first biomaterial used in clinical studies.


Based on positive results in animal studies, confirming the biological<br />

concept of matrix assisted cell technology a bilayer Type 1/3 collagen<br />

membrane was sutured instead of the periosteal flap to the defect<br />

site and the cell suspension is injected underneath. This method still<br />

requires suturing of the membrane and low control of the seeding<br />

process of the cellsuspension. The clinical results of five years followup<br />

are comparable to the periosteum technique. In biopsy studies<br />

hyaline-like tissue have been found. The preseeding of the membrane<br />

was the next development, allowing a more controlled distribution of<br />

the cultured cells and also allows glueing of the graft to the defect. In<br />

this technique the chondrocytes are seeded in a fibrin glue<br />

suspension, however the impact of fibringlue on chondrocytes is still<br />

controversial and experimental and studies on the stability of such<br />

fixation show minor mechanical properties. Hyaluronan In a next step<br />

a preseeded hyaluronan based scaffold has been introduced in the<br />

indication of a chronic cartilage defect. The material consists of a non<br />

woven mesh of hyaluronan fibers which are modified by esterification<br />

by benzylesters to improve the biocompatibility of the material. Since<br />

the hyaluronan is a natural constituent of cartilage and the material<br />

desolves into hyaluronan and is a promising candidate for chondrocyte<br />

transplantation. The in-vitro studies revealed the Hyaff 11 mesh as an<br />

appropriate scaffold for chondrocytes regarding the chondrocytic<br />

phenotype, however the animal studies on the material are sparse<br />

and no conclusive controlled data available. However, the clinical<br />

performance of the material in three- and five-years follow-up studies<br />

document successful performance in over 80 % across the study<br />

population. Stageing the data according to age and severity of the<br />

cartilage defect, patients under the age of 35 and single circumscribed<br />

defects do a lot better with success rates over 90 %. The precultured<br />

hyaluronan matrix shows good attachment of the chondrocytes and<br />

also allows an arthroscopic implantation. The preformed shaping of<br />

the defect and the rough surface of the graft allows fixation without<br />

glueing and pressfit technique and reduces joint morbitiy due to the<br />

arthroscopic approach. PLA/PGA membranes Similar arthroscopic<br />

techniques are performed with precultured matrices consisting of<br />

PLA/PGA meshes, which are secured with technically demanding<br />

intraosseous suture-knot technique or lately with easier pin fixation.<br />

The cell seeded construct shows comparable results to ACT in<br />

midterm follow-up in cohort studies and the stable, mechanically<br />

resistant graft allows also treating circumscribed osteoarthritic<br />

lesions with satisfying outcomes. Recent techniques using minced<br />

cartilage pieces glued to the membrane have been developed,<br />

assuming that outgrowth of chondrocytes from the minced tissue<br />

provide adequate chondrogenic stimulation to facilitate cartilage<br />

regeneration. The minced tissue is harvested with a newly developed<br />

shaver system which allows automized seeding on the artificial<br />

membrane. The 2 years clinical follow up shows better results<br />

compared to microfracture. Collagen Gel The 3D-culture system in<br />

gels allows the chondrocytes to maintain the chondrocytic phenotype,<br />

especially when unpassaged cells are used. The distribution of the<br />

cells resembles more the sparsely dispersion of cells like in natural<br />

cartilage and a high bioactivity of the cell is achieved. The soft<br />

collagen gel needs a well debrided defect with stable walls to allow a<br />

press fit stabilisation which is supported by fibrin glue. The graft<br />

shows a complete fill of the defect and a perfect bonding to the<br />

adjacent cartilage. Clinical results of case controlled studies and<br />

multicenter data show comparable results to ACT and better<br />

performance with regards to effusion and surgery time. Fibrin Fibrin<br />

as a biomaterial was used in liquid form in a glue-cell suspension or<br />

lately also in a spongelike matrix. The fibrin technique allows a<br />

homologous approach to the cell transplantation and the support of<br />

FGF growth factor stimulates cell proliferation and differentiation to<br />

shorten the necessary culture period. First clinical case series show<br />

promising results of the material. The spongelike appearance serves<br />

as a more stable construct than liquid glues. Other combinations of<br />

gels and membranes are available to combine 3D environment with a<br />

biomechanical stable fixation technique. Clinical performance The<br />

clinical results of scaffold techniques are comparable to the<br />

midtermresults of ACT, hence longterm results are not yet available.<br />

Especially circumscribed lesions on the condyle in the younger<br />

patients are doing well in all the studies. However, most studies are<br />

case series and only few are controlled trials including controls. The<br />

advantage of the use of biomaterials is the shorter surgery time, the<br />

easier handling and fixation, the secure and biological appropriate<br />

environment for the chondrocytes during the transplantation process,<br />

the smaller incision including arthroscopic techniques and the more<br />

predictable biological regeneration process avoiding overgrowth like<br />

hypertrophy of the periosteum graft. The safety reports reveal only<br />

minor adverse events most of the time and from the ethic standpoint<br />

no bridges are burned for further treatment options including<br />

revisions, mosaicplasty or redos of cell transplantation. The use of<br />

biomaterials without cultured cells is of course the logistical and<br />

legistical easier process and serves economical interest of companies<br />

and healthcare stake holders. Most of the matrices were developed<br />

Extended Abstracts 149<br />

for the cell based therapies and are now serving as scaffolds for the<br />

bloodclot techniques like microfracture. Some early midtermresults<br />

show increased healing response but <strong>–</strong>like microfracture alone-<br />

inadequate tissue formation and thinning of the surface layer by<br />

intracartilagenous bone formation. However clinical and experimental<br />

studies are on the way and will help to develop the optimal biomaterial<br />

based cartilage repair procedure. Up to now the evidence in<br />

experimental studies is low to suggest successful use of cell-free<br />

constructs and most animal experiments show advantages of the cell<br />

augmented techniques, however the addition of growth hormones,<br />

chondrocyte nutrients or application nanotechnology aspects in<br />

biomimetic materials may allow such technique. First applications of<br />

biomimetic matrices using hydroyapatite and collagen in a gradient<br />

concentration from the osseus to the cartilaginous layer have shown<br />

good results. However, cell based therapies without biomaterials like<br />

chondrospheres or stemcells may be a relevant alternative to<br />

biomaterials and make the difference at last.<br />

References:<br />

1) Bartlett W, Skinner JA, Gooding CR, Canington RW, Flanagan AM,<br />

Briggs TW, Bentley G (2005) Autologous chondrocyte implantation<br />

versus matrix-induced autologous chondrocyte implantation for<br />

osteochondral defects of the knee: a prospective, randomised study.<br />

J Bone Joint Surg Br 87:640-645<br />

2) Behrens P, Bitter T, Kurz B, Russlies M (2006) Matrix-associated<br />

autologous chondrocyte transplantation/implantation (MACT/<br />

MACI)-5-year follow-up. Knee 13: 194-202<br />

3) Bentley G, Biant LC, Carrington RWT, Akmal M, Goldberg A,<br />

Williams AM, Skinner JA, Pringle J (2003) A prospective, randomized<br />

comparison of autologous chondrocyte implantation versus<br />

mosaicplasty for ostechondral defects of the knee. J Bone Joint Surg<br />

Br 85:223-230<br />

4) Brittberg M, Lindahl A, Nilsson A, Ohlsson C, Isaksson O, Peterson<br />

L (1994) Treatment of deep cartilage defects in the knee with<br />

autologous chondrocyte transplantation. N Eng J Med 331:889-895<br />

5) Erggelet C, Sittinger M, Lahm A (2003) The arthroscopic<br />

implantation of autologous chondrocytes for the treatment of fullthickness<br />

cartilage defects of the knee joint. Arthroscopy 19:108-110<br />

6) Gobbi A, Kon E, Berruto M, Francisco R, Filardo G, Marcacci M<br />

(2006) Patellofemoral full-thickness chondral defects treated with<br />

Hyalograft-C: a clinical, arthroscopic, and histologic review. Am J<br />

Sports Med 34:1763-1773<br />

7) Gooding CR, Bartlett W, Bentley G, Skinner JA, Carrington R,<br />

Flanagan A (2006) A prospective, randomised study comparing<br />

two techniques of autologous chondrocyte implantation for<br />

osteochondral defects in the knee: Periosteum covered versus type<br />

I/III collagen covered. Knee 13:203-210<br />

8) Jakobsen RB, Engebretsen L, Slauterbeck JR (2005) An analysis<br />

of the quality of cartilage repair studies. J Bone Joint Surg Am<br />

87:2232-2239 9) Knutsen G, Engebretsen L, Ludvigsen TC, Drogset<br />

JO, Grontvedt T, Solheim E, Strand T, Roberts S, Isaksen V, Johansen<br />

O (2004) Autologous chondrocyte implantation compared with<br />

microfracture in the knee. A randomized trial. J Bone Joint Surg Am<br />

86:455-464<br />

10) Knutsen G, Drogset JO, Engebretsen L, Grontvedt T, Isaksen V,<br />

Ludvigsen TC, Roberts S, Solheim E, Strand T, Johansen O (2007)<br />

A randomized trial of autologous chondrocyte implantation with<br />

microfracture. Findings at five years. J Bone Joint Surg Am 89:2105-<br />

2112<br />

11) Marcacci M. Kon E, Zaffagnini S, Filardo G, Delcogliano M, Neri<br />

MP, Iacono F, Hollander AP (2007) Arthroscopic second generation<br />

autologous chondrocyte implantation. Knee Surg Sports Traumatol<br />

Arthrosc 16:610-619<br />

12) Nehrer S, Dorotka R, Domayer S, Stelzeneder D, Kotz R. Am J<br />

Sports Med. 2009 Nov;37 Suppl 1:81S-87S. Epub 2009 Oct 27.<br />

13) Nehrer S, Chiari C, Domayer S, Barkay H, Yayon A. Clin Orthop<br />

Relat Res. 2008 Aug;466(8):1849-55. Epub 2008 Jun 5.<br />

14) Nehrer S, Domayer S, Dorotka R, Schatz K, Bindreiter U, Kotz . R<br />

(2006) Three-year clinical outcome after chondrocyte transplantation<br />

using a hyaluronan matrix for cartilage repair. Eur J Radiol 57:3-8


150<br />

Extended Abstracts<br />

15) Ossendorf C, Kaps C, Kreuz PC, Burmester GR, Sittinger M,<br />

Erggelet C (2007) Treatment of posttraumatic and focal osteoarthritic<br />

cartilage defects of the knee with autologous polymer-based threedimensional<br />

chondrocyte grafts: 2-year clinical results. Arthritis Res<br />

Ther 9:R41<br />

16) Steinwachs M, Kreuz PC (2007) Autologous chondrocyte<br />

implantation in chondral defects of the knee with a type I/III collagen<br />

membrane: a prospective study with a 3-year follow-up. Arthroscopy<br />

23:381-387<br />

17) Zheng MH, Willers C, Kirilak L, Yates P, Xu J, Wood D, Shimmin A<br />

(2007) Matrix-induced autologous chondrocyte implantation (MACI):<br />

biological and histological assessment. Tissue Eng 13:737-746<br />

15.3.1<br />

Importance of subchondral bone in cartilage repair<br />

H. Madry<br />

Homburg/Germany<br />

Introduction: The subchondral bone lies immediately below the<br />

calcified zone of the articular cartilage, separated from it by the<br />

cement line. It consists of two mineralized layers, the subchondral<br />

bone plate and the subarticular spongiosa. The subchondral bone<br />

plate is composed of dense cancellous bone. Adjacent to the<br />

articular cartilage, this bone consists of relatively thick plates which<br />

join together to enclose relatively narrow intervening spaces. In the<br />

deeper regions of the subchondral bone, these spaces are gradually<br />

enlarged and become elongated in a direction parallel to the<br />

diaphysis, forming the subarticular spongiosa. Its trabeculae arise<br />

from the subchondral bone plate. The subchondral bone is subjected<br />

to a range of static and dynamic forces, such as shear, tension,<br />

compression. These mechanical loads are mainly absorbed by the<br />

extracellular matrix of the articular cartilage and are transmitted<br />

to the subchondral bone. The subchondral bone and the articular<br />

cartilage form the osteochondral unit.<br />

Content: Marrow stimulation techniques for articular cartilage<br />

repair, resulting in a surgically-created communication between<br />

a symptomatic cartilage defect and the bone marrow, affect the<br />

subchondral bone. Recent data from experimental studies in small<br />

and large animal models and clinical investigations suggest significant<br />

alterations of the subchondral bone microarchitecture following<br />

marrow stimulation. Initially, early bone resorption is followed by<br />

bone remodeling as in fracture healing. Caroline Hoehmann and<br />

Michael Buschman have shown in the rabbit model that microfracture<br />

leads to bone compaction as indicated by significantly higher bone<br />

density and trabecular thickness, compared to drilling. The depth<br />

of drilling also plays a role - deep drilling leading to a larger volume<br />

of bone remodelling than shallow drilling. We have recently shown<br />

in a sheep model that drilling induces significant alterations of<br />

nearly all microarchitectural parameters of the subchondral bone<br />

plate and the subarticular spongiosa after 6 months. These include,<br />

but are not limited, the bone volume (BV/TV), bone surface /<br />

volume ratio, trabecular thickness, separation, and pattern factor<br />

and bone mineral density. Particularly of interest, these observed<br />

microarchitectural changes were similar to the structural patterns<br />

observed in osteoporosis and osteoarthritis such as a decrease in<br />

BV/TV, bone mineral density, and trabecular thickness vis-à-vis an<br />

increase in trabecular separation. In addition, the appearance of<br />

subchondral bone cysts, the formation of intralesional osteophytes,<br />

and the upward migration of the subchondral bone plate are key<br />

features that have been described both in small and large animal<br />

models and in patients. Subchondral bone cyst formation may<br />

result from penetration of the subchondral bone combined with<br />

immediate full weight bearing. A recent long-term investigation of<br />

ovine subchondral bone microarchitecture found subchondral bone<br />

cysts in 62% of cartilage defects that were treated by drilling and<br />

intralesional osteophytes in 26% of cases.<br />

An intralesional osteophyte is defined as new bone formation<br />

apical to the cement line, projected into the articular cartilage.<br />

Upward migration of the entire subchondral bone plate into the<br />

cartilaginous repair tissue has been also observed in several animal<br />

studies. Such new bone formation may be a factor playing a role<br />

in the degeneration of cartilaginous repair tissue over time. In<br />

addition, also the subchondral plate surrounding treated defects is<br />

affected. The subchondral plate has been shown to thicken following<br />

subchondral drilling, suggesting that marrow stimulation may also<br />

induce long-term changes in the subchondral bone adjacent to the<br />

treated defects.<br />

Clinical results after marrow stimulation of small symptomatic<br />

articular cartilage defects are promising, especially within the first 2<br />

years after surgery. In agreement with the data generated in animal<br />

models, however, clinical evidence suggests that marrow stimulation<br />

may induce similar alterations in the subchondral bone plate such<br />

as upward migration, intralesional osteophytes or subchondral bone<br />

cysts, which may play a role in the degeneration of the repair tissue<br />

over time. Also, autologous chondrocyte implantation (ACI) has been<br />

reported to have a three-fold higher failure rate when applied to<br />

articular cartilage defects previously treated with marrow stimulation<br />

techniques compared with previously untreated defects.<br />

In general, the lower resolution of imaging techniques applicable<br />

in clinical investigations compared with experimental tools such<br />

as high-resolution micro CT makes the differentiation between a<br />

laminar upward migration of the entire subchondral bone plate<br />

and a localized formation of intralesional osteophytes difficult.<br />

Although MRI is a valuable tool for to monitor cartilage repair,<br />

depiction of subchondral bone microarchitecture is still not possible<br />

in patients. <strong>Cartilage</strong> defects treated by microfracture developed<br />

more subchondral bone reaction after 3 years compared with the<br />

baseline using MRI. Such changes of the subchondral bone plate<br />

may predispose for the degeneration of the repair tissue in a pattern<br />

similar to osteoarthritis, when the thickened and stiff subchondral<br />

bone plate increases friction and shear forces. A bone marrow edema<br />

is a frequent but non-specific signal pattern that can be related<br />

to ischemia (i.e. osteonecrosis, bone marrow edema syndrome),<br />

mechanical (bone bruise, microfracture) or reactive (osteoarthritis,<br />

postoperative bone marrow edema) causes. Often, the presence<br />

of such a bone marrow edema is a first sign of a corresponding<br />

chondral lesion.<br />

Taken together, alterations of the subchondral bone are associated<br />

with the clinical use of marrow stimulation techniques for articular<br />

cartilage repair. These changes have been confirmed in various<br />

small and large animal models of different types of cartilage defects.<br />

The emerging experimental and clinical data suggest that a possible<br />

deterioration of the subchondral bone plate and subarticular<br />

spongiosa underlying the cartilage defect might be an additional,<br />

previously underestimated factor that significantly influences the<br />

long-term outcome of articular cartilage repair following marrow<br />

stimulation and possibly other reconstructive surgical techniques.<br />

More data are therefore needed to better understand the clinical<br />

relevance of subchondral cysts as they are often unrelated to the<br />

symptoms of the patients. Clinical evidence confirms that marrow<br />

stimulation induces upward migration of the subchondral bone plate<br />

and formation of bone marrow edemas, intralesional osteophytes or<br />

bone cysts in approximately 25% of patients, possibly playing a role<br />

in the degeneration and failure of the repair tissue. Future studies<br />

will also have to focus on the containment of these subchondral bone<br />

alterations, e.g. by optimizing surgical techniques or postoperative<br />

regimes. Also, highly resolving imaging techniques need to be<br />

developed and clinically tested to gain more insights into the clinical<br />

mechanisms of subchondral bone changes.<br />

A deeper comprehension of the subchondral bone alterations<br />

associated with cartilage repair will translate into further improved<br />

techniques to preserve and restore the entire osteochondral unit.<br />

References:<br />

H. Madry, C.N. van Dijk and M. Mueller-Gerbl, The basic science of<br />

the subchondral bone, Knee Surg. Sports Traumatol. Arthrosc. 18<br />

(2010), 419-433.<br />

P.C. Kreuz, M.R. Steinwachs, C. Erggelet, S.J. Krause, G. Konrad,<br />

M. Uhl and N. Sudkamp, Results after microfracture of fullthickness<br />

chondral defects in different compartments in the knee,<br />

Osteoarthritis <strong>Cartilage</strong> 14 (2006), 1119-1125.<br />

K. Mithoefer, R.J. Williams, 3rd, R.F. Warren, H.G. Potter, C.R. Spock,<br />

E.C. Jones, T.L. Wickiewicz and R.G. Marx, The microfracture technique<br />

for the treatment of articular cartilage lesions in the knee. A prospective<br />

cohort study, J. Bone Joint Surg. Am. 87 (2005), 1911-1920.<br />

H. Chen, J. Sun, C.D. Hoemann, V. Lascau-Coman, W. Ouyang, M.D.<br />

McKee, M.S. Shive and M.D. Buschmann, Drilling and microfracture<br />

lead to different bone structure and necrosis during bone-marrow<br />

stimulation for cartilage repair, J. Orthop. Res. 27 (2009), 1432-<br />

1438.


H. Chen, A. Chevrier, C.D. Hoemann, J. Sun, W. Ouyang and<br />

M.D. Buschmann, Characterization of subchondral bone repair<br />

for marrow-stimulated chondral defects and its relationship to<br />

articular cartilage resurfacing, Am. J. Sports Med. 39 (2011), 1731-<br />

1740.<br />

P. Orth, L. Goebel, U. Wolfram, M.F. Ong, S. Gräber, D. Kohn,<br />

M. Cucchiarini, A. Ignatius, D. Pape and H. Madry, Effect of<br />

subchondral drilling on the microarchitecture of subchondral<br />

bone: analysis in a large animal model at 6 months, Am. J. Sports<br />

Med. (2012) PMID:22223716. in press.<br />

D. Lajeunesse, G. Hilal, J.P. Pelletier and J. Martel-Pelletier,<br />

Subchondral bone morphological and biochemical alterations in<br />

osteoarthritis, Osteoarthritis <strong>Cartilage</strong> 7 (1999), 321-322.<br />

T. Minas, A.H. Gomoll, R. Rosenberger, R.O. Royce and T. Bryant,<br />

Increased failure rate of autologous chondrocyte implantation<br />

after previous treatment with marrow stimulation techniques, Am.<br />

J. Sports Med. 37 (2009), 902-908.<br />

D.D. Frisbie, S. Morisset, C.P. Ho, W.G. Rodkey, J.R. Steadman and<br />

C.W. McIlwraith, Effects of calcified cartilage on healing of chondral<br />

defects treated with microfracture in horses, Am. J. Sports Med. 34<br />

(2006), 1824-1831.<br />

D.B. Saris, J. Vanlauwe, J. Victor, K.F. Almqvist, R. Verdonk, J.<br />

Bellemans and F.P. Luyten, Treatment of symptomatic cartilage<br />

defects of the knee: characterized chondrocyte implantation<br />

results in better clinical outcome at 36 months in a randomized<br />

trial compared to microfracture, Am. J. Sports Med. 37 Suppl 1<br />

(2009), 10S-19S.<br />

Acknowledgments:<br />

Supported in part by AGA and GOTS.<br />

15.3.2<br />

Digital imaging of subchondral bone density<br />

F.P.J.G. Lafeber, M. Kinds, F. Intema, S.C. Mastbergen<br />

Utrecht/Netherlands<br />

Introduction: Subchondral bone changes in osteoarthritis: Osteoarthritis<br />

(OA) is a degenerative joint disease characterized by cartilage destruction<br />

and changes in subchondral bone. Joints most affected are hip, knees,<br />

and spine, but every joint can be affected including ankle joints.<br />

Subchondral bone changes are a distinctive feature in OA development,<br />

and they include sclerosis, cyst formation, bone attrition, bone marrow<br />

lesions, and osteophytes. Subchondral bone changes alter the joint‘s<br />

mechanical as well as biochemical environment and have a direct and<br />

indirect influence on the overlaying articular cartilage. Consequently,<br />

subchondral bone has been identified as an attractive target for<br />

treatment in OA, and should be monitored in detail. (1)<br />

Content: Subchondral bone remodelling is related to clinical<br />

improvement after clinical effective treatment of OA.(2) Joint distraction<br />

as a treatment of OA has been shown to provide pain relief and<br />

functional improvement through mechanisms that are not well<br />

understood. It was demonstrated that subchondral bone remodelling<br />

is associated with clinical improvement in OA patients treated with<br />

joint distraction. In twenty-six patients with advanced post-traumatic<br />

ankle OA, treated with joint distraction for 3 months using an Ilizarov<br />

frame, bone density (BD) change were analysed by computed<br />

tomography (CT) scans. Longitudinal, manually segmented CT<br />

datasets for a given patient were brought into a common spatial<br />

alignment. Changes in BD (Hounsfield Units; relative to baseline) were<br />

calculated at the weight-bearing region, extending subchondral to a<br />

depth of 8 mm. Clinical outcome was assessed using the ankle OA<br />

scale. Baseline scans demonstrated subchondral sclerosis with local<br />

cysts. At 1 and 2 years of follow-up, an overall decrease in BD (-23%<br />

and -21%, respectively) was observed. Interestingly, BD in originally<br />

low-density (cystic) areas increased. Joint distraction resulted in a<br />

decrease in pain (from 60 to35, scale of 100) and functional deficit<br />

(from 67 to 36). Improvements in clinical outcomes were best<br />

correlated with disappearance of low-density (cystic) areas (R=0.69).<br />

From this study it is concluded that treatment of advanced posttraumatic<br />

ankle OA with joint distraction results in BD normalization<br />

that is associated with clinical improvement. While overall BD<br />

decreased, BD in cystic lesions actually increased (normalization of<br />

Extended Abstracts 151<br />

BD) and a correlation was found between clinical improvement and<br />

the resolution of subchondral bone cysts. Feasibility of bone density<br />

evaluation using plain digital radiography.(3) Clearly, (a change in)<br />

subchondral BD is an important feature of OA. For measurement of BD<br />

several methods have been reported on, including dual energy digital<br />

radiography (DEDR), (quantitative) computed tomography (QCT; as<br />

described above), radiographic absorptiometry, but most importantly<br />

dual energy X-ray absorptiometry (DEXA) which is the most validated<br />

and commonly used method. However, for evaluation of structural<br />

changes due to OA, in general practice, plain radiographs are<br />

commonly acquired. If the quantitative evaluation of clinically relevant<br />

BD changes on these radiographs is proved feasible, it obviates the<br />

need to acquire additional methods. In general practice subchondral<br />

BD changes are commonly subjectively assessed. Although the use of<br />

film-screen radiography has been described in the evaluation of BD,<br />

this technique has been almost completely replaced by digital<br />

radiography. The accuracy of digital radiography in BD measurement<br />

has received no attention, however. One important feature of digital<br />

radiography is that image post-processing (PP) is incorporated in the<br />

scan protocol. This PP generally includes adjustment of contrast<br />

curves and application of non-linear image filters to optimize image<br />

quality parameters such as contrast and noise. PP aims at improving<br />

diagnostic readability, rather than allowing quantitative analyses to<br />

assess BD changes for longitudinal evaluation. Furthermore, the<br />

acquisition settings including tube voltage (in kilovolt: kV), exposure<br />

(in milli ampere seconds: mAs), and filtration can vary between<br />

technologists, exam rooms, and institutes, which may influence crosssectional<br />

or longitudinal BD evaluation. To enable quantification of<br />

BD, independently of PP and acquisition settings, the inclusion of an<br />

aluminium (step) wedge in the radiographic field-of-view has been<br />

suggested.(4) In this way the grey values of the bone can be expressed<br />

in mm aluminium equivalents (mm Al). The feasibility of BD evaluation<br />

using digital radiography was evaluated. A bone density standard<br />

(BDS) of hydroxyapatite (HA) mimicked a BD range of 1.0 - 5.75 g/cm2.<br />

Digital radiographs were acquired with variation in acquisition<br />

settings, and with clinical and minimal PP. An aluminium step wedge<br />

served as an internal reference to express the grey values of the BDS<br />

in mm aluminium equivalents (mm Al). The relation between actual BD<br />

and BD normalized to the reference wedge was evaluated with linear<br />

regression analyses for radiographs with variations in PP and<br />

acquisition settings. Precision of BD measurement of the BDS was<br />

evaluated for application in clinical practice. The correlation between<br />

actual BD and BD normalized to the reference was improved by<br />

changing PP from clinical (R2 = 0.96) to minimal (R2 = 0.98). Higher<br />

tube voltage [kilovolt (kV)] improved the correlation further. Even for<br />

clinical PP, average standard deviation (SD) was 0.97 mmAl, much<br />

smaller than the change of 2.51 mm Al clinically observed in early OA,<br />

which implies the feasibility of BD measurements on digital radiographs<br />

It was demonstrate that BD measurement on plain digital radiographs<br />

is feasible. However, since variations in digital radiography settings at<br />

acquisition influence the outcome, BD measurements should be<br />

interpreted with caution. In clinical practice variations in acquisition<br />

settings within institutes and specifically between institutes (in<br />

multicenter trials) have been found to vary in a relevant range.<br />

Furthermore, differences in performance between different brands<br />

and types of digital radiography acquisition systems will exist. Digital<br />

radiographs without a reference need to be evaluated with caution<br />

since in optimization of images, PP algorithm plays an important role.<br />

For example, during longitudinal evaluation of a knee joint, the clinical<br />

PP algorithm might yield radiographs with a similar appearance while<br />

BD actually changed based on disease (e.g. OA). On the other hand,<br />

radiographs might appear different as a result of variations in PP<br />

settings (or different radiography systems) within and between centres<br />

rather than as a result of BD changes. Although in clinical practice<br />

variations in digital radiography and PP settings will occur that<br />

influence BD measurement, the addition of a reference enables an<br />

adequate assessment of the grey values. A limitation of the use of a<br />

BDS might be that it is in general a simplified representation of tissue<br />

composition of a human knee without anatomical resemblance.<br />

However, the mean subchondral BD values determined with DEXA at<br />

the medial tibia were 2.21 g/cm2 for a healthy human knee joint, with<br />

a linear correlation with BD values on digital radiographs (R2 = 0.91),<br />

and1.70 g/cm2 for a cadaver knee joint, which shows that the BDS<br />

represented a clinically relevant range. The BDS experiments indicated<br />

that the precision of BD measurement could be increased by using<br />

minimal PP rather than clinical PP and by applying relatively high kV.<br />

The relation between actual BD and BD normalized to the reference is<br />

weak when low tube voltage (44 kV) is used especially at larger BD<br />

values, which might be due to relatively more absorption of the beam<br />

by the knee joint. Although the application of higher kV improves<br />

linearity of the relation between actual and normalized BD, patient<br />

exposure needs to be taken into account. Improved accuracy without<br />

additional patient exposure can be reached by using higher kV in<br />

combination with lower mAs. Applying minimal PP to improve accuracy


152<br />

Extended Abstracts<br />

is not easily applicable in regular clinical practice since clinical PP is<br />

required to provide optimal diagnostic image quality, and in general<br />

cannot easily be bypassed in clinical practice. Overall, the BDS<br />

experiments and the comparison to clinical data indicate that BD<br />

measurement using digital radiography is feasible in a clinically<br />

relevant range. Variations in acquisition and PP settings within and<br />

between clinics can have profound effect on BD evaluation and should<br />

therefore be considered with caution. As compared to the default<br />

clinical protocol, the accuracy of BD measurements can be improved<br />

by applying only minimal image PP and a relatively high kV. Provided<br />

properly performed, plain digital radiography may yield, in addition to<br />

OA characteristics, reliable data on BD which reduces the need for<br />

additional imaging techniques.<br />

References:<br />

1) Bijlsma JWJ, Berenbaum F, and Lafeber FPJG. Osteoarthritis: an<br />

update with relevance for clinical practice. Lancet. 2011; 377:2115-26.<br />

2) Intema F, Thomas TP, Anderson DD, Elkins JM, Brown TD, Amendola<br />

A, Lafeber FPJG, and Saltzman CL. Subchondral bone remodeling is<br />

related to clinical improvement after joint distraction in the treatment<br />

of ankle osteoarthritis. Osteoarthritis and <strong>Cartilage</strong> 2011; 19: 668-675.<br />

3) Kinds MB, Bartels LW, Marijnissen ACA, Vincken K, Viergever MA,<br />

Lafeber FPJG, andDeJong HWAM. Feasibility of bone density evaluation<br />

using plain digital radiography. Osteoarthritis and <strong>Cartilage</strong> 2011; 19:<br />

1343-1348.<br />

4) Marijnissen AC, Vincken KL, Vos PA, Saris DB, Viergever MA, Bijlsma<br />

JW, Bartels LW, Lafeber FP. Knee Images Digital Analysis (KIDA): a<br />

novel method to quantify individual radiographic features of knee<br />

osteoarthritis in detail. Osteoarthritis <strong>Cartilage</strong>. 2008;16:234-43.<br />

Acknowledgments:<br />

-Orthopaedics & Rehabilitation, The University of Iowa, Iowa City, IA,<br />

USA<br />

-Image Sciences Institute, University Medical Centre (UMC), Utrecht,<br />

The Netherlands<br />

15.3.3<br />

Fresh Osteochondral Allografting in the Knee<br />

W.D. Bugbee<br />

La Jolla/United States of America<br />

Introduction: The concept of treating articular cartilage diseases with<br />

bone and cartilage substitution in the knee has now a history of more<br />

than a century, since the first joint transplantation described by Lexer<br />

in 1908.1,2 Animal and clinical studies concerning transplantation<br />

and immunology were carried out in the sixties, demonstrating that<br />

transplanted fresh cadaver cartilage is viable.3-5 In the seventies Gross<br />

and colleagues began reporting on their experience with osteochondral<br />

allograft for post traumatic and periarticular tumor reconstruction.6,7<br />

In the eighties Meyers and Convery firstly applied this technique to<br />

specific chondral and osteochondral diseases such as chondromalacia,<br />

osteoarthritis and osteonecrosis,8 developing the shell-shaped graft.<br />

Later in the nineties that Garrett first reported on the use of allograft<br />

plugs for the treatment of osteochondritis dissecans of the knee.9 In<br />

the last twenty years a large number of basic scientific and clinical<br />

studies have been performed by a number of investigators. These<br />

studies and the increasing availability of fresh allografts, have led to<br />

an increasing popularity of fresh allografts and the inclusion of this<br />

procedure as part of the “cartilage repair paradigm” for the treatment<br />

of chondral or osteochondral lesions in the knee.10,11,12<br />

Content: ALLOGRAFT RECOVERY, PROCESSING AND STORAGE<br />

Historically, in North America, fresh osteochondral allograft procedures<br />

were performed at university based centers that had associated<br />

tissue banks which independently established recovery, processing<br />

and release protocols. Fresh osteochondral allografts (OCA) were<br />

typically stored in lactated Ringer’s solution and transplanted fresh<br />

within one week after donor death. Under this model approximately<br />

100 allografts per year were implanted in North America in the<br />

eighties and nineties. Beginning around 1998, commercially supplied<br />

allografts became available in the United States through a number<br />

of tissue banks that established new protocols under the oversight<br />

of the Food and Drug Administration (FDA). Commercial distribution<br />

of grafts required a prolonged storage interval (10-45 days) to allow<br />

for completion of recovery and testing protocols. This resulted in an<br />

increase in the number of allografting procedures performed in the US<br />

to approximately 2000 per year.<br />

Allograft tissue recovery is performed within 12-24 hours of donor<br />

death.13 Suitable donors are generally between 15 and 35 years old with<br />

macroscopically healthy articular cartilage. Since the transplantation<br />

procedure is based on cartilage substitution, a process that maintains<br />

allograft cartilage tissue health during storage is mandatory. Many<br />

studies have been carried out to identify the ideal storage media and<br />

to evaluate the effects of hypothermic storage on chondrocytes and<br />

extracellular matrix.14-19<br />

Osteochondral allografts can be stored frozen, cryopreserved or fresh.<br />

Each of these options affects chondrocyte viability, immunogenicity,<br />

and length of time to transplantation. Frozen grafts showed a<br />

chondrocyte survivorship of less than 5%, because of the freezing<br />

process at -80°C.20 As chondrocytes are responsible for maintenance<br />

of the extracellular matrix, studies have shown that the matrix in these<br />

frozen allografts tends to deteriorate over time. 21,22 Along with<br />

the decreased chondrocyte viability, fresh-frozen allografts showed<br />

decreased immunogenicity.23<br />

With cryopreservation it is possible to maintain chondrocyte viability<br />

during this freezing process by adding glycerol and dimethyl sulfoxide<br />

(DMSO) to the tissue. Theoretically, the addition of these chemicals<br />

prevents ice formation within cells. Multiple studies have reported<br />

variable results, with chondrocyte survival ranging from 20% to<br />

70%.24-27 Unfortunately, viable cells were found only at the surface<br />

of the articular cartilage layer.28 Fresh allografts proved to have the<br />

highest rates of chondrocyte viability of the three different methods<br />

of storage.19,25,29,30 Fresh grafts are usually placed in tissue culture<br />

medium at 4°C (or potentially 37°C). Chondrocyte viability is significantly<br />

affected by length of storage, with little effect from storage times less<br />

than one week.31,32 The time of storage before implantation is a key<br />

point. Studies have shown a time-dependent decreased chondrocyte<br />

viability and degradation of biomechanical properties of fresh grafts<br />

stored for greater than 14 days.33-35 Currently the trend of the<br />

tissue banks is to hold transplants for a minimum of 14 days, to allow<br />

completion of microbiologic and serologic testing prior to release.36<br />

More recently a new off-the-shelf alternative to classic OCA has<br />

been developed and released in the US market: The Chondrofix®<br />

Osteochondral Allograft (Zimmer, Inc. 1800 West Center Street<br />

Warsaw, IN, USA). This product is an osteochondral allograft<br />

consisting of decellularized hyaline cartilage and cancellous bone,<br />

recovered by an accredited tissue bank, processed to be sterile and<br />

viral-inactivated, hydrated, precut, and ready for implantation. The<br />

relative advantages include an off the shelf availability, sterility and<br />

ease of use while potential limits are availability in sizes only up<br />

to 15mm and the absence of viable cells within the graft. Currently<br />

no published peer reviewed data is available, however preliminary<br />

experience suggests a place for this product in the pool of newer<br />

alternatives for chondral and osteochondral repair or replacement.<br />

INDICATION FOR ALLOGRAFTS<br />

The structural features and multi shaping possibilities makes<br />

osteochondral allografts suitable for the treatment of a wide spectrum<br />

of diseases, which can be grouped into two main paradigms (Table<br />

1). The first treatment paradigm includes complex reconstruction<br />

procedures, such as post traumatic deformity and degenerative<br />

lesions associated with intra-articular fracture malunion, most<br />

commonly of the tibial plateau,40,41 and unicompartmental<br />

arthrosis or multifocal chondrosis including patellofemoral<br />

degeneration.42-44 This group also includes massive type 3 or 4<br />

osteochondritis dissecans (OCD),9,10 osteonecrosis,45 as well as<br />

other diseases primarily affecting the subchondral bone. The second<br />

treatment paradigm is formed by conditions primarily affecting<br />

articular cartilage. These include large chondral defects treated<br />

primarily with an allografts or defect that have been previously<br />

treated by another cartilage repair technique such as microfracture,<br />

OATS or Autologous Chondrocyte Implantation that have failed or<br />

that have developed compromise of the subchondral bone.<br />

Although the knee is the most common joint for osteochondral<br />

allografting, experience in other joints has been reported. Several<br />

case series have been reported in the ankle joint. Good results<br />

have been shown with the use of allografts in the treatment of large<br />

osteochondral lesions of the talus.46-50 Mixed results have been<br />

demonstrated for Bipolar shell grafting for ankle osteoarthritis.


51,52 Experience with allografts has also been described in the<br />

hip or in the shoulder, as treatment of femoral or humeral head<br />

osteonecrosis or for osteochondral lesions associated with shoulder<br />

instability. 53-55<br />

RESULTS<br />

Clinical results of fresh osteochondral allografts in the knee joint<br />

have shown encouraging long-term results, with overall success<br />

rates from 50% to 95%.10,30,32,41,45,58. The most commonly<br />

treated lesion location is the femoral condyle. Table 2 outlines the<br />

major studies reporting outcomes of osteochondral allografting of<br />

isolated lesions of the femoral condyle.<br />

In post traumatic reconstruction of the tibial plateau, Shasha et al. in<br />

2003 41 reported the long term outcome of sixty-five patients treated<br />

with fresh tibial osteochondral allografts. At a mean of twelve years,<br />

forty-four patients had an intact graft, while twenty-one had had<br />

conversion to a total knee arthroplasty at an average of ten years.<br />

The reported survival rate was 95% at five years, 80% at ten years<br />

and 65% at fifteen years.<br />

The “San Diego experience” with osteochondral allografting in<br />

the knee extends almost thrity years. In 1983, an institutional<br />

review board (IRB) approved osteochondral allografting program<br />

was established for the evaluation and treatment of complex or<br />

advanced articular cartilage disease. This comprehensive program<br />

was comprised of scientific and clinical components, including<br />

retrieval studies. Over the last 30 years, we have collected outcomes<br />

data on all patients unergoing fresh osteochondral allografting with<br />

the purpose of better defining the indications and understanding<br />

clinical outcome. Since 1983, 515 patients have undergone 576<br />

knee allografting procedures. Of those, 328 patients (354 knees)<br />

currently have minimum 2-year follow-up and 187 patients (222<br />

knees) do not have 2-year follow-up data (59 patients are less than<br />

2 years from surgery). The following results includes only the 354<br />

knees with minimum 2-year follow-up. The mean follow-up was 86<br />

months (range 24<strong>–</strong>309 months). Patient characteristics and details<br />

regarding the allograft are presented in table 3 . Objective clinical<br />

outcome showed improvements in both pain and function on all<br />

measures utilized (table 4). Subjectively, 96% of patients reported<br />

satisfaction (73% extremely satisfied), 93% reported less pain, and<br />

94% reported better function as a result of the allograft procedure.<br />

Almost all patients (92%) stated they would have the surgery again<br />

under similar circumstances.<br />

Seventy-two knees (20%) underwent a reoperation that included<br />

removal or revision of the allograft and were defined as clinical<br />

failures. The 72 failures included 41 total knee arthroplasties, 23<br />

revision allografts, 4 partial knee arthroplasties, 2 patellectomies,<br />

and 2 knee fusions. The mean time to failure was 40 months (range,<br />

3<strong>–</strong>165 months). Survivorship was 82% at 5 years, 72% at 10 years,<br />

and 70% at 25 years. The best outcome, by diagnosis, was seen in<br />

the patients with osteochondral dissecans (12% failures). Of the<br />

osteochondral dissecans non-failures, the mean modified D’Aubigne<br />

and Postel score 64 improved from 12.9 to 16.7, the mean IKDC 65<br />

pain score improved from 5.2 to 2.2, the mean IKDC function score<br />

improved from 3.9 to 8.1, and the mean KS-F score 66 improved from<br />

76 to 92 (all p


154<br />

Extended Abstracts<br />

Annotation: Table 4 Results of objective outcome measures<br />

*paired t-test<br />

References:<br />

1. Lexer E. Substitution of whole or half joints from freshly amputated<br />

extremities by free plastic operation. Surg Gynecol Obstet 1908;6601-<br />

6607<br />

2. Lexer E. Joint transplantations and arthroplasty. Surg Gynec Obst<br />

1925;40:782-809<br />

3. Campbell CJ, Ishida H, Takahashi H, Kelly F. The transplantation of<br />

articular cartilage: an experimental study in dogs. J Bone Joint Surg<br />

[Am] 1963;45-A:1579-1592<br />

4. Craigmyle MBL. An autoradiographic and histochemical study of<br />

long-term cartilage grafts in the rabbit. J Anat 1958;92:467-72.<br />

5. Depalma AF, Tsaltas TT, Mauler GG. Viability of osteochondral<br />

grafts as determined by uptake of S35. J Bone Joint Surg [Am] 1963;45-<br />

A:1565-1578<br />

6. Locht RC, Gross AE, Langer F. Late osteochondral allograft resurfacing<br />

for tibial plateau fractures. J Bone Joint Surg Am. 1984;66(3):328-335<br />

7. Bell RS, Davis A, Allan DG, Langer F, Czitrom AA, Gross AE. Fresh<br />

osteochondral allografts for advanced giant cell tumors at the knee. J<br />

Arthroplasty. 1994;9(6):603-609<br />

8. Meyers MH, Akeson W, Convery FR. Resurfacing of the knee with<br />

fresh osteochondral allograft. J Bone Joint Surg Am. 1989;71(5):704-<br />

713<br />

9. Garrett JC. Fresh osteochondral allografts for treatment of articular<br />

defects in osteochondritis dissecans of the lateral femoral condyle in<br />

adults. Clin Orthop Relat Res. 1994;(303):33-37<br />

10. Emmerson BC, Görtz S, Jamali AA, Chung C, Amiel D, Bugbee WD.<br />

Fresh osteochondral allografting in the treatment of osteochondritis<br />

dissecans of the femoral condyle. Am J Sports Med. 2007;35(6):907-<br />

914.<br />

11. Görtz S, Bugbee WD. Allografts in articular cartilage repair. J Bone<br />

Joint Surg Am. 2006;88(6):1374-1384<br />

12. Sgaglione NA, Chen E, Bert JM, Amendola A, Bugbee WD. Current<br />

strategies for nonsurgical, arthroscopic, and minimally invasive<br />

surgical treatment of knee cartilage pathology. Instr Course Lect.<br />

2010;59:157-80<br />

13. Burchardt H. The biology of bone graft repair. Clin Orthop<br />

1983;174:28-42<br />

14. Williams SK, Amiel D, Ball ST, et al. Prolonged Storage Effects on<br />

the Articular <strong>Cartilage</strong> of Fresh Human Osteochondral Allografts. J Bone<br />

Joint Surg Am 2003;85-A(11):2111<strong>–</strong>2120<br />

15. Allen RT, Robertson CM, Pennock AT, et al. Analysis of Stored<br />

Osteochondral Allografts at the Time of Surgical Implantation. Am J<br />

Sports Med 2005;33(10):1479<strong>–</strong>1494<br />

16. Pennock AT, Robertson CM, Wagner F, Harwood FL, Bugbee WD,<br />

Amiel D. Does Subchondral Bone Affect the Fate of Osteochondral<br />

Allografts During Storage? Am J Sports Med 2006;34(4):586<strong>–</strong>591<br />

17. Robertson CM, Allen TR, Bugbee WD, Harwood FL, Healey RM,<br />

Amiel D. Upregulation of Apoptotic and Matrix-related Gene Expression<br />

During Fresh Osteochondral Allograft Storage. Clin Orthop Relat Res<br />

2006;442:260<strong>–</strong>266<br />

18. Pennock AT, Wagner F, Harwood FL, Bugbee WD, Amiel D. Prolonged<br />

Storage of Osteochondral Allografts: Does the Addition of Fetal Bovine<br />

Serum Improve Chondrocyte Viability? J Knee Surg 2006;19(4):265<strong>–</strong><br />

272<br />

19. Pallante AL, Bae WC, Chen AC, Görtz S, Bugbee WD, Sah RL.<br />

Chondrocyte Viability is Higher after Prolonged Storage at 37°C than<br />

at 4°C for Osteochondral Grafts. Am J Sports Med. 2009 Nov;37 Suppl<br />

1:24S-32S<br />

20. Enneking WF, Mindell ER. Observations on massive retrieved<br />

human allografts. J Bone Joint Surg Am 1991;73:1123<strong>–</strong>1142<br />

21. Enneking WF, Campanacci DA. Retrieved human allografts: a<br />

clinicopathological study. J Bone Joint Surg Am 2001;83:971<strong>–</strong>986<br />

22. Acosta CA, Izal I, Ripalda P, et al. Cell viability and protein composition<br />

in cryopreserved cartilage. Clin Orthop Relat Res 2007;460:234<strong>–</strong>239<br />

23. Gortz S, Bugbee WD. Allografts in articular cartilage repair. Instr<br />

Course Lect 2007;56:469<strong>–</strong>480<br />

24. Rodrigo JJ, Thompson E, Travis C. Deep-freezing versus 4 degrees<br />

preservation of avascular osteocartilaginous shell allografts in rats.<br />

Clin Orthop Relat Res 1987;218:268<strong>–</strong>275<br />

25. Schachar N, McAllister D, Stevenson M, et al. Metabolic and<br />

biochemical status of articular cartilage following cryopreservation<br />

and transplantation: A rabbit model. J Orthop Res 1992;10:603<strong>–</strong>609<br />

26. Jamali AA, Hatcher SL, You Z. Donor cell survival in a fresh<br />

osteochondral allograft at twenty-nine years. A case report. J Bone<br />

Joint Surg Am 2007;89(1):166<strong>–</strong>169<br />

27. Bobic V. Arthroscopic osteochondral autograft transplantation in<br />

anterior cruciate ligament reconstruction: a preliminary clinical study.<br />

Knee Surg Sports Traumatol Arthrosc 1996;3:262<strong>–</strong>264<br />

28. Judas F, Rosa S, Teixeira L, et al. Chondrocyte viability in fresh and<br />

frozen large human osteochondral allografts: effect of cryoprotective<br />

agents. Transplant Proc 2007;39(8):2531<strong>–</strong>254<br />

29. Malinin TI, Wagner JL, Pita JC, et al. Hypothermic storage and<br />

cryopreservation of cartilage. Clin Orthop 1985;187:15<strong>–</strong>26<br />

30. Bugbee WD, Convery RF. Osteochondral allograft transplantation.<br />

Clin Sports Med 1999;18:67<strong>–</strong>75<br />

31. Sammarco VJ, Gorab R, Miller R, et al. Human articular cartilage<br />

storage in cell culture medium: guidelines for storage of fresh<br />

osteochondral allografts. Orthopedics 1997;20:497<strong>–</strong>500<br />

32. Gross AE, Shasha N, Aubin P. Long-term follow-up of the use of<br />

fresh osteochondral allografts for posttraumatic knee defects. Clin<br />

Orthop Relat Res 2005;435:79<strong>–</strong>87<br />

33. Ball ST, Amiel D, Williams SK, et al. The effects of storage on fresh<br />

human osteochondral allografts. Clin Orthop Relat Res 2004;418:246<strong>–</strong><br />

252<br />

34. Pearsall AW IV, Tucker JA, Hester RB, et al. Chondrocyte viability<br />

in refrigerated osteochondral allografts used for transplantation within<br />

the knee. Am J Sports Med 2004;32:125<strong>–</strong>131<br />

35. Malinin T, Temple HT, Buck BE. Transplantation of osteochondral<br />

allografts after cold storage. J Bone Joint Surg Am 2006;88(4):762<strong>–</strong><br />

770<br />

36. Vangsness CT Jr, Garcia IA, Mills CR, Kainer MA, Roberts MR,<br />

Moore TM. Allograft transplantation in the knee: tissue regulation,<br />

procurement, processing, and sterilization. Am J Sports Med. 2003<br />

May-Jun;31(3):474-481<br />

37. Gross AE. Use of fresh osteochondral allografts to replace traumatic<br />

joint defects. In: Czitrom AA, Gross AE, eds. Allografts in orthopaedic<br />

practice. Williams & Wilkins, 1992:67-82<br />

38. McDermott AGP, Langer F, Pritzker KPH, Gross AE. Fresh<br />

smallfragment osteochondral allografts. Clin Orthop 1985;197:96-102<br />

39. Stevenson S, Shaffer JW, Goldberg VM. The humoral response to<br />

vascular and nonvascular allografts of bone. Clin Orthop Relat Res.<br />

1996;(326):86-95<br />

40. Ghazavi MT, Pritzker KP, Davis AM, et al. Fresh osteochondral<br />

allografts for posttraumatic osteochondral defects of the knee. J Bone<br />

Joint Surg Br 1997;79:1008<strong>–</strong>1013<br />

41. Shasha N, Krywulak S, Backstein D. Long-term follow-up of fresh<br />

tibial osteochondral allografts for failed tibial plateau fractures. J Bone<br />

Joint Surg Am 2003;85(Suppl 2):33<strong>–</strong>39<br />

42. Jamali AA, Emmerson BC, Chung C, et al. Fresh osteochondral<br />

allografts: results in the patellofemoral joint. Clin Orthop Relat Res<br />

2005;437:176<strong>–</strong>185<br />

43. Gross AE, Silverstein EA, Falk J, et al. The allotransplantation of<br />

partial joints in the treatment of osteoarthritis of the knee. Clin Orthop<br />

Relat Res 1975;108:7<strong>–</strong>14<br />

44. Beaver RJ, Mahomed M, Backstein D, et al. Fresh osteochondral<br />

allografts for post-traumatic defects in the knee. A survivorship<br />

analysis. J Bone Joint Surg Br 1992;74:105<strong>–</strong>110


45. Flynn JM, Springfield DS, Mankin HJ. Osteoarticular allografts to<br />

treat distal femoral osteonecrosis. Clin Orthop 1994;303:38<strong>–</strong>43<br />

46. Gross AE, Agnidis Z, Hutchison CR. Osteochondral defects of the<br />

talus treated with fresh osteochondral allograft transplantation. Foot<br />

Ankle Int. 2001;22(5):385-391<br />

47. Raikin SM. Fresh osteochondral allografts for large-volume<br />

cystic osteochondral defects of the talus. J Bone Joint Surg Am.<br />

2009;91(12):2818-2826<br />

48. Görtz S, De Young AJ, Bugbee WD. Fresh osteochondral allografting<br />

for osteochondral lesions of the talus. Foot Ankle Int. 2010;31(4):283-<br />

290<br />

49. Hahn DB, Aanstoos ME, Wilkins RM. Osteochondral lesions of the<br />

talus treated with fresh talar allografts. Foot Ankle Int. 2010;31(4):277-<br />

282<br />

50. El-Rashidy H, Villacis D, Omar I, Kelikian AS. Fresh osteochondral<br />

allograft for the treatment of cartilage defects of the talus: a<br />

retrospective review. J Bone Joint Surg Am 2011;93(17):1634-1640<br />

51. Meehan R, McFarlin S, Bugbee W, Brage M. Fresh ankle<br />

osteochondral allograft transplantation for tibiotalar joint arthritis.<br />

Foot Ankle Int. 2005;26(10):793-802<br />

52. Giannini S, Buda R, Grigolo B, et al. Bipolar fresh osteochondral<br />

allograft of the ankle. Foot Ankle Int. 2010 Jan;31(1):38-46<br />

53. Meyers MH. Resurfacing of the femoral head with fresh<br />

osteochondral allografts. Long-term results. Clin Orthop Relat Res<br />

1985;197:111<strong>–</strong>114<br />

54. Johnson DL, Warner JJ. Osteochondritis dissecans of the humeral<br />

head: treatment with a matched osteochondral allograft. J Shoulder<br />

Elbow Surg 1997;6:160<strong>–</strong>163<br />

55. Chapovsky F, Kelly JD IV. Osteochondral allograft transplantation<br />

for treatment of glenohumeral instability. Arthroscopy 2005;21:1007<br />

56. Pylawka TK, Wimmer M, Cole BJ, Virdi AS, Williams JM. Impaction<br />

affects cell viability in osteochondral tissues during transplantation. J<br />

Knee Surg. 2007 Apr;20(2):105-110<br />

57. Borazjani BH, Chen AC, Bae WC, et al. Effect of impact on<br />

chondrocyte viability during insertion of human osteochondral grafts. J<br />

Bone Joint Surg Am. 2006;88(9):1934-1943<br />

58. Görtz S, De Young AJ, Bugbee WD. Fresh osteochondral allografting<br />

for steroid-associated osteonecrosis of the femoral condyles. Clin<br />

Orthop Relat Res. 2010;468(5):1269-1278<br />

59. McCulloch PC, Kang RW, Sobhy MH, et al. Prospective evaluation of<br />

prolonged fresh osteochondral allograft transplantation of the femoral<br />

condyle: minimum 2-year follow-up. Am J Sports Med 2007;35:411<strong>–</strong><br />

420<br />

60. LaPrade RF, Botker J, Herzog M, Agel J. Refrigerated osteoarticular<br />

allografts to treat articular cartilage defects of the femoral condyles.<br />

A prospective outcomes study. J Bone Joint Surg Am. 2009;91(4):805-<br />

811<br />

61. Davidson PA, Rivenburgh DW, Dawson PE, Rozin R. Clinical,<br />

histologic, and radiographic outcomes of distal femoral resurfacing<br />

with hypothermically stored osteoarticular allografts. Am J Sports Med.<br />

2007;35(7):1082-1090<br />

62. Williams RJ, Ranawat AS, Potter HG, Carter T, Warren RF. Fresh<br />

Stored Allografts for the Treatment of Osteochondral Defects of the<br />

Knee. J Bone Joint Surg Am. 2007;89(4):718-726<br />

63. Levy YD, Görtz S, Pulido PA, McCauley JC, Bugbee WD. Long-tem<br />

followup of fresh osteochondral allografting of the femoral condyle.<br />

Paper No. 709, presented at the annual meeting of the American<br />

Academy of Orthopaedic Surgeons, San Francisco, CA, February 7-11,<br />

2012<br />

64. D’Aubigne RM, Postel M. Functional results of hip arthroplasty with<br />

acrylic prosthesis. J Bone Joint Surg Am. 1954;36:451-475<br />

65. Irrgang JJ, Anderson AF, Boland AL, et al. Development and validation<br />

of the <strong>International</strong> Knee Documentation Committee subjective knee<br />

form. Am J Sports Med. 2001;29:600-613<br />

66. Insall JN, Dorr LD, Scott RD, Scott WN. Rationale of the Knee <strong>Society</strong><br />

clinical rating system. Clin Orthop Relat Res. 1989;(248):13-14<br />

Acknowledgments:<br />

Shiley Center for Orthopaedic Research and Education<br />

Extended Abstracts 155<br />

18.1<br />

Stem Cell Therapy for Joint <strong>Repair</strong><br />

F. Barry<br />

Galway/Ireland<br />

Introduction: Adult mesenchymal stem cells (MSCs) isolated<br />

from bone marrow and a variety of connective tissues have been<br />

extensively tested in the treatment of bone and caetilage repair and<br />

in osteoarthritis (OA). In addition, fully allogeneic transplantation<br />

of human MSCs is tolerated in the immunocompetent host and<br />

allogeneic therapy has been effective in the treatment of graftversus-host<br />

disease. However, there are many aspects of the<br />

biology of MSCs that are poorly described and a more exhaustive<br />

characterization is necessary to exploit these cells fully in the context<br />

of tissue repair. Adequate translation of MSC therapy will only be<br />

successful if the following are addressed: (1) development of new<br />

cell-specific markers, (2) deciphering the therapeutic mechanism<br />

of action and unravelling the paracine signals that contribute to<br />

tissue repair, (3) understanding clonal heterogeneity in cultured<br />

populations, (4) ensuring that batch variability is controlled and (5)<br />

understanding the nature of host immunomodulation by transplanted<br />

MSCs and allogencity. Further studies are also necessary to gaoin<br />

a comprehensive understanding of how MSCs may contribute to<br />

cartilage repair and how they may impede the progression of OA.<br />

This review will address aspects of the characterization of MSCs and<br />

the development of new markers and will also discuss their use in<br />

cartilage repair models.<br />

Content: Characterization of MSCs While the preclinical and clinical<br />

testing of hMSCs has advanced rapidly over the last few years,<br />

the development of efficient and standardised methods for their<br />

isolation and characterization has not. Most attempts to isolate<br />

these cells depend on adherent culture systems with a significant<br />

risk of preparing heterogeneous or poorly characterized populations.<br />

There is evidence that hMSCs isolated by current methods are not<br />

homogenous and in fact consist of mixtures of progenitors and<br />

other cells. The uncertain nature of primary isolates of hMSCs, the<br />

possibility of culture-induced heterogeneity and the lack of highly<br />

specific markers raise issues of regulatory compliance that may<br />

impede clinical testing. There is a risk that emerging clinical data<br />

will be compromised by this lack of standardization and that current<br />

methods of cell isolation may not result in the most therapeutically<br />

effective cells. The lack of MSC-specific markers has also made it<br />

difficult to construct meaningful hierarchical models of multilineage<br />

differentiation and to probe the in vivo antecedents of cultured<br />

cells. Presently there are several antibodies that are routinely used<br />

to characterize MSCs from human bone marrow by flow cytometry<br />

and other methods and some have been adopted as release tests<br />

for clinical grade cells. These include CD105, CD73, CD90, STRO-1<br />

and CD271. None of these represents a canonical marker of MSCs<br />

and therefore the homogeneity, reproducibility and consistency<br />

of isolated populations is not assured. MSC reagents have been<br />

generated using a variety of methods including monoclonal antibody<br />

technology, by screening a naive human antibody phage display<br />

library or by preparing aptamers. Despite these efforts specific and<br />

unambiguous markers have not been forthcoming. To overcome<br />

potential immune tolerance and to generate a new antibody<br />

discovery platform for hMSCs we prepared an avian immune phage<br />

display library. We hypothesized that the phylogenetic distance<br />

would greatly increase the probability of generating high affinity<br />

recombinant antibodies to MSC surface proteins and overcome<br />

the difficulties associated with conserved mammalian sequences.<br />

The use of the avian immunoglobulin system for generation of<br />

recombinant antibodies to human proteins has been demonstrated<br />

previously. In this study we immunized chickens with early passage<br />

hMSCs and generated an antibody phage display library from the<br />

spleen and marrow. After stringent screening of the library and the<br />

removal of antibodies reactive with peripheral blood mononuclear<br />

cells we randomly isolated four recombinant single-chain antibody<br />

fragments (scFvs) termed TMSC1, 2, 3 and 4. We found that these<br />

bound to a population of cells in human bone marrow that were<br />

CD45low, lineage negative and which were enriched in early passage<br />

directly plated hMSCs. One of the scFvs, TMSC3 was particularly<br />

useful in the direct isolation of MSCs from human marrow. TMSC3+<br />

cells isolated using magnetic cell separations (MACS) were<br />

fibroblastic, proliferative and exhibited trilineage differentiation.<br />

MSCs in <strong>Cartilage</strong> <strong>Repair</strong> There are several factors which influence<br />

the selection of cells for cartilage repair therapy. These include<br />

ease of isolation, differentiation potential, evidence of engraftment<br />

and tissue repair and the absence of a host immune response.<br />

Chondrocytes represent one type of repair cells that are cultured<br />

ex vivo and then delivered to the cartilage surface, but it is not<br />

clear that these are the best choice. Stem cells with chondrogenic


156<br />

Extended Abstracts<br />

potential may represent a more effective and more readily available<br />

cellular therapeutic for this purpose. Several tissue-engineering<br />

approaches have been used for cartilage repair, for example the<br />

fixation of implanted chondrocytes beneath a sutured flap of ectopic<br />

tissue or a collagen membrane. Other approaches have centered<br />

on the use of cells loaded on a scaffold and delivered to a lesion<br />

site. These methods are applicable to the repair of focal defects<br />

of defined dimensions but not to the treatment of complex lesions<br />

that cover a large surface area of the joint that may be associated<br />

with severe and progressive inflammatory conditions such as<br />

OA. It is attractive to hypothesize that MSCs could have a role in<br />

cartilage protection by direct resurfacing of the articular cartilage.<br />

However, it seems more likely, based on low levels of engraftment,<br />

that transplanted MSCs are effective mediators of repair by virtue<br />

of their capacity to deliver to the host a cascade of cytoprotective,<br />

antiaptoptotic, immunomodukatory or proangiogenic factors.<br />

Based in this principle, the capacity of MSCs to enhance tissue<br />

restoration in the complex degenerate environment of the OA joint<br />

may be high. The MSC Niche There is little clarity surrounding the<br />

niche, or tissue-specific microenvironment, in which MSCs reside.<br />

Despite this, they can be easily isolated and differentiated in vitro<br />

into chondrocytes which synthesize an abundant extracellular<br />

matrix rich in type II collagen, aggrecan and other cartilage-specific<br />

components. Despite the lack of understanding of these cells and<br />

their natural history, it is clear that they have therapeutic potential<br />

in a broad variety of clinical applications. At this point we have<br />

an incomplete understanding of the regulation of differentiation,<br />

commitment and plasticity of the MSC population isolated from<br />

marrow. What is clear is that their proliferative activity in vitro is<br />

high and that, when exposed to TGF-beta, they have a chondrogenic<br />

capacity that far exceeds that of primary chondrocytes in culture.<br />

It is also clear that MSCs are fully tolerated in an allogeneic setting<br />

when delivered to an immunocompetent host. This creates new<br />

opportunities for the therapeutic use of these cells without the need<br />

for a tissue biopsy. There are other properties of MSCs that make<br />

them attractive candidates in cartilage repair: (1) they have an ability<br />

to resist hypoxic stress and remain active after transplantation when<br />

host cells are compromised, (2) they may deliver a series of repair<br />

cytokines and chemokines to the transplantation site which stimulate<br />

a repair response in host cells and (3) they may have the capacity to<br />

migrate in a targeted fashion to injured tissues. Despite this, cellbased<br />

cartilage repair remains a challenging objective. Most efforts<br />

to date have relied on the local delivery of undifferentiated cells<br />

to the cartilage surface, often in association with a biocompatible<br />

scaffold. It is useful now to consider newer approaches, including<br />

the application of cells which are predifferentiated to match the<br />

tissue target, and to devise new approaches that will ensure optimal<br />

engraftment and retention at the site of injury. In the case of articular<br />

cartilage engraftment and retention of delivered cells is difficult to<br />

achieve.<br />

18.2<br />

Stem cell - based therapeutic approaches to joint surface repair<br />

C. De Bari<br />

Aberdeen/United Kingdom<br />

Introduction: Arthritis, with osteoarthritis (OA) accounting for most<br />

of this burden, is a leading cause of disability in the western world<br />

and its total costs have been estimated over $65 billion annually in<br />

United States. Post-traumatic OA represents 13% of knee OA and<br />

73% of ankle OA. Symptomatic chronic full-thickness defects of<br />

the knee joint surface require surgical treatment for both symptom<br />

relief and to prevent possible evolution towards OA. In a simplistic<br />

way, the evolution towards secondary OA may result from a failure<br />

of reparative processes to counteract the tissue damage induced<br />

by injuring factors. Hence, joint surface repair represents an<br />

important therapeutic goal. Regenerative medicine has opened up<br />

the unprecedented opportunity to promote repair through tissue<br />

regeneration (1). The current prototype of cell-based biological<br />

repair of the joint surface was first described in 1994 by Brittberg<br />

and colleagues (2), who reported treatment of symptomatic defects<br />

of the joint surface in humans by implantation of autologous,<br />

culture-expanded articular chondrocytes underneath a periosteal<br />

flap. Chondrocytes were obtained with a cartilage biopsy from a<br />

healthy and minor-load bearing area of the same joint surface.<br />

This study showed symptomatic relief in 14 out of 16 patients with<br />

lesions of the femoral condyle at 2 years follow up (2). Currently,<br />

ACI represents gold standard of cell therapy for cartilage repair<br />

with up to 20 years’ follow-up showing ACI being an effective and<br />

durable solution for the treatment of large full-thickness cartilage<br />

lesions of the knee joint (3). Articular chondrocytes, however, are<br />

difficult to expand because of their limited proliferative capacity<br />

and rapid de-differentiation in vitro, resulting in the loss of their<br />

capacity to form cartilage when transplanted in vivo (4). The use of<br />

mesenchymal stem cells (MSCs) as chondrocyte substitutes in an<br />

ACI-equivalent procedure is intensely pursued because MSCs are<br />

easily accessible, easy to isolate and to expand in culture, and they<br />

have ability to form cartilage and bone. In addition, MSCs appear to<br />

be immune privileged under specific conditions. Altogether, these<br />

properties would allow upscaling and generation of large batches of<br />

quality controlled MSC preparations ready for allogeneic use, thus<br />

circumventing the limitations and patient-to-patient variability of<br />

autologous cell protocols. Preclinical and clinical studies are needed<br />

to compare MSCs with articular chondrocytes to see whether<br />

implantation of MSCs will result in a cartilage tissue that is as<br />

durable as the one following implantation of articular chondrocytes.<br />

Importantly, the use of MSCs would extend the application of cellbased<br />

technologies to non-localised chronic lesions in OA patients,<br />

as reported (5).<br />

Content: Originally isolated from bone marrow, MSCs are now<br />

known to be obtainable virtually from any connective tissue of<br />

the adult human body (6). We previously reported the isolation<br />

and characterization of MSCs from the adult human synovium and<br />

periosteum, and provided evidence of their multipotency at the<br />

single cell level using a series of in vitro and in vivo assays (7-12).<br />

There is evidence to indicate that human MSCs from different tissues<br />

possess distinctive biological properties (13-14) and the issue<br />

remains as to whether MSCs would be capable of forming stable<br />

hyaline-like cartilage as opposed to the transient cartilage template<br />

that is replaced with bone during the process of endochondral<br />

ossification. The propensity of bone marrow MSCs to form a<br />

cartilage tissue with a high frequency of chondrocyte hypertrophy<br />

(15) could explain the excessive advancement of the bone front<br />

due to endochondral ossification and leading to a thinner articular<br />

cartilage as observed in experimental animals (16). The variability in<br />

the biological properties of MSC populations is likely to affect the<br />

outcome of clinical applications; hence, the need to standardize<br />

MSC bioprocessing to obtain MSC preparations with consistent and<br />

reproducible biological potency, quality-controlled for therapeutic<br />

applications with specific clinical indications (1). At the same<br />

time, the availability of different MSCs with distinct differentiation<br />

properties is also desirable because it allows a broader choice in the<br />

optimisation of different tissue repair protocols. For example, MSCs<br />

with a preferential osteogenic differentiation would be desirable<br />

for preparation of biological bone substitutes while for articular<br />

cartilage repair the ideal MSCs should be able to form hyaline-like<br />

cartilage that is resistant to endochondral ossification. The use of<br />

MSCs, in suspension or in combination with biomaterials, requires<br />

assessment of potency for quality control in order to ensure clinical<br />

effectiveness at least in structural outcome. The availability of<br />

large batches of “off-the-shelf” quality-controlled cell populations<br />

will enhance consistency of treatments while abating costs; it will<br />

also eliminate the need for two operations and enable large scale


production. This however poses obvious risks of rejection. There<br />

is evidence that MSCs can be poorly immunogenic in vivo under<br />

specific conditions (17). However, the differentiation into a mature<br />

phenotype of the implanted stem cells is likely to result in the loss<br />

of the immunological privilege with consequent rejection. MSCs<br />

could also be implanted immediately after their purification. A<br />

huge effort in the field is directed towards developing therapeutic<br />

protocols employing freshly purified cells in a one-stop procedure.<br />

Minimally manipulated cellular preparations are still required to<br />

undergo identity characterization and potency assessment, but they<br />

are expected to greatly simplify autologous procedures, deliverable<br />

to patients in one single intervention, and the regulatory paths. In<br />

addition to the use of ex vivo manipulated MSCs, another approach<br />

to the repair of the joint surface could be the activation of intrinsic<br />

regenerative mechanisms by using medications that target the<br />

stem cells naturally present in their own environments and related<br />

reparative signalling pathways. In this respect, several jointassociated<br />

tissues such as synovial membrane and fluid, fat pad,<br />

periosteum, bone marrow, and even the articular cartilage itself,<br />

have been reported to contain cells that, after isolation and culture<br />

expansion, display properties of MSCs. However, it remains to be<br />

seen whether “professional” MSCs exist in vivo in their own tissue/<br />

organ environments or are a clinically relevant artefact due to ex vivo<br />

manipulations. Recently, by combining a double nucleoside labelling<br />

scheme with a clinically relevant mouse model of joint surface injury<br />

(18), we provided data on the identification and characterization<br />

of endogenous resident MSC niches in the knee joint synovium in<br />

vivo (19). We are currently investigating endogenous MSCs in joint<br />

tissues, their role in the mechanisms underlying articular cartilage<br />

maintenance and healing and, more generally, how signals in the<br />

niches are coupled to functional events and related outcomes in joint<br />

homeostasis, remodelling and repair. Bone marrow transplantation<br />

in haematology best illustrates the success of a cell therapy that<br />

has evolved with the increasing understanding of cell phenotypes,<br />

functions, and niches. Classical ACI has already evolved into<br />

second and third generation ACI. In a near future, novel strategies<br />

for biological joint resurfacing will be developed in parallel to<br />

refinements of ACI-based protocols. It is anticipated that the type<br />

of intervention will be dependent on clinical indication and factors<br />

such as size and depth of the lesion, status of the surrounding<br />

cartilage and other joint tissues. Interventions will not be mutually<br />

exclusive and will be likely to span from ACI (with chondrocytes or<br />

stem cells) to pharmacological targeting, by using drugs, of the joint<br />

stem cell niches in order to regenerate joint tissues. Ultimately, such<br />

interventions will have to address two main issues: symptom relief<br />

and durable repair for effective prevention of OA.<br />

References:<br />

1. Roberts S, Genever P, McCaskie A, De Bari C. Prospects of stem<br />

cell therapy in osteoarthritis. Regen Med. 2011; 6 (3): 351-66. 2.<br />

Brittberg M, Lindahl A, Nilsson A, Ohlsson C, Isaksson O, Peterson<br />

L. Treatment of deep cartilage defects in the knee with autologous<br />

chondrocyte transplantation. N Engl J Med. 1994; 331 (14): 889-95<br />

3. Peterson L, Vasiliadis HS, Brittberg M, Lindahl A. Autologous<br />

chondrocyte implantation: a long-term follow-up. Am J Sports Med.<br />

2010; 38 (6): 1117-24. 4. Dell’Accio F, De Bari C, Luyten FP. Molecular<br />

markers predictive of the capacity of expanded human articular<br />

chondrocytes to form stable cartilage in vivo. Arthritis Rheum.<br />

2001; 44 (7): 1608-19. 5. Wakitani S, Imoto K, Yamamoto T, Saito<br />

M, Murata N, Yoneda M. Human autologous culture expanded bone<br />

marrow mesenchymal cell transplantation for repair of cartilage<br />

defects in osteoarthritic knees. Osteoarthritis <strong>Cartilage</strong> 2002; 10<br />

(3): 199-206. 6. Augello A, Kurth TB, De Bari C: Mesenchymal stem<br />

cells: a perspective from in vitro cultures to in vivo migration and<br />

niches. Eur Cell Mater. 2010; 20: 121-33. 7. De Bari C, Dell’Accio F,<br />

Tylzanowski P, Luyten FP. Multipotent mesenchymal stem cells from<br />

adult human synovial membrane. Arthritis Rheum. 2001; 44 (8):<br />

1928-42. 8. De Bari C, Dell’Accio F, Luyten FP: Human periosteumderived<br />

cells maintain phenotypic stability and chondrogenic<br />

potential throughout expansion regardless of donor age. Arthritis<br />

Rheum. 2001; 44 (1): 85-95. 9. De Bari C, Dell’Accio F, Vandenabeele<br />

F, Vermeesch JR, Raymackers JM, Luyten FP. Skeletal muscle repair<br />

by adult human mesenchymal stem cells from synovial membrane.<br />

J Cell Biol. 2003; 160 (6): 909-18. 10. De Bari C, Dell’Accio F, Luyten<br />

FP. Failure of in vitro-differentiated mesenchymal stem cells from<br />

the synovial membrane to form ectopic stable cartilage in vivo.<br />

Arthritis Rheum. 2004; 50 (1): 142-50. 11. De Bari C, Dell’Accio F,<br />

Vanlauwe J, Eyckmans J, Khan I, Archer CW, Jones E, McGonagle D,<br />

Pitzalis C, Luyten FP: Mesenchymal multipotency of adult human<br />

periosteal cells demonstrated by single cell lineage analysis.<br />

Arthritis Rheum. 2006; 54 (4): 1209-21. 12. Karystinou A, Dell’Accio<br />

F, Kurth TB, Wackerhage H, Khan IM, Archer CW, Jones EA, Mitsiadis<br />

Extended Abstracts 157<br />

TA, De Bari C: Progenitor cell subsets revealed by clonal analysis of<br />

human synovial membrane mesenchymal stem cells. Rheumatology<br />

(Oxford). 2009; 48 (9): 1057-64. 13. Sakaguchi Y, Sekiya I, Yagishita<br />

K, Muneta T. Comparison of human stem cells derived from various<br />

mesenchymal tissues: superiority of synovium as a cell source.<br />

Arthritis Rheum. 2005; 52 (8): 2521-9. 14. De Bari C, Dell’Accio F,<br />

Karystinou A, Guillot PV, Fisk NM, Khan IM, Archer CW, Jones EA,<br />

McGonagle D, Mitsiadis TA, Donaldson AN, Luyten FP, Pitzalis C.<br />

A biomarker-based mathematical model to predict bone forming<br />

potency of human synovial and periosteal mesenchymal stem<br />

cells. Arthritis Rheum. 2008; 58 (1): 240-50. 15. Scotti C, Tonnarelli<br />

B, Papadimitropoulos A, Scherberich A, Schaeren S, Schauerte<br />

A, Lopez-Rios J, Zeller R, Barbero A, Martin I. Recapitulation of<br />

endochondral bone formation using human adult mesenchymal stem<br />

cells as a paradigm for developmental engineering. Proc Natl Acad<br />

Sci U S A. 2010; 107 (16): 7251-6. 16. Qiu YS, Shahgaldi BF, Revell WJ,<br />

Heatley FW. Observations of subchondral plate advancement during<br />

osteochondral repair: a histomorphometric and mechanical study<br />

in the rabbit femoral condyle. Osteoarthritis <strong>Cartilage</strong> 2003; 11 (11):<br />

810-20. 17. MacDonald G, Augello A, De Bari C: Role of mesenchymal<br />

stem cells in reestablishing immunologic tolerance in autoimmune<br />

rheumatic diseases. Arthritis Rheum. 2011; 63 (9): 2547-57. 18.<br />

Eltawil NM, De Bari C, Achan P, Pitzalis C, Dell’Accio F: A novel in vivo<br />

murine model of cartilage regeneration. Age and strain-dependent<br />

outcome after joint surface injury. Osteoarthritis <strong>Cartilage</strong>. 2009; 17<br />

(6): 695-704. 19. Kurth TB, Dell’Accio F, Crouch V, Augello A, Sharpe<br />

PT, De Bari C. Functional mesenchymal stem cell niches in adult<br />

mouse knee joint synovium in vivo. Arthritis Rheum. 2011; 63 (5):<br />

1289-300.<br />

Acknowledgments:<br />

The author is grateful to support to his research from Arthritis<br />

Research UK.<br />

19.1.1<br />

Basics on ultrastructural-multiparametric MR techniques<br />

S. Apprich 1 , G.H. Welsch 1 , S. Zbyn 1 , B. Schmitt 1 , S. Trattnig 2<br />

1 Vienna/Austria, 2 Wien/Austria<br />

Introduction: Osteoarthritis (OA) changes in hyaline articular<br />

cartilage are characterized by important changes in the biochemical<br />

composition of cartilage. The macromolecular network of cartilage<br />

consists mainly of collagen and proteoglycans. Normally, the<br />

highly organized collagen network serves as the tissue’s structural<br />

framework and is the principal source of tensile and shears strength.<br />

Glucosaminoglycans (GAGs) are repeating disaccharides with<br />

carboxyl and sulfate groups attached to the larger aggrecan molecule<br />

(proteoglycan) that is part of the extracellular matrix network of<br />

cartilage. GAG molecules possess considerable net negative charge<br />

and confer compressive strength to the cartilage. Loss of GAGs and<br />

increased water content represent the earliest stage of cartilage<br />

degeneration, while the collagenous component of the extracellular<br />

matrix still remains intact. Several MR imaging techniques are<br />

available that enable detection of biochemical changes that precede<br />

the morphologic degeneration in cartilage. All of these techniques<br />

attempt to selectively demonstrate the GAG components and/or the<br />

collagen fiber network of the extracellular matrix and are usually<br />

summarized as “compositional imaging” of cartilage.<br />

Content: Delayed Gadolinium enhanced MRI of <strong>Cartilage</strong> (dGEMRIC)<br />

The dGEMRIC and sodium (23 Na) MR imaging techniques are based<br />

on similar principles, with positive sodium ions being attracted by<br />

the negatively fixed charged density of the GAG side chains. These<br />

electrostatic forces are responsible for a direct relationship between<br />

the local sodium concentration and fixed charged density with a<br />

strong correlation between fixed charged density and GAG content<br />

(1,2). dGEMRIC is based on the fact that GAGs contain negatively<br />

charged side chains, which lead to an inverse distribution of<br />

negatively charged contrast agent molecules (eg, gadolinium) with<br />

respect to GAG concentration (3,4). Drawbacks of this technique<br />

are the need to use a double dose of a gadolinium-based contrast<br />

agent (0.2 mmol per kilogram of body weight) and the requirement<br />

for a delay between intravenous administration of the agent and<br />

the start of the MR examination (usually 60<strong>–</strong>90 minutes) to allow<br />

complete penetration of the contrast agent into the cartilage. Varus<br />

malalignment is associated with a lower dGEMRIC index on the medial<br />

side, while the opposite trend is evident in valgus malalignment<br />

(5). Correlations between dGEMRIC index and pain, as measured


158<br />

Extended Abstracts<br />

by the Western Ontario and McMaster Universities Arthritis Index,<br />

were evident in patients with hip dysplasia (5). dGEMRIC studies<br />

have demonstrated that moderate exercise can improve knee<br />

cartilage GAG (estimated by T1 in the presence of gadopentetate<br />

dimeglumine) in patients at high risk for OA (6). In patients with an<br />

injury to the anterior cruciate ligament, lower GAG concentrations<br />

were found in the medial compartment of the femoral and tibial<br />

articular cartilage of the injured knee when compared with the<br />

contralateral (uninjured) knee (7). In patients with femoroacetabular<br />

impingement, correlations were observed between dGEMRIC index,<br />

pain, and alpha angle, suggesting that hips with more femoral<br />

deformity may show signs of early OA (8).<br />

T1rho mapping T1 rho is a time constant that characterizes magnetic<br />

relaxation of spins under the influence of a radiofrequency field<br />

parallel to the spin magnetization. The resultant contrast is sensitive<br />

to the low-frequency interactions between water molecules and their<br />

local macromolecular environment, such as GAG and collagen, which<br />

are the main constituents of the extracellular matrix in cartilage. In<br />

early studies, changes in T1 rho were found in proteoglycan-depleted<br />

cartilage plugs, but, on the other hand, other investigators (9-13)<br />

reported that T1 rho did not correspond to a modified dGEMRIC<br />

technique or to the proteoglycan distribution seen at histologic<br />

examination, which suggests that several factors contribute to<br />

variations in T1 rho . Recently, rapid 3D in vivo T1 rho mapping<br />

techniques of knee cartilage at high field strength (3 T) have been<br />

developed and applied in patients with OA (14,15). Other in vivo<br />

studies (15) have shown increased cartilage T1 rho values in patients<br />

with OA compared with those values in control subjects, which<br />

suggests the potential of T1 rho imaging for noninvasive evaluation<br />

of diseased cartilage. In cartilage overlying traumatic bone marrow<br />

lesions, the average T1 rho values were significantly higher than<br />

those in surrounding cartilage, demonstrating that macromolecular<br />

changes in cartilage may be related to traumatic bone marrow damage<br />

(12). Sodium imaging Previous research (1, 16, 17) has already shown<br />

that 23 Na MR imaging has a potential advantage over conventional<br />

proton MR imaging for the investigation of biochemical markers in<br />

cartilage during the early stages of OA. Although 23 Na MR imaging<br />

has high specificity and does not require any exogenous contrast<br />

agent, it does require special hardware (multinuclear) capabilities,<br />

specialized RF coils (transmit-receive sodium coils), and, likely, 3D<br />

very short echo time sequences. These challenges currently limit the<br />

use of 23 Na MR imaging in the clinical and research settings. T2 and<br />

T2* mapping T2 mapping has been used to describe the composition<br />

of hyaline articular cartilage in the knee joint on the basis of collagen<br />

structure and hydration (18). In addition to the transverse relaxation<br />

time (T2) of articular cartilage, T2* relaxation measures have recently<br />

been investigated (2) for depiction of the collagen matrix. In healthy<br />

articular cartilage, an increase in T2 values from deep to superficial<br />

cartilage layers can be observed; this is based on the anisotropy of<br />

collagen fibers running perpendicular to cortical bone in the deep<br />

layer of cartilage and parallel to the surface in the superficial layer<br />

(19). Therefore, zonal evaluation of articular cartilage is important<br />

in T2 analyses. Analyses of T2 relaxation times in the knee have<br />

been performed previously, usually at 1.5 T or, more recently, 3.0<br />

T, demonstrating the ability to depict abnormalities before there<br />

is evident morphologic change. In vivo MR imaging studies (20)<br />

have demonstrated that cartilage T2 values are related to age.<br />

<strong>Cartilage</strong> T2 values seem to be associated with the severity of OA,<br />

and there are variations between tibial and femoral cartilage T2 (21).<br />

A significant correlation between cartilage T2 and the severity and<br />

grade of cartilage and meniscus lesions has been demonstrated.<br />

Subjects with high activity levels had significantly higher prevalence<br />

and grade of abnormalities and higher T2 values than did subjects<br />

with low activity levels (22). Chemical Exchange Saturation Transfer<br />

(gagCEST) Chemical exchange saturation transfer (CEST) (23)imaging<br />

has recently been presented as a technique with the potential<br />

to measure PG content in cartilage. This technique exploits the<br />

biochemical properties of GAG, i.e., the chemical exchange of labile<br />

protons with bulk water (gagCEST). It was shown that labile <strong>–</strong>NH<br />

(δ=3.2 ppm offset from the water resonance) and <strong>–</strong>OH (δ=0.9 to 1.9<br />

ppm) protons of GAG can be used as CEST agents through selective<br />

saturation of their resonance signals (23). This selectivity is also the<br />

fundamental difference between gagCEST and T1 rho relaxation, with<br />

the latter being caused by a sum of non-distinguishable exchange<br />

effects. Recent studies aimed mostly at general optimization of<br />

gagCEST imaging technique, but also the feasibility of gagCEST<br />

imaging in patients was demonstrated at 7 Tesla (24). In the latter<br />

study, a strong correlation was found between gagCEST results and<br />

sodium imaging, which is a sensitive and highly specific method to<br />

determine cartilage GAG content at 7 Tesla.<br />

References:<br />

1 . Shapiro EM , Borthakur A , Gougoutas A , Reddy R . 23Na MRI<br />

accurately measures fi xed charge density in articular cartilage.<br />

Magn Reson Med 2002 ; 47 (2): 284 <strong>–</strong> 291 .<br />

2 . Welsch GH , Mamisch TC , Hughes T , et al . In vivo biochemical 7.0<br />

Tesla magnetic resonance: preliminary results of dGEMRIC, zonal<br />

T2, and T2* mapping of articular cartilage . Invest Radiol 2008 ; 43<br />

(9): 619 <strong>–</strong> 626<br />

3 . Bashir A , Gray ML , Burstein D . Gd-DTPA2-as a measure of<br />

cartilage degradation .Magn Reson Med 1996 ; 36 (5): 665 <strong>–</strong> 673 .<br />

4 . Bashir A , Gray ML , Boutin RD , Burstein D . Glycosaminoglycan<br />

in articular cartilage: in vivo assessment with delayed Gd(DTPA)<br />

(2-)-enhanced MR imaging . Radiology 1997 ; 205 (2): 551 <strong>–</strong> 558 .<br />

5 . Kim YJ , Jaramillo D , Millis MB , Gray ML , Burstein D . Assessment<br />

of early osteoarthritis in hip dysplasia with delayed gadoliniumenhanced<br />

magnetic resonance imaging of cartilage . J Bone Joint<br />

Surg Am 2003 ; 85-A (10): 1987 <strong>–</strong> 1992 .<br />

6 . Roos EM , Dahlberg L . Positive effects of moderate exercise<br />

on glycosaminoglycan content in knee cartilage: a four-month,<br />

randomized, controlled trial in patients at risk of osteoarthritis .<br />

Arthritis Rheum 2005 ; 52 (11): 3507 <strong>–</strong> 3514 .<br />

7 . Fleming BC , Oksendahl HL , Mehan WA , et al . Delayed gadoliniumenhanced<br />

MR imaging of cartilage (dGEMRIC) following ACL injury .<br />

Osteoarthritis <strong>Cartilage</strong> 2010 ; 18 (5): 662 <strong>–</strong> 667 .<br />

8 . Jessel RH , Zilkens C , Tiderius C , Dudda M , Mamisch TC , Kim<br />

YJ . Assessment of osteoarthritis in hips with femoroacetabular<br />

impingement using delayed gadolinium enhanced MRI of cartilage<br />

. J Magn Reson Imaging 2009 ; 30 (5): 1110 <strong>–</strong> 1115 ..<br />

9 . Duvvuri U , Reddy R , Patel SD , Kaufman JH , Kneeland JB , Leigh<br />

JS . T1rho-relaxation in articular cartilage: effects of enzymatic<br />

degradation . Magn Reson Med 1997 ; 38 (6): 863 <strong>–</strong> 867 .<br />

10 . Akella SV , Regatte RR , Gougoutas AJ , et al .Proteoglycaninduced<br />

changes in T1rhorelaxation of articular cartilage at 4T .<br />

Magn Reson Med 2001 ; 46 (3): 419 <strong>–</strong> 423 .<br />

11 . Menezes NM , Gray ML , Hartke JR , Burstein D . T2 and T1rho<br />

MRI in articular cartilage systems . Magn Reson Med 2004 ; 51 (3):<br />

503 <strong>–</strong> 509 .<br />

12 . Regatte RR , Akella SV , Wheaton AJ , et al . 3D-T1rho-relaxation<br />

mapping of articular cartilage: in vivo assessment of early<br />

degenerative changes in symptomatic osteoarthritic subjects . Acad<br />

Radiol 2004 ; 11 (7): 741 <strong>–</strong> 749 .<br />

13 . Mlynárik V , Szomolányi P , Toffanin R , Vittur F , Trattnig S<br />

.Transverse relaxation mechanisms in articular cartilage . J Magn<br />

Reson 2004 ; 169 (2): 300 <strong>–</strong> 307 .<br />

14 . Li X , Han ET , Busse RF , Majumdar S .In vivo T(1rho) mapping<br />

in cartilage using 3D magnetization-prepared angle-modulated<br />

partitioned k-space spoiled gradient echo snapshots (3D MAPSS) .<br />

Magn Reson Med 2008 ; 59 (2): 298 <strong>–</strong> 307 .<br />

15 . Li X , Benjamin Ma C , Link TM , et al . In vivo T(1rho) and T(2)<br />

mapping of articular cartilage in osteoarthritis of the knee using 3 T<br />

MRI . Osteoarthritis <strong>Cartilage</strong> 2007 ; 15 (7): 789 <strong>–</strong> 797 .<br />

16 . Shapiro EM , Borthakur A , Dandora R , Kriss A , Leigh JS , Reddy<br />

R . Sodium visibility and quantitation in intact bovine articular<br />

cartilage using high fi eld (23)Na MRI and MRS . J Magn Reson 2000<br />

; 142 (1): 24 <strong>–</strong> 31 .<br />

17 . Borthakur A , Shapiro EM , Beers J , Kudchodkar S , Kneeland JB<br />

, Reddy R . Sensitivity of MRI to proteoglycan depletion in cartilage:<br />

comparison of sodium and proton MRI . Osteoarthritis <strong>Cartilage</strong><br />

2000 ; 8 (4): 288 <strong>–</strong> 293 .<br />

18 . Mosher TJ , Dardzinski BJ . <strong>Cartilage</strong> MRI T2 relaxation time<br />

mapping: overview and applications . Semin Musculoskelet Radiol<br />

2004 ; 8 (4): 355 <strong>–</strong> 368 .<br />

19 . Mosher TJ , Smith HE , Collins C , et al .Change in knee cartilage<br />

T2 at MR imaging after running: a feasibility study . Radiology 2005<br />

; 234 (1): 245 <strong>–</strong> 249 . 20 . Mosher TJ , Dardzinski BJ , Smith MB .<br />

Human articular cartilage: influence of aging and early symptomatic


degeneration on the spatial variation of T2—preliminary findings at<br />

3 T . Radiology 2000 ; 214 (1): 259 <strong>–</strong> 266 .<br />

21 . Dunn TC , Lu Y , Jin H , Ries MD , Majumdar S . T2 relaxation<br />

time of cartilage at MR imaging: comparison with severity of knee<br />

osteoarthritis . Radiology 2004 ; 232 (2): 592 <strong>–</strong> 598 .<br />

22 . Stehling C , Liebl H , Krug R , et al . Patellar cartilage: T2 values<br />

and morphologic abnormalities at 3.0-T MR imaging in relation to<br />

physical activity in asymptomatic subjects from the osteoarthritis<br />

initiative . Radiology 2010 ; 254 (2): 509 <strong>–</strong> 520 . concentration in vivo<br />

by chemical exchange-dependent saturation transfer (gagCEST).<br />

Proc Natl Acad Sci 2008;105(7):2266-2270.<br />

23. Schmitt B, Zbyn S, Stelzeneder D, Jellus V, Paul D, Lauer L,<br />

Bachert P, Trattnig S. <strong>Cartilage</strong> Quality Assessment by Using<br />

Glycosaminoglycan Chemical Exchange Saturation Transfer and<br />

23Na MR Imaging at 7 T. Radiology 2011;260(1):257-264.<br />

19.1.2<br />

Clinical application of ultrastructural-multiparametric MR<br />

techniques in patients after repair surgery<br />

G.H. Welsch<br />

Vienna/Austria<br />

Introduction: Articular cartilage lesions are a common pathology of<br />

the knee joint and many patients could benefit from cartilage repair.<br />

Such surgical treatment options may offer the possibility for patients<br />

with cartilage defects to avoid the development of osteoarthritis<br />

or delay its progression. Different sophisticated cartilage repair<br />

techniques, including arthroscopic or open surgical approaches, as<br />

well as marrow-stimulation techniques, osteochondral grafting, and<br />

chondrocyte implantation/transplantation, require knowledgeable<br />

and high quality follow-up. Although clinical findings are the primary<br />

criteria, a more objective outcome measure would possibly have the<br />

ability to provide predictive values. Advanced magnetic resonance<br />

imaging (MRI) is able to depict the morphological and biochemical<br />

condition of the cartilage repair tissue and the surrounding<br />

structures [1-4]. Within the ICRS a consensus paper on the recent<br />

possibilities and advances in imaging of cartilage repair tissue has<br />

been published in the journal cartilage [5]. An ideal MRI protocol<br />

for articular cartilage and cartilage repair should provide accurate<br />

assessment of cartilage thickness and volume, reveal information<br />

about signal changes within cartilage, and demonstrate clear<br />

delineation of the cartilage surface, the cartilage and bone interface,<br />

as well as the subchondral bone and also provide information about<br />

the biochemical composition of articular cartilage and cartilage repair<br />

tissue. To evaluate such MRI protocols, cartilage repair procedures,<br />

such as arthroscopic or open surgical approaches as well as marrowstimulation<br />

techniques, osteochondral grafting, and chondrocyte<br />

implantation/transplantation could be used. The uniqueness of<br />

cartilage repair in the use of advanced morphological and especially<br />

biochemical MR methodologies is bases in the fact that these<br />

procedures include defined areas of repair cartilage in addition to an<br />

often-intact surrounding cartilage in mostly young patients. Thus,<br />

modified and healthy cartilage can be compared within one subject.<br />

Nevertheless, the aim of an ideal MRI protocol must be its ease of<br />

implementation in scientific studies as well as day-to-day clinical<br />

routine. The combination of high-resolution morphological MR<br />

evaluation with biochemical assessment in a clinically applicable<br />

scan time needs to exploit high-field MRI (1.5T, 3.0T) together with<br />

advanced imaging techniques and sophisticated coil technology.<br />

Aim of this manuscript is to demonstrate advanced MRI techniques<br />

to depict the morphological and especially the biochemical<br />

constitution of cartilage repair tissue as a multiparametric approach<br />

in the follow-up of cartilage repair procedures.<br />

Content: Morphological MRI The morphological shape of the repair<br />

tissue, the degree of repair filling (1), the integration of the cartilage<br />

repair tissue to the border zone (2), the structure of the surface (3),<br />

the structure of the whole repair tissue (4), the signal intensity (5),<br />

the constitution of the subchondral lamina (6), the the constitution<br />

of the subchondral bone (7), possible adhesions (8), and possible<br />

effusion (9) can be combined in the nine variables of the magnetic<br />

resonance observation of cartilage repair tissue (MOCART) scoring<br />

system [6] which is claimed to allow subtle and suitable assessment<br />

of the articular cartilage repair tissue. The MR assessment of the<br />

MOCART score is based on standard MR sequences, also<br />

recommended by the <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong> (ICRS)<br />

Extended Abstracts 159<br />

[7]. Depending on the locality of the area of cartilage repair, the MR<br />

evaluation of the cartilage repair tissue is performed on sagittal,<br />

axial or coronal two-dimensional (2D) planes using high spatial<br />

resolution together with a slice thickness of 2-4 mm. Following the<br />

current standard procedure, the recommended MR sequences by<br />

the ICRS and the recommended sequences for the MOCART scoring<br />

visualize the area of cartilage repair and the adjacent cartilage as<br />

well as the surrounding structures in 2D. In contrast, new isovoxel<br />

sequences have the potential for high-resolution isotropic imaging<br />

with a voxel size down to 0.4mm3, and can be reformatted in every<br />

plane without any loss of spatial resolution. Hence the cartilage<br />

repair tissue can be visualized three-dimensionally (3D) in every<br />

plane, its subsequent classification and grading by an MR-based<br />

scoring system benefits and a new 3D-MOCART [8]. This new MOCART<br />

score can be achieved by standard 2D MR sequences as well as by<br />

modern isotropic MR sequences. Besides the possibility of a<br />

depiction of the repair tissue in more than one plane, other benefits<br />

of this new MOCART score are the better and more precise description<br />

of the repair tissue itself however also of the underlying subchondral<br />

bone plate. Hence e.g. the cartilage hypertrophy can be scored in<br />

more detail. Within the subchondral bone, osteophytes (as very<br />

often present after marrow stimulating techniques) are included in<br />

the score. Furthermore other changes within the subchondral bone<br />

and the bone marrow are better included into this score scheme.<br />

This results in now 11 variables and a better scoring algorithm. This<br />

helps especially in longitudinal evaluations of a specific patient over<br />

time. dGERMIC and other glycosamnioglycan specific techniques Of<br />

the major macromolecules, glycosamnioglycans (GAG), are important<br />

to the cartilage tissue’s biochemical and biomechanical function.<br />

GAG are the main source of fixed charge density (FCD) in cartilage,<br />

often decreased in reparative cartilage after cartilage repair [9].<br />

Intravenously administered gadolinium diethylenetriamine<br />

pentaacetate anion, (Gd-DTPA2-) penetrates the cartilage through<br />

both the articular surface and the subchondral bone. The contrast<br />

equilibrates in inverse relation to the FCD, which is, in turn, directly<br />

related to the GAG concentration; therefore, T1, which is determined<br />

by the Gd-DTPA2- concentration, becomes a specific measure of<br />

tissue GAG concentration, suggesting that T1 mapping enhanced by<br />

delayed administration of Gd-DTPA2- (T1 dGEMRIC) has the potential<br />

for monitoring GAG content of cartilage in vivo [10; 11]. Besides<br />

standard inversion recovery (IR) evaluation, a new approach for fast<br />

T1 mapping has shown promising results and is increasing the<br />

clinical applicability of the dGEMRIC technique [12]. Since GAG<br />

content is responsible for cartilage function, particularly its tensile<br />

strength, the monitoring of the development of GAG content in<br />

cartilage repair tissues may provide information about the quality of<br />

the repair tissue. A recent study by our group showed dGEMRIC to<br />

be able to differentiate between different cartilage repair tissues<br />

with higher delta DR1 values, and thus, lower GAG content for<br />

cartilage repair tissue after MFX compared to MACT [13]. As the<br />

mapping of the GAG concentration is desirable in the follow-up of<br />

cartilage repair procedures and the presented dGEMRIC technique<br />

has the limitation of contrast agent administration and a time delay<br />

before post-contrast MRI, a recently described technique for the<br />

assessment of GAG concentration in vivo by chemical exchangedependent<br />

saturation transfer (CEST) may have potential in future<br />

applications on articular cartilage [14]. At high fields and ultra-high<br />

fields, sodium MR imaging might become a gold-standard in future<br />

approaches. Sodium MRI has been validated as direct and<br />

quantitative method of computing FCD and, hence, proteoglycan<br />

content. Achieving high enough signal-to-noise ratio, the sensitivity<br />

of sodium MRI is high enough for detecting small changes in<br />

proteoglycan content. T2 and other collagen specific techniques<br />

Changes in water and collagen content and tissue anisotropy are<br />

most frequently analyzed using the transverse relaxation time (T2)<br />

of cartilage [15]. The collagen fiber orientation, its three-dimensional<br />

organization and curvature can be reliably visualized by cartilage T2<br />

mapping, however influencing its appearance at 55° (with respect to<br />

the main magnetic field (B0)) resulting in the magic angle [16; 17]. In<br />

healthy articular cartilage, an increase in T2 values from deep to<br />

superficial cartilage layers can be observed based on the anisotropy<br />

of collagen fibers running perpendicular to cortical bone in the deep<br />

layer of cartilage [18]. Histologically validated animal studies have<br />

shown this zonal increase in T2 values as a marker of hyaline or<br />

hyaline-like cartilage structure after cartilage repair procedures<br />

within the knee [19; 20]. To visualize this zonal variation is essential<br />

for cartilage T2 mapping and high enough spatial resolution has to<br />

be provided. In cartilage repair tissue, full-thickness T2 values, as<br />

zonal evaluation have been shown able to visualize cartilage repair<br />

maturation [21; 22]. Another study by our group further showed the<br />

ability of zonal T2 evaluation to differentiate cartilage repair tissue<br />

after MFX and MACT [23]. In a clinical approach, the differentiation of<br />

two matrices after MACT was possible based on T2 mapping showing<br />

the dependency of the ultra-structure of the repair tissue based on


160<br />

Extended Abstracts<br />

the initial matrix it was build from / seeded on [24]. Hence the<br />

specific collagen structure of the repair tissue can be depicted and<br />

quantified by T2 relaxation time mapping. In addition to standard 2D<br />

multi-echo spin-echo T2 relaxation, T2*- weighted 3D gradient-echo<br />

articular cartilage imaging has shown reliable results in the<br />

evaluation of chondromalacia of the knee [25]. In recent studies, T2*<br />

mapping, with its potentially short scan times, was correlated to<br />

standard T2, and showed information comparable to that obtained<br />

for articular cartilage in the knee, but with overall lower T2* values<br />

(ms) [26]. Furthermore, also for T2*, a clear zonal variation between<br />

deep and superficial cartilage layers can be shown. Another<br />

advanced approach to combine morphological and biochemical T2<br />

cartilage imaging is based on a Double-Echo-Steady-State (DESS)<br />

sequence, which generates two signal echoes that are characterized<br />

by a different contrast behavior where the underlying T2 can be<br />

calculated. Further emerging biochemical MR techniques that are<br />

gaining more and more importance are, besides others, diffusion<br />

weighted imaging (DWI), magnetization transfer contrast (MTC) or<br />

T1rho. The cartilage composition or component visualized by these<br />

methodologies is not yet clearly validated. However initial studies<br />

are showing very promising results and additional information<br />

besides dGERMIC and T2 can be gained also in patients after cartilage<br />

repair. Concluding, a combination of morphological and biochemical<br />

MRI might provides the chance to gain a precise follow-up of the<br />

knee joint and may offer a predictive value for the future development<br />

and performance of the cartilage repair tissue and the joint. Ongoing<br />

approaches in MRI of joint after cartilage repair surgery try to<br />

furthermore include the state of the whole joint as assessed e.g. by<br />

the Whole-Organ Magnetic Resonance Imaging Score (WORMS) in<br />

relation to the area of cartilage repair. This is of outmost importance<br />

as the status of the whole joint predicts the follow-up of the repaired<br />

joint possibly even more as the repair tissue itself.<br />

References:<br />

1 Potter HG, et. al. Clin Sports Med, 28:77-94. ^<br />

2 Trattnig S, et. al. Eur Radiol, 19:1582-1594.<br />

3 Welsch GH, et. al. Semin Musculoskelet Radiol, 12:196-211. doi:<br />

10.1055/s-0028-1083104<br />

4 Welsch GH, et. al. J Magn Reson Imaging, 33:180-188.<br />

5 Trattnig S, et. al. <strong>Cartilage</strong>, 2:5-26.<br />

6 Marlovits S, et. al. Eur J Radiol, 52:310-319.<br />

7 Brittberg M, et. al. J Bone Joint Surg Am, 85-A Suppl 2:58-69.<br />

8 Welsch GH, et. al. Invest Radiol, 44:603-612.<br />

9 Watanabe A, et. al. Radiology, 239:201-208.<br />

10 Tiderius CJ, et. al. Magnet Reson Med, 49:488-492.<br />

11 Williams A, et. al. Am J Roentgenol, 182:167-172.<br />

12 Trattnig S, et. al. J Magn Reson Imaging, 26:974-982.<br />

13 Trattnig S, et. al.Eur Radiol.<br />

14 Ling W, et. al. Proceedings of the National Academy of Sciences of<br />

the United States of America, 105:2266-2270.<br />

15 Mosher TJ, et. al.Semin Musculoskelet Radiol, 8:355-368.<br />

16 Goodwin DW, et. al. Acad Radiol, 5:790-798.<br />

17 Goodwin DW, et. al. AJR Am J Roentgenol, 174:405-409.<br />

18 Smith HE, et. al. J Magn Reson Imaging, 14:50-55.<br />

19 Watrin-Pinzano A, et. al.Magn Reson Mater Phy, 17:219-228.<br />

20 White LM, et. al. Radiology, 241:407-414.<br />

21 Trattnig S, et. al. Invest Radiol, 42:442-448. 22 Welsch GH, et. al.<br />

J Orthop Res, 27:957-963.<br />

23 Welsch GH, et. al. Radiology, 247:154-161.<br />

24 Welsch GH, et. al. Am J Sports Med, 38:934-942.<br />

25 Murphy BJ (et. al.Skeletal Radiol, 30:305-311.<br />

26 Welsch GH, et. al.Eur Radiol. 2010<br />

Acknowledgments:<br />

The authors achnowledge funding of the Austrian National Founds<br />

(OENB, FWF) and the German Research Foundation (DFG)<br />

19.1.3<br />

Quantitative X-ray and ultrasound methodology for cartilage<br />

repair<br />

J.S. Jurvelin 1 , T. Viren 1 , H. Kokkonen 1 , J. Liukkonen 1 , K. Kulmala 1 , A.<br />

Joukainen 1 , I. Kiviranta 2 , J. Salo 1 , H. Kröger 1 , J. Töyräs 1<br />

1 Kuopio/Finland, 2 Helsinki/Finland<br />

Introduction: Improved possibilities for cartilage reconstruction<br />

require accurate diagnostic tools both to evaluate the results,<br />

and to find out the right patients early enough to take advantage<br />

of these treatment methods.At present, quantitative MRI methods<br />

provide noninvasive means to follow the outcome of cartilage<br />

repair. Unfortunately, due to limited availability and high costs,<br />

MRI investigations may not always be readily at disposal. Further,<br />

limited resolution of MRI impair its applicability for initial cartilage<br />

diagnostics. Traditional diagnostic techniques of osteoarthritis (OA),<br />

including clinical examination and plain X-ray imaging can only detect<br />

late and major tissue changes. Using arthroscopic inspection and<br />

probe palpation, cartilage injury is possible to evaluate and grade,<br />

but the reliability of this classification is poor, especially in the early<br />

cartilage changes (Spahn et al. 2011). Due to immature diagnostics,<br />

many cartilage defects are only seen incidentally during arthroscopy.<br />

This can be seen as one reason to delayed increase in the number<br />

of cartilage repair with modern techniques. Novel noninvasive or<br />

minimally invasive methods are needed to successfully develop new<br />

cartilage repair approaches. Especially, sensitive and quantitative<br />

methods should be in use to evaluate the extent and severity of the<br />

original injury and to follow the healing process. In the past, several<br />

methodologies, based on mechanical (Lyyra et al. 1995), acoustic<br />

(Laasanen et al. 2003), electric (Quenneville et al. 2004) and optical<br />

(Han et al. 2003) techniques have been proposed for evaluation<br />

purposes. However, none of those has propagated to extensive<br />

clinical use.<br />

Content: Based on long-term basic science research two novel<br />

quantitative techniques, relying on use of contrast agent enhanced<br />

computed tomography (CECT) (Palmer et al. 2006, Silvast et al.<br />

2009, Aula et al. 2009), or high frequency ultrasound (Viren et<br />

al. 2009, Huang et al. 2009) are under development for cartilage<br />

diagnostics. Both techniques have been systematically developed<br />

for cartilage repair assesment in in vitro and in situ studies with<br />

animal models and are now in the stage of in vivo testing in humans.<br />

Contrast enhanced computed tomography (CECT) Analogously to<br />

delayed gadolinium enhanced MRI of cartilage (dGEMRIC), CECT<br />

has been introduced for quantitative imaging of articular cartilage<br />

proteoglycan (PG) content (Palmer et al. 2006). In this method<br />

anionic contrast agent is assumed to distribute into cartilage<br />

inversely proportionally to spatial distribution of fixed charge<br />

density (FCD). It has also been shown that in degenerated, or in<br />

acutely injured, cartilage significantly higher contrast agent intake<br />

locates in injured tissue than in intact cartilage (Kokkonen et al.<br />

2011). CECT technique enables simultaneous quantitative analysis<br />

of the properties of articular cartilage and subchondral bone (Aula<br />

et al. 2009). Subtle subchondral changes have been shown to be<br />

important in OA diagnostics. An additional advantage is higher<br />

resolution with the use of isotrophic voxels (conical beam CT),<br />

allowing free reconstruction of curved joint facet surfaces. Our<br />

earlier study showed that CECT combined with finite element (FE)<br />

analysis could be used to determine solute diffusion coefficients<br />

which appeared to depend on the composition and structure of the<br />

cartilage (Kulmala et al. 2010). Therefore, instead of investigating<br />

the equilibrium distribution of the contrast agent, the nonequilibrium<br />

diffusion properties of cartilage monitored with CECT<br />

may be assessed to evaluate articular cartilage of clinical patients<br />

in vivo (Kokkonen et al. 2011). Further, the optimal time points and<br />

other practical issues regarding the in vivo imaging were evaluated.<br />

Contrast agent concentration maximum in the cartilage was<br />

achieved at 30 to 60 minutes after the injection and the CT-contrast<br />

agent concentration in the knee joint was still adequate at two<br />

hours after the injection. Normalized contrast agent concentration<br />

was higher in the OA knee than in the healthy knee. Thus, CECT<br />

showed potential for clinical evaluation of cartilage integrity. The<br />

first in vivo CECT of human knees have been conducted using intraarticular<br />

injection, however, premises to use intravenous injection<br />

for introduction of the contrast agent are under investigations. The<br />

challenge is to obtain contrast agent concentrations in cartilage<br />

high enough for quantitation with CECT (Bansal et al. 2012).. Miniinvasive<br />

ultrasound arthroscopy Ultrasound imaging application,<br />

based on FDA approved ultrasound catheters (Viren et al. 2009,<br />

Kaleva et al. 2011), enables imaging of cartilage surfaces during<br />

arthroscopy. Catheters are thin, and they give a detailed structure of<br />

cartilage with high resolution (~40 μm) Further, several quantitative<br />

ultrasound backscatter parameters, such as integrated reflection


coefficient (IRC), apparent integrated backscatter (AIB) and<br />

ultrasound roughness index (URI), sensitive to integrity of cartilage<br />

and subchondral bone, can be calculated. Articular surfaces of<br />

bones from horse intercarpal joints, featuring both intact tissue<br />

and spontaneously healed chondral or osteochondral defects, were<br />

imaged ex vivo with an arthroscopic ultrasound device. It was possible<br />

to detect lesion sites and adjacent intact tissue based on IRC, AIB<br />

and URI. The ultrasound backscattering from the inner structures of<br />

the cartilage matched well with the histological findings, reflecting<br />

abnormal intrinsic collagen architecture within repaired cartilage.<br />

Surface roughness and the integrity of the intact and repaired rabbit<br />

cartilage could also be quantitatively evaluated with the minimally<br />

invasive ultrasound technique. Furthermore, qualitative information<br />

about integration of the repair tissue, integrity of the surface of<br />

repair tissue and the internal structure could be extracted from the<br />

ultrasound images. In the clinical setting, the diagnostic potential<br />

of high frequency ultrasound is two-fold. It can be used during<br />

arthroscopy as an additional method to see the detailed structure of<br />

cartilage, or it can be seen as a possible diagnostic tool to map the<br />

condition of cartilage on the outpatient visits. We have already used<br />

it successfully during arthroscopy in diagnostics of the earliest signs<br />

of OA, severity of acute cartilage injuries and the outcome of surgical<br />

repair of the injuries. The technique has revealed the high diagnostic<br />

potential, and it will be integrated more closely with the arthroscopic<br />

instrumentation. Further, consistent quantitative measurements set<br />

need for the perpendicularity of the ultrasound pulse incidence as<br />

well as for accurate localization of measurements. These issues<br />

need to be further investigated and technically solved. Additionally,<br />

the optimal frequency or frequency range remains to be determined,<br />

as it determines the iamge resolution and penetration of the<br />

ultrasound into the tissue. Correction of the effects of overlying<br />

cartilage in bone measurements should also be further investigated.<br />

As the thin ultrasound probe can be inserted into joint through a<br />

small needle, the use of the technique without arthroscopic control<br />

is investigated.<br />

References:<br />

Aula AS et al.: Simultaneous computed tomography of articular<br />

cartilage and subchondral bone. Osteoarthritis <strong>Cartilage</strong> 17:1583-8,<br />

2009<br />

Bansal PN et al.: Cationic contrast agents improve quantification of<br />

glycosaminoglycan (GAG) content by contrast enhanced CT imaging<br />

of cartilage. J Orthop Res Dec 23 [Epub ahead of print]<br />

Han CW at al.: Analysis of rabbit articular cartilage repair after<br />

chondrocyte implantation using optical coherence tomography.<br />

Osteoarthritis <strong>Cartilage</strong> 11: 111-21, 2003;<br />

Huang YP, Zheng YP. Intravascular Ultrasound (IVUS): A Potential<br />

Arthroscopic Tool for Quantitative Assessment of Articular <strong>Cartilage</strong>.<br />

Open Biomed Eng J 26:13-20, 2009<br />

Kaleva E et al.: Arthroscopic ultrasound assessment of articular<br />

cartilage in human knee joint: A potential diagnostic method.<br />

<strong>Cartilage</strong> 2: 246-53, 2011<br />

Kokkonen HT et al.: Contrast enhanced computed tomography of<br />

human knee cartilage in vivo. Baltic bone and cartilage, Malmö,<br />

2011<br />

Kulmala KA et al.: Diffusion coefficients of articular cartilage for<br />

different CT and MRI contrast agents. Med Eng Phys. 32: 878-82,<br />

2010<br />

Laasanen MS et al.: Mechano-acoustic diagnosis of cartilage<br />

degeneration and repair. J Bone Joint Surg Am. 2003;85-A Suppl<br />

2:78-84.<br />

Lyyra T et al.: Indentation instrument for the measurement of<br />

cartilage stiffness under arthroscopic control. Med Eng Phys. 1995;<br />

17(5):395-9.<br />

Palmer AW et al.: Analysis of cartilage matrix fixed charge density<br />

and three-dimensional morphology via contrast-enhanced<br />

microcomputed tomography. Proc Natl Acad Sci U S A. 103: 19255-<br />

60, 2006<br />

Quenneville E et al.: Fabrication and characterization of nonplanar<br />

microelectrode array circuits for use in arthroscopic diagnosis of<br />

cartilage diseases. IEEE Trans Biomed Eng. 2004; 51(12):2164-73.<br />

Extended Abstracts 161<br />

Silvast TS et al.: Diffusion and near-equilibrium distribution of MRI<br />

and CT contrast agents in articular cartilage. Phys Med Biol 54:<br />

6823-36, 2009<br />

Spahn G et al. Reliability in arthroscopic grading of cartilage lesions:<br />

results of a prospective blinded study for evaluation of inter-observer<br />

reliability. Arch Orthop Trauma Surg. 131: 377-81, 2011<br />

Virén T et al.: Minimally invasive ultrasound method for intraarticular<br />

diagnostics of cartilage degeneration. Ultrasound Med Biol<br />

35:1546-5, 2009<br />

Acknowledgments:<br />

Financial support from the Academy of Finland, Sigrid Jusélius<br />

Foundation, Sakari Sohlberg Foundation, Kuopio University Hospital<br />

(EVO 5041715) and University of Eastern Finland (strategic spearhead<br />

funding) is acknowledged.<br />

19.2.1<br />

Current Treatment Paradigm for Single-Stage <strong>Cartilage</strong> <strong>Repair</strong><br />

A.H. Gomoll<br />

Boston/United States of America<br />

Introduction: <strong>Cartilage</strong> repair remains in evolution. Single stage<br />

technologies appear attractive to reduce cost and patient morbidity<br />

alike. This presentation will review several of these technologies that<br />

are currently either on the market or in later stage clinical trials.<br />

Content: Single stage procedures can be divided in cell-free<br />

implants, mainly scaffolds, and cell-based implants, which are<br />

further sub-divided according to whether auto- and allograft tissue<br />

is being utilized. From a US perspective, there are currently no<br />

cell-free implants available on the market. Several devices are in<br />

current late-stage trials that are based on multi-phasic collagen<br />

plugs for osteochondral repair. Autograft cell-based technologies<br />

are attractive, and one device (CAIS, Mitek, Raynham, USA) is<br />

currently undergoing phase III evaluation. Autologous cartilage is<br />

harvested with a special shaving device, morcellized and secured<br />

onto a resorbable polymer mesh with fibrin glue, which in turn is<br />

stapled into the defect. Allograft cartilage has traditionally been<br />

used in fresh osteochondral allograft transplantation. However,<br />

the limited time span between graft harvest and implantation<br />

complicates availability and scheduling. Therefore, a new preserved<br />

osteochondral allograft (Chondrofix, Zimmer, Warsaw, USA) has<br />

entered the market last year. During processing, cells and lipids<br />

are being removed from the tissue, allowing shelf-storage at room<br />

temperature. For defects limited to the articular surface with intact<br />

subchondral bone, a cartilage-only allograft product (DeNovo NT,<br />

Zimmer, Warsaw, USA) has become available in the US, which<br />

utilizes morcellized juvenile (ave. donor age 3 years) cartilage,<br />

which is secured into the defect with fibrin glue. Both CAIS and<br />

DeNovo NT rely on chondrocyte migration out of the cartilage tissue<br />

with subsequent matrix production to fill the defect. Pre-clinical<br />

models have been encouraging, but clinical evidence so far has been<br />

limited. Lastly, an allogenic cell-based implant (DeNovo ET, Zimmer,<br />

Warsaw, USA) is in late stage clinical trial in the US, using allogenic<br />

chondrocytes that are grown in a matrix-free process to form a disklike<br />

implant.


162<br />

Extended Abstracts<br />

19.2.2<br />

Emerging technologies<br />

A.A.M. Dhollander 1 , K.F. Almqvist 1 , P.C. Verdonk 2 , R. Verdonk 1 , G.<br />

Verbruggen 3 , J. Victor 1<br />

1 Gent/Belgium, 2 Gent-zwijnaarde/Belgium, 3 Ghent/Belgium<br />

Introduction: In the synovial joints, tissues such as cartilage,<br />

meniscus/acetabular labrum, subchondral bone and the musclecapsule-ligament<br />

complex constitute a functional unit, which is<br />

called the synovial joint organ. Chondrocytes in articular cartilage<br />

use mechanical signals in conjunction with other environmental and<br />

genetic factors to regulate their metabolic activity. This results in<br />

an alteration of the structure and composition of the extracellular<br />

matrix (ECM) to meet physical demands of the body. Articular<br />

cartilage serves to absorb mechanical shocks and to distribute joint<br />

loads more evenly across the underlying bone structures. Articular<br />

cartilage lesions are found during the course of degenerative joint<br />

disease, or as a result of articular trauma. In normal circumstances,<br />

ECM homeostasis is maintained by an equilibrium between anabolic<br />

and catabolic pathways. The most relevant growth factors and<br />

cytokines known to be involved in cartilage metabolism are produced<br />

by the chondrocytes themselves. Excessive mechanical load due to<br />

obesity, malalignment, occupation, … can lead to structural changes<br />

in joints and finally to joint degeneration. The first detectable<br />

event is a loss of PGs, followed by the disruption of the collagen<br />

network. The initially smal focal degenerative lesions may gradually<br />

increase in depth and length. Chondocytes react to these changes<br />

by increasing their metabolic activity. However, catabolic processes<br />

predominate over the anabolic ones.<br />

Content: Surgical treatment for articular cartilage injury is of major<br />

interest to orthopaedic surgeons because most lesions of articular<br />

cartilage do not heal spontaneously and may predispose the<br />

joint to the subsequent development of secondary osteoarthritis.<br />

Various techniques have been used to treat this injury with variable<br />

success rates. The ultimate aim of treatment is the restoration of<br />

normal knee function by regenerating hyaline cartilage in the defect<br />

and complete integration of the regenerated cartilage with the<br />

surrounding cartilage and underlying bone. Currently, three major<br />

trends can be observed among one-stage cartilage repair strategies:<br />

one trend based on bone marrow stimulating techniques, one based<br />

on acellular scaffolds and finally, cellular based techniques. Several<br />

marrow-stimulating procedures directed at the recruitment of bone<br />

marrow cells have been widely used to treat local cartilage defects.<br />

In these type of procedures, mesenchymal stem cells (MSCs)<br />

migrate in the fibrin network of the blood clot. However, the fibrin<br />

clot is not mechanically stable to withstand the tangential forces. An<br />

implanted exogenous scaffold may improve the mechanical stability<br />

of the fibrin clot and its specific molecular composition may provide<br />

a proper stimulus for chondrogenic differentiation and cartilage<br />

regeneration. In this presentation we will discuss the combination<br />

of microfracture with a collagen type I/III scaffold (Autologous<br />

Matrix Induced Chondrogenesis, AMIC) (1) and platelet-rich plasma<br />

(PRP, AMIC plus) (2) and with a cell-free matrix chondrotissue (3),<br />

which consists of an absorbable non-woven polyglycolic acid textile<br />

treated with hyaluronic acid. Then two acellular scaffold designs will<br />

be discussed. The use of these bioabsorbable scaffolds for repair<br />

of chondral and osteochondral defects has recently been explored<br />

in laboratory and preclinical investigations. Such matrix scaffolds,<br />

implanted alone or in combination with cells, allow immediate<br />

filling of the defect and support local migration of chondrogenic<br />

and osteogenic cells that synthesize new ground substance. First,<br />

we will give an update about the TruFit plug (Smith & Nephew,<br />

Andover, MA) (4). This osteochondral scaffold plug is an acellular<br />

synthetic polymer scaffold that is inserted into the osteochondral<br />

bone to provide a stable scaffold that in theory could encourage<br />

the regeneration of a full thickness of articular cartilage to repair<br />

chondral defects. Secondly, we will discuss the MaioRegen<br />

scaffold. This is an osteochondral nanostructured biomimetic<br />

scaffold (FinCeramica Faenza SpA, Faenza, Italy). It has a porous,<br />

3-dimensional composite, trilayered structure, to reproduce the<br />

cartilaginous layer, the tide mark, and the subchondral bone (5).<br />

In the last part of the lecture, we will highlight one-stage cellularbased<br />

approaches. It is now over 20 years since the first patient was<br />

operated on with autologous chondrocyte implantation (ACI). This<br />

two-stage technique has gained wide scientific and clinical support<br />

for use in the repair of focal articular lesions. However, during in<br />

vitro propagation of the chondrocytes, dedifferentiation of the cells<br />

can occur, and afterward these fibroblast-like chondrocytes show<br />

different biosynthetic properties than the original cartilage cells in<br />

the knee joint. To avoid the issue of dedifferentiation during in vitro<br />

propagation of chondrocytes, instantaneously delivered allogenic<br />

chondrocytes could be used. In this part of the lecture, we will<br />

discuss the use of a biodegradable, alginate-based biocompatible<br />

scaffold containing human allogenic chondrocytes for the treatment<br />

of cartilage lesions in the knee (6). Finally, we will talk about a new<br />

one-stage cellular based approach combining chondrocytes with<br />

mesenchymal stem cells seeded on a polymer-based scaffold.<br />

References:<br />

· Benthien J.P., Behrens P. Autologous matrix-induced chondrogenesis<br />

(AMIC) combining microfracturing and a colla- gen I/III matrix for<br />

articular cartilage resurfacing. <strong>Cartilage</strong>. 2010; 1:65-68.<br />

· Dhollander A.A.M., De Neve F., Almqvist K.F., Verdonk R., Lambrecht<br />

S., Elewaut D., Verbruggen G., Verdonk P.C.M.: Autologous Matrix<br />

Induced Chondrogenesis (AMIC) combined with Platelet Rich Plasma<br />

Gel (PRP). Technical description and a five pilot patients report. Knee<br />

Surg. Sports Traumatol. Arthrosc., 2011, 19, 536-542.<br />

· Dhollander A.A.M., Verdonk P.C.M., Lambrecht S., Almqvist<br />

K.F., Elewaut D., Verbruggen G., Verdonk R.: The combination of<br />

microfracture and a cell-free polymer-based implant immersed with<br />

autologous serum for cartilage defect coverage. Knee Surg. Sports<br />

Traumatol. Arthrosc. 2011, Nov 9 [epub ahead of print].<br />

· Dhollander A.A.M., Liekens K., Almqvist K.F., Verdonk R., Lambrecht<br />

S., Elewaut D., Verbruggen G., Verdonk P.C.M.: A Pilot Study of the<br />

Use of an Osteochondral Scaffold Plug for <strong>Cartilage</strong> <strong>Repair</strong> in the<br />

Knee and How to Deal with Early Clinical Failures. Arthroscopy, 2011,<br />

Oct 18 [epub ahead of print].<br />

· Kon E., Delcogliano M., Filardo G., Busacca M., Di Martino A.,<br />

Marcacci M.: Novel nano-composite multilayered biomaterial for<br />

osteochondral regeneration: a pilot clinical trials. Am J Sports Med.,<br />

2011, 39, 1180-1190.<br />

· Dhollander A.A.M., Verdonk P.C.M., Lambrecht S., Verdonk R.,<br />

Elewaut D., Verbruggen G., Almqvist K.F.: Mid-term Results of the<br />

Treatment of <strong>Cartilage</strong> Defects in the Knee using Alginate Beads<br />

containing Human Mature Allogenic Chondrocytes. Am. J. Sports<br />

Med., 2011, Sep 29 [epub ahead of print].<br />

Acknowledgments:<br />

The authors like to thank Dr. W. Huysse and Prof. Dr. K. Verstraete<br />

from the department of Radiology, Ghent University Hospital,<br />

Belgium for their support concerning the use of MRI facilities.<br />

19.2.3<br />

Concomitant procedures<br />

A. Getgood<br />

Cambridge/United Kingdom<br />

Introduction: The cause of full thickness articular cartilage injury is<br />

multifactorial. Traumatic injury is often associated with meniscus<br />

and ligament damage, whilst early degeneration has been shown<br />

to be associated with lower limb mal-alignment. The combination<br />

of altered kinematics and reduced biological function can lead to<br />

progressive articular cartilage damage and ultimately osteoarthritis.<br />

When faced with a patient with a symptomatic articular cartilage<br />

injury, the philosophy of addressing the biomechanical function of<br />

the joint first, prior to performing articular cartilage restoration is<br />

followed. A number of articular cartilage repair technologies have<br />

been developed which aim to restore the mechanical and biological<br />

function of the articulating surface. However, for these to be<br />

successful, the root cause of the problem should be identified and<br />

addressed. This presentation will outline the different concomitant<br />

procedures available and will describe the experience of using them<br />

in combination with articular cartilage restoration.<br />

Content: Patient assessment A full history and examination is<br />

performed, during which the personality of both the patient and the<br />

cartilage defect is ascertained. The patients’ occupation and activity<br />

level is important, as may preclude certain procedures. Associated<br />

medical conditions are of significance, as the complication rate<br />

associated with osteotomy has been shown to be greater in diabetics<br />

and smokers. A clear understanding of patients’ symptoms is<br />

important. Does pain predominate, or is instability the major issue.<br />

Is there evidence of mechanical symptoms? Physical examination


will include assessment of gait, looking for evidence of dynamic<br />

laxity, assessment of alignment, core stability, quadriceps tone<br />

and function and evidence of synovitis or effusion. It is important<br />

to try to ascertain which compartment is predominantly affected,<br />

to see if this correlates with the imaging. A thorough exam of the<br />

ligaments, looking for evidence of laxity, as well as the tracking<br />

and control of the patella is made. Plain radiographs including AP/<br />

lateral and 30 degree flexed weight bearing PA views are routinely<br />

ordered. If there is evidence of patellofemoral involvement, a skyline<br />

view is requested. Standardised weight bearing long leg alignment<br />

views are also requested. Further imaging will include MRI scan<br />

with articular cartilage specific sequences, and if any history of<br />

bony trauma, a CT scan. A period of rehabilitation is recommended,<br />

which may help alleviate symptoms, but also optimise the<br />

patients condition pre-operatively. This is particularly important in<br />

patellofemoral conditions. Surgical decision making: A full profile of<br />

the patient and the associated articular cartilage lesion can now be<br />

made once all of the imaging is available. If non-operative measures<br />

have failed, the decision to perform articular cartilage restoration<br />

with or without concomitant procedures is made. The philosophy<br />

is to correct the biomechanical environment of the knee prior to<br />

performing any biological procedures. The goal is to restore normal<br />

alignment, ligamentous stability and structure and function of the<br />

meniscus. A combination of re-alignment osteotomy, ligament<br />

reconstruction, meniscus reconstruction/allograft transplantation<br />

can be performed along with cartilage repair, however, depending<br />

on how many procedures are required to be performed, a staged<br />

reconstruction may be more appropriate in some circumstances. Realignment<br />

osteotomy: Long leg alignment radiographs are used to<br />

ascertain the mechanical axis of the lower limbs. Measurement of<br />

the distal femoral and proximal tibial articular angles are also made<br />

to work out where the deformity is, thereby allowing the deformity<br />

correction to be addressed on the appropriate side of the joint[1].<br />

In a knee with an articular cartilage lesion, the preference is to<br />

unload the joint out of the affected compartment, thereby providing<br />

a favourable environment for the repair procedure to mature. On<br />

the tibial side the opening wedge technique is preferred on medial<br />

and lateral sides for varus and valgus deformities respectively. If<br />

performing a lateral opening wedge varus osteotomy, care must be<br />

taken not to create joint line obliquity of greater than 10 degrees.<br />

On the femoral side, the valgus deformity is most common, in which<br />

a medial closing wedge technique is preferred. In patellofemoral<br />

cases, anteromedialisaton of the tibial tubercle can be utilised to<br />

both offload the joint surface, but also improve patella tracking,<br />

particularly if the tibial tubercle-trochlea groove (TT-TG) distance<br />

is greater than 20mm[2]. Ligament reconstruction: Ligamentous<br />

instability is addressed with a mixture of reconstructive techniques.<br />

Anatomic anterior cruciate ligament reconstruction is performed with<br />

the use of either hamstring or patella tendon autograft. Collateral<br />

ligament instability may be addressed with either autograft,<br />

allograft of occasionally synthetic ligaments. If multiple ligament<br />

reconstruction is to be performed, a combination of grafts may often<br />

be utilised. A combination of ligament reconstruction and osteotomy<br />

can be used to address combined coronal and sagittal plane laxity. In<br />

the presence of patella instability, a medial patellofemoral ligament<br />

reconstruction may be indicated. Meniscus reconstruction: The<br />

primary goal of meniscus surgery is to preserve tissue, particularly<br />

on the lateral side. Meniscus repair should therefore be performed<br />

when possible. Biological adjuncts, such as platelet rich plasma,<br />

fibrin clots and microfracture of the intercondylar notch can be used<br />

to try and augment the healing process. In the event of segmental<br />

failure, scaffolds are used to re-build the meniscus and restore<br />

structure and function[3]. The pre-requisite for meniscal scaffolds,<br />

is an intact peripheral rim and posterior and anterior horn. Meniscus<br />

allograft transplantation: In the event of meniscus rim or anterior/<br />

posterior horn loss, arthroscopic meniscus allograft transplantation<br />

is performed. Deep frozen allografts are implanted into the knee<br />

and held with sutures, attached to the posterior and anterior horns,<br />

delivered through bone tunnels and tied over a bony bridge on the<br />

anteromedial tibia. The periphery is then sutured to the capsule via<br />

a combination of inside-out and all-inside suture devices. Saving the<br />

failing knee: Patients presenting with ICRS grade IV degenerative<br />

change in one or more compartment, associated with meniscus loss,<br />

were treated with meniscus allograft transplantation in combination<br />

with one or more of osteotomy, ligament reconstruction and articular<br />

cartilage repair. Outcome data was collected pre-operatively then<br />

annually including IKDC, Lysholm, Tegner and KOOS scores. MRI and/<br />

or second look arthroscopy was also performed at one year post op.<br />

Results: Of the 58 meniscus allograft transplants performed in our<br />

institution, 26 patients had grade IV ICRS degenerative change and<br />

had combination ‘salvage surgery’. The mean age at surgery was 37<br />

years (19<strong>–</strong>49). 19 have minimum 12 month follow up with 17 (89%)<br />

of those rated as good or excellent at latest follow-up. MRI showed<br />

normal appearances of the graft in 17, with minimal graft extrusion.<br />

Extended Abstracts 163<br />

Second look arthroscopy was performed in 9 showing encouraging<br />

recovery of the chondral surface. Eight showed good peripheral<br />

integration of the graft, with only 1 failure requiring removal (15<br />

months). One patient has been converted to arthroplasty at 4 years.<br />

Three required arthroscopic arthrolysis for adhesions. Conclusions:<br />

Salvage of the failing knee can be successful if a number of<br />

procedures are combined to firstly address biomechanical deficits,<br />

followed by biological reconstruction. It remains unclear as to which<br />

of these procedures have the greatest impact on patient symptom<br />

relief and cessation of disease progression.<br />

References:<br />

1. Brinkman JM, Lobenhoffer P, Agneskirchner JD, et al. Osteotomies<br />

around the knee: patient selection, stability of fixation and bone<br />

healing in high tibial osteotomies. J Bone Joint Surg Br 2008;90<br />

1548-57<br />

2. Caton JH, Dejour D. Tibial tubercle osteotomy in patello-femoral<br />

instability and in patellar height abnormality. Int Orthop 2010; 34<br />

305-9<br />

3. Efe T, Getgood A, Schofer MD, et al. The safety and short-term<br />

efficacy of a novel polyurethane meniscal scaffold for the treatment<br />

of segmental medial meniscus deficiency. Knee Surg Sports<br />

Traumatol Arthrosc 2011;<br />

Acknowledgments:<br />

This work was not supported by any external funding source.<br />

19.3.1<br />

Can biomarkers be used as outcome measures in cartilage repair<br />

and osteoarthritis?<br />

S. Lohmander<br />

Lund/Sweden<br />

Introduction: The monitoring of cartilage regeneration and<br />

osteoarthritis in natural history or intervention studies carries with<br />

it the need for outcome measures. Without appropriate outcome<br />

measures, results cannot be documented or validated. We should<br />

consider patient-reported outcomes on symptoms, function and<br />

quality of life as the gold standard, the clinical endpoint. Other<br />

outcomes may include functional tests, imaging techniques to<br />

monitor structure and quality of joint tissues, and molecular<br />

biomarkers to reflect the turnover, structure and state of joint tissues.<br />

Studies on cartilage repair and osteoarthritis with currently available<br />

outcome measures require long observation times and trials, and<br />

there is therefore a great need for new measures that could predict<br />

the long-term clinical outcome after a shorter observation time.<br />

Considerable efforts are therefore being invested in the development<br />

of biomarkers that reflect normal and pathological processes in the<br />

joint. Biomarkers developed for osteoarthritis will likely find use<br />

also in studies of cartilage repair and regeneration.<br />

Content: For osteoarthritis biomarkers, a terminology named<br />

BIPEDS was proposed, which classifies these biomarkers into five<br />

categories corresponding to their proposed use: Burden of disease,<br />

Investigational, Prognostic, Efficacy of Intervention, Diagnostic and<br />

Safety. Biomarkers that are likely to have the earliest beneficial<br />

impact on clinical trials fall into two categories. The first are those<br />

markers that would allow us to select for trials subjects that are<br />

most likely to respond or progress (prognostic markers) within<br />

a reasonable time for a clinical study, which for an osteoarthritis<br />

study could be one to two years. The second category of biomarkers<br />

includes those that provide early feedback for preclinical decisionmaking<br />

and for trial organizers that an intervention has the desired<br />

effect on the primary molecular target (efficacy markers). Both types<br />

of biomarkers are highly desirable in chronic conditions where<br />

conventional clinical outcomes may take years to present. These<br />

two categories of biomarkers could reduce the burden and risk of<br />

clinical trials by delivering essential early information, speeding up<br />

the product development cycle and making osteoarthritis a more<br />

manageable target for developing new drugs. Many biomarkers are<br />

being tested in relation to their utility for studies on osteoarthritis,<br />

and exist in various states of validation. The term validation here<br />

refers to the evidence for a marker in support of its use as a surrogate<br />

endpoint, and includes verification of analytical performance


164<br />

Extended Abstracts<br />

characteristics and correlation of the biomarker with a specific<br />

biological process. Naturally, validation of a biomarker against a<br />

gold standard endpoint depends critically also on the performance<br />

and specificity of that gold standard endpoint. A second useful<br />

classification system divides biomarkers into four categories<br />

according to their current level of qualification. Exploration level<br />

biomarkers are research and development tools with in vitro and/<br />

or preclinical evidence but without consistent information linking<br />

the biomarker to clinical outcomes in humans. Demonstration<br />

level biomarkers are associated with clinical outcomes in humans<br />

but have not been reproducibly demonstrated in clinical studies.<br />

Characterization level biomarkers are reproducibly linked to clinical<br />

outcomes in more than one prospective clinical study in humans.<br />

Surrogacy level biomarkers can substitute for a clinical endpoint,<br />

corresponding to ‘‘surrogate end point’’ as mentioned above, and<br />

require agreement with regulatory authorities as an FDA registrable<br />

endpoint. The table below lists a selection of commercially available<br />

osteoarthritis-related biomarkers and an approximation with<br />

regard to their level of qualification. However, that qualification is<br />

as always dependent on a specific context, limiting generalizability<br />

in the early development phase. There are at this time no qualified<br />

biomarkers that can be considered as surrogate clinical endpoints in<br />

OA. It should further be noted that the majority of currently studied<br />

biomarkers, including those exemplified below, lack a detailed<br />

verification of the tissue source and molecular structures that<br />

generate the signal in the assay, severely limiting our ability to draw<br />

conclusions from results generated by the assays. The development<br />

and validation of biomarkers with utility for osteoarthritis will require<br />

the combination of reproducible and specific biomarker assays with<br />

large prospective, controlled clinical trials. To demonstrate utility<br />

of e.g. efficacy biomarkers, these trials will need to show structural<br />

joint protection and/or clinically relevant efficacy.<br />

References:<br />

Bauer DC et al. Classification of osteoarthritis biomarkers: a<br />

proposed approach. Osteoarthritis <strong>Cartilage</strong> 2006;14:723-7.<br />

Felson DT, Lohmander LS. Whither Osteoarthritis Biomarkers?<br />

Osteoarthritis <strong>Cartilage</strong> 2009;17:419-22.<br />

Kraus VB, Burnett B, Coindreau J, Cottrell S, Eyre D, Gendreau M,<br />

Gardiner J, Garnero P, Hardin J, Henrotin Y, Heinegard D, Ko A,<br />

Lohmander LS, Matthews G, Menetski J, Moskowitz R, Persiani S,<br />

Poole R, Rousseau JC, Todman M. Application of Biomarkers in the<br />

Development of Drugs Intended for the Treatment of Osteoarthritis.<br />

OARSI FDA Osteoarthritis Biomarkers Working Group: FDA initiative<br />

for the coordination of a critical appraisal related to the design of<br />

clinical development programs for drugs, biological products &<br />

medical devices for the treatment/prevention of OA. Osteoarthritis<br />

<strong>Cartilage</strong> 2011;19:515-42.<br />

van Spil W, DeGroot J, Lems W, Oostveen J, Lafeber F. Serum and<br />

urinary biochemical markers for knee and hip osteoarthritis:<br />

a systematic review applying the consensus BIPED criteria.<br />

Osteoarthritis <strong>Cartilage</strong> 2010;18:605-12.<br />

Wagner J A, Williams S A, Webster C J. Biomarkers and surrogate end<br />

points for fit-for-purpose development and regulatory evaluation of<br />

new drugs. Clin Pharmacol Ther 2007;81:104-7.<br />

19.3.2<br />

Emerging molecular biomarker technologies and the way forward<br />

A.R. Poole<br />

Quebecr/<strong>Canada</strong><br />

Introduction: Molecular or biochemical biomarkers of cartilage<br />

metabolism offer the potential to detect and monitor cartilage damage<br />

in arthritis, the progression of this damage and the effectiveness of<br />

therapy designed to arrest cartilage damage and/or stimulate its<br />

repair. In addition these biomarkers can be used to examine the<br />

metabolism of cartilage in healthy individuals that may predispose<br />

them to joint damage and who may exhibit different capacities for<br />

joint repair. In this presentation we will look at the turnover of type<br />

II collagen, the dominant component of the extracellular matrix of<br />

cartilage, without which cartilage cannot exist.<br />

Content: The most important requirements for the development of<br />

successful skeletal biomarker technology are the ability to accurately<br />

identify the source of the biomarker; the molecular event(s) that<br />

generates it; what the biomarker assay measures in terms of the<br />

molecular fragment and where best to measure this biomarker-in<br />

synovial fluid, serum or urine since different results can be obtained<br />

with each of these body fluids. In this presentation we look at the<br />

development of technology to detect the synthesis and degradation<br />

of type II collagen in cartilage. The focus on cartilage degradation is<br />

on the cleavage of type II collagen by collagenases which is a key<br />

event in health and disease and is increased in articular cartilage in<br />

osteoarthritis (OA; Dejica et al, 2008) and rheumatoid arthritis (RA).<br />

The first immunoassay that was developed (Col2-3/4Clong or C2C)<br />

produced a competitive ELISA inhibition assay that measured the<br />

primary cleavage neoepitope generated by collagenases (Poole et al,<br />

2004). Thereby, any collagen II fragments containing this neoepitope<br />

would be detected. To detect collagen II synthesis a competitive<br />

ELISA immunoassay (CPII) was also developed, in this case to detect<br />

the c-propeptide of collagen II that is removed extracellularly as<br />

newly synthesized collagen is secreted (Nelson et al, 1998). These<br />

assays, used in combination and singly, have proved of value in<br />

ascertaining the amount of collagen II cleavage and synthesis in<br />

normal individuals and in patients with OA and rheumatoid arthritis<br />

(RA). Much of what is measured appears to relate to the cleavage<br />

of newly synthesized type II collagen in the pericellular domain<br />

around chondrocytes.These assays used in combination can detect<br />

differences between individuals with early pre- and radiographic<br />

knee osteoarthritis (OA) and those without knee OA (Cibere et al,<br />

2010).They reveal differences between those with knee OA who<br />

exhibit progression and those who do not (Cahue et al, 2007). They<br />

indicate early responses to disease-modifying therapy in patients<br />

with RA (Mullan et al, 2007). Surprisingly, in healthy active military<br />

trainees exposed to identical training programs these assays also<br />

revealed differences in those who subsequently succumbed to<br />

cruciate ligament rupture and those who did not (Svoboda et al,<br />

2012). Clearly differences in cartilage metabolism reflected by the<br />

differences in these cartilage biomarkers revealed differences in<br />

cartilage metabolism that put people either at risk for joint injury<br />

or not at risk.The reasons for this are currently under investigation.<br />

But what it signifies is that cartilage metabolism can vary between<br />

individuals of the same age and these differences may also identify<br />

people at risk for OA and whose disease is progressing. Such<br />

detectable differences in cartilage metabolism may also identify<br />

those who may respond differently to therapies designed to<br />

stimulate cartilage repair. All these studies focussed on cartilage<br />

metabolism measured systemically in serum peripheral blood. What<br />

we then discovered was that the cleavage assay, that also worked<br />

in urine, produced different results to what were seen in serum. The<br />

increase in cleavage of type II collagen seen in OA cartilage and in<br />

urine was not detectable in serum in patients with OA. It was then<br />

discovered that urine contained a dominant 45mer peptide of type<br />

II collagen that contained the C2C cleavage neoepitope and which<br />

was increased in OA (Nemerovskiy et al, 2007).This appeared to<br />

be reflect more the pathology of cartilage collagen cleavage rather<br />

than its turnover in health and disease .The cleavage 45mer peptide<br />

seemed to be masked in serum by the cleavage products generated<br />

by physiology.By developing a new sandwich assay (C2C HUSA)<br />

that is designed to detect this 45mer fragment in type II collagen it<br />

is now possible to use this new urine assay to better discriminate<br />

between cartilage turnover in those without knee OA and those<br />

with pre-radiographic and more advanced damage to articular<br />

cartilage in radiographic OA (Ha et al, 2012). These advances point<br />

to the great importance of looking at specific molecular events in<br />

biomarker development in detail. Otherwise a biomarker assay can<br />

be developed that measure something other than what you think<br />

it measures, as has been the case on some occasions. As we have<br />

seen, skeletal biomarkers have the potential to detect and monitor<br />

cartilage synthesis and degradation. A balance between synthesis<br />

and degradation is viewed as essential in maintaining healthy<br />

cartilage.Changes in this balance can be measured using currently<br />

available biomarker assays. In studies of cartilage repair one<br />

would expect that synthesis would be favoured over degradation,<br />

determined from assay ratios.From what has been discussed above<br />

it would be important to use these assays in studies of cartilage<br />

repair using both serum for synthesis and degradation as well as<br />

urine for degradation.


References:<br />

Cahue, S et al. Osteoarthritis <strong>Cartilage</strong> 15:819-23,2007; Cibere, J et<br />

al. Arthritis Rheum 60: 1372-80, 2009; Dejica, VM et al. Am J Pathol<br />

173: 161-69, 2008; Ha,N et al. In preparation;<br />

Mullan, RH et al. Arthritis Rheum 56: 2919-28, 2007; Nelson,F et al. J<br />

Clin Invest 102: 2115-25, 1998; Nemirovskiy, OV et al. Anal Biochem<br />

361: 93-101, 2007; Poole AR et al. J Immunological Meth 294: 145-<br />

53,2004.<br />

Acknowledgments:<br />

These studies were funded by Canadian Institutes of Health;<br />

NIAMS,National Institutes of Health; Canadian Arthritis Network;<br />

IBEX Technologies Inc.<br />

19.3.3<br />

Genetic influence on cartilage repair<br />

L.J. Sandell<br />

St. Louis/United States of America<br />

Introduction: Emerging evidence suggests that genetic components<br />

contribute significantly to cartilage degeneration in osteoarthritis<br />

pathophysiology but little evidence is available on genetics of<br />

cartilage regeneration. Therefore, we investigated cartilage<br />

regeneration in genetic murine models using common inbred strains<br />

and a set of recombinant inbred lines generated from LG/J (healer<br />

of ear-wounds) and SM/J (non-healer) inbred strains. Using the two<br />

healing extreme recombinant inbred lines, we tested the relationship<br />

between the ability to regenerate cartilage and the susceptibility to<br />

osteoarthritis (OA).<br />

Content: To investigate cartilage regeneration, we created an acute<br />

full-thickness cartilage injury through microsurgery in the trochlear<br />

groove of 265 mice by the method of Fitzgerald and colleagues (1).<br />

Knee joints were sagittally sectioned and stained with toluidine<br />

blue to evaluate regeneration. The ear-wound phenotype was also<br />

established. For ear-wound, a bilateral 2-mm through-and-through<br />

puncture was made and healing outcomes measured after 30-days.<br />

Broad-sense heritability and genetic correlations were calculated<br />

for both phenotypes. Time-course studies from recombinant inbred<br />

lines show no significant regeneration until 16-weeks post-surgery;<br />

at that time, the strains can be segregated into three categories:<br />

good, intermediate and poor healers. Broad-sense heritability<br />

showed that both cartilage regeneration and ear-wound closure are<br />

significantly heritable traits. The genetic correlations between the two<br />

healing phenotypes were found to be extremely high. We concluded<br />

that articular cartilage regeneration is heritable, the phenotypic<br />

differences between the lines are due to genetic differences and a<br />

strong genetic correlation between the two phenotypes (cartilage<br />

regeneration and ear-wound healing) exists indicating that they<br />

plausibly share a common genetic basis (2). We are currently studying<br />

gene expression differences in cartilage and bone in several of the<br />

recombinant inbred strains of mice already used for phenotyping by<br />

employing novel technology of QuantiGene Plex assay. This beadbased<br />

assay utilizes RNA from tissue lysates prepared from formalinfixed<br />

paraffin embedded sections. We are quantifying genes related<br />

to tissue healing, osteoarthritis, DNA repair, cell cycle control and<br />

other relevant pathways to determine differences among strains. We<br />

have also examined the susceptibility to osteoarthritis of our genetic<br />

mouse lines. We compared strain-dependent development of posttraumatic<br />

osteoarthritis and its association with tissue regeneration<br />

in two recombinant inbred lines LGXSM-6 and LGXSM-33 generated<br />

from LG/J and SM/J intercross. Our findings above indicated that<br />

LGXSM-6 can regenerate both articular cartilage and ear-hole punch<br />

while LGXSM-33 cannot. In this study, osteoarthritis was induced<br />

in over 70 mice through the transection of the medial meniscotibial<br />

ligament and cartilage and bone changes were evaluated by the<br />

method of Glasson (3). <strong>Cartilage</strong> damage showed that LGXSM-33<br />

developed a significantly higher grade of osteoarthritis than LGXSM-6.<br />

Bone analysis showed that LGXSM-33 had substantial subchondral<br />

bone and trabecular bone thickening post-surgery, while LGXSM-6<br />

showed bone loss over time. We also confirmed that LGXSM-6 can<br />

heal ear tissues significantly better than LGXSM-33. Thus, we found<br />

that osteoarthritis is negatively correlated with the degree of tissue<br />

regeneration (4).<br />

Extended Abstracts 165<br />

References:<br />

1. Fitzgerald J, Rich C, Burkhardt D, Allen J, Herzka AS, Little CB.<br />

Evidence for articular cartilage regeneration in MRL/MpJ mice.<br />

Osteoarthritis <strong>Cartilage</strong>. 2008;16(11):1319-26.<br />

2. Rai MF, Hashimoto S, Johnson EE, Janiszak K, Fitzgerald J, Heber-<br />

Katz E, Cheverud JM, Sandell LJ. Heritability of Articular <strong>Cartilage</strong><br />

Regeneration and its Association with Ear-Wound Healing. Arthritis<br />

Rheumatism In Press. doi: 10.1002/art.34396. PMID: 22275233.<br />

2012<br />

3. Glasson SS. In vivo osteoarthritis target validation utilizing<br />

genetically-modified mice. Curr Drug Targets 2007;8:367-76.<br />

4. Hashimoto S, Rai MF, Janiszak K, Cheverud JM, Sandell LJ.<br />

<strong>Cartilage</strong> and Bone Changes during Development of Post-Traumatic<br />

Osteoarthritis in Selected LGXSM Recombinant Inbred Mice.<br />

Osteoarthritis <strong>Cartilage</strong> In Press. doi:10.1016/j.joca.2012.01.022.<br />

2012<br />

Acknowledgments:<br />

The project was supported by an ARRA Grand Opportunity Grant,<br />

Award Number RC2 AR058978; the micro-CT and histological<br />

analysis were supported by the Musculoskeletal Research Center,<br />

Grant Award Number P30 AR057235, from the National Institute of<br />

Arthritis, Musculoskeletal and Skin Diseases.<br />

21.1.1<br />

Role of lubrication and joint homeostasis: <strong>Cartilage</strong> Boundary<br />

Lubricating Ability of Full-Length Recombinant Human PRG4 -<br />

Alone and In Combination with Hyaluronan<br />

S. Abubacker 1 , N. Masala 1 , S. Morrison 1 , G.D. Jay 2 , T.A. Schmidt 1<br />

1 Calgary/<strong>Canada</strong>, 2 Providence/United States of America<br />

Introduction: The proteoglycan 4 (PRG4) gene [1] encodes for mucinlike<br />

O-linked glycosylated proteins with several names, including<br />

lubricin [2] and superficial zone protein [3]. PRG4 proteins, herein<br />

referred to as PRG4, are synthesized and secreted by various cells<br />

within the joint, including synoviocytes [4] and articular chondrocytes<br />

in the superficial zone of cartilage [3]. As such, PRG4 is present in<br />

synovial fluid (SF) and at the surfaces of tissues within the joint,<br />

including articular cartilage [5]. Mutations in the PRG4 gene cause an<br />

autosomal recessive disorder in humans, camptodactylyarthropathycoxa<br />

vara-pericarditis (CACP), which results in juvenile-onset,<br />

noninflammatory, precocious joint failure [6]. PRG4-null mice show<br />

early signs of wear and higher friction than normal mouse joints -<br />

suggesting that friction is coupled with wear at the cartilage surface<br />

in vivo [7]. Therefore, normal expression of PRG4 is necessary for<br />

normal joint health. PRG4 has multiple biophysical properties [8,<br />

9], including the boundary lubrication of cartilage [10] (hence the<br />

classic name lubricin [11]), that contribute to the overall maintenance<br />

and integrity of the joint. Boundary lubrication is an operative and<br />

essential mechanism of articular cartilage lubrication in synovial<br />

joints, where surface-to-surface contact occurs and surface bound<br />

molecules provide lubrication. This mode of lubrication is thought<br />

to be important for the protection and maintenance of articular<br />

cartilage, since the apposing cartilage surfaces within the joint<br />

make contact over ~10% of the total area - where most of the<br />

friction may occur [12]. PRG4 has demonstrated in vitro boundary<br />

lubricating ability, in a dose-dependent manner, at a physiologically<br />

relevant cartilage-cartilage interface [10]. PRG4 has also been shown<br />

to act synergistically with hyaluronan (HA), through an unknown<br />

mechanism, to further reduce friction to levels near that of SF [10, 13].<br />

PRG4-deficient human SF lacks normal in vitro boundary lubricating<br />

ability [7], which suggests PRG4 is an important boundary lubricant<br />

that contributes to the low-friction articulation of cartilage surfaces<br />

in vivo. Moreover, the destructive joint phenotype associated with<br />

the lack of PRG4 in vivo [6, 7], clearly demonstrates the importance of<br />

both boundary lubrication and PRG4 for joint health. Recent studies<br />

suggest that a decrease of PRG4 concentration in SF following a knee<br />

injury, and the associated decreased boundary lubricating ability of<br />

SF, contributes to ensuing cartilage surface damage. In patients with<br />

acute knee injuries or progressive chronic inflammatory arthritis,<br />

cartilage damage has been associated with decreased boundary<br />

lubricating ability of SF [14]. This association was also observed in an<br />

animal model following anterior cruciate ligament (ACL) transection,<br />

as well as decreased levels of PRG4 in SF [15]. Decreased PRG4


166<br />

Extended Abstracts<br />

concentrations have been reported in SF of patients soon after<br />

an ACL injury [16], which is associated with an increased risk of<br />

subsequent progressive degeneration leading to osteoarthritis<br />

(OA) [17]. More recent studies collectively suggest that restoring<br />

normal boundary lubrication with PRG4 in situations where it is lost<br />

or diminished, could contribute to the maintenance of cartilage in<br />

vivo. Indeed, the diminished in vitro cartilage boundary lubricating<br />

ability of human OA SF deficient in PRG4, could be restored with<br />

PRG4 supplementation [18]. Furthermore, local administration of<br />

PRG4 [19], as well as both a truncated version [20] and more recently<br />

a full-length [21] recombinant human PRG4 (rh-PRG4), has been<br />

demonstrated to be therapeutically effective in preventing cartilage<br />

degeneration in a rat model of OA. Previous supporting studies<br />

demonstrated the ability of truncated and full-length rh-PRG4 to<br />

both bind to an articular surface [22] and reduce friction at a glasscartilage<br />

interface [23]. While PRG4’s molecular interactions with the<br />

articular surface, and with other lubricant molecules in SF, remains<br />

to be fully elucidated, local administration of rh-PRG4 appears to<br />

be a promising biotherapeutic intervention to potentially halt or<br />

slow the progression of OA. Recent advances in protein expression<br />

technology [24] has enabled the abundant expression of full-length<br />

rh-PRG4, which could be used for such therapeutic applications. The<br />

objectives of this study were to 1) biochemically characterize fulllength<br />

rh-PRG4, and 2) assess the cartilage boundary lubricating<br />

properties of rh-PRG4, both alone and in combination with HA.<br />

Content: METHODS: (rh-)PRG4 Preparation. rhPRG4. rh-PRG4 was<br />

enriched from media conditioned by Chinese hamster ovary cells,<br />

transfected with the rh-PRG4 gene, by concentration and filtration<br />

in a 100kDa MW cut-off filter. Total concentration was determined<br />

by bicinchoninic acid assay, and then determined for rh-PRG4<br />

based on purity of the preparation as assessed by SDS-PAGE<br />

protein stain (see below). PRG4. PRG4 was purified from culture<br />

medium conditioned by mature bovine cartilage explants from the<br />

superficial zone using anion exchange chromatography, essentially<br />

as described previously [10]. Biochemical Characterization. Samples<br />

were analyzed by SDS-PAGE followed by protein staining or western<br />

blotting, as described previously [13, 25]. Briefly, non-reduced (NR)<br />

and reduced (R) samples, with and without additional enzymatic<br />

treatment with neuraminidase and O-glycanase (Prozyme) to remove<br />

O-linked glycosylations, were electrophoresed on 3-8% Tris Acetate<br />

gels (Invitrogen). Gels were then stained for protein (SimplyBlue<br />

Stain, Invitrogen) or electroblotted to a 0.2 μm polyvinylidene<br />

fluoride membrane. Membranes were blocked, probed with a variety<br />

of anti-PRG4 Ab (pAb LPN and J108N [26], mAb 5C11 [26] and 9G3<br />

[19]) or lectins (peanut agglutinin: PNA, wheat germ agglutinin:<br />

WGA) and developed with an ECL chemiluminescent substrate<br />

and visualized with a Bio Imaging System. <strong>Cartilage</strong> Lubricating<br />

Ability. Lubricating ability of rh-PRG4 was tested with a cartilageon-cartilage<br />

friction test in the boundary lubrication regime using<br />

normal bovine osteochondral cores, as described previously [27].<br />

Briefly, after incubation in the test lubricant overnight, samples<br />

were compressed, allowed to stress relax, then subjected to relative<br />

rotation at an effective velocity of 0.3mm/s (to ensure a boundary<br />

mode of lubrication) with prespin durations, Tps, of 1200-1.2s. Static,<br />

μstatic,Neq, and kinetic, , friction coefficients<br />

were calculated [27]. Two sets of tests were performed to determine<br />

the boundary lubricating ability of the rh-PRG4, compared to native<br />

PRG4, at a physiological concentration of 450 μg/ml, both alone or<br />

in combination with 1.5MDa HA at a physiological concentration<br />

[28] of 3.33 mg/ml. Phosphate buffered saline (PBS) and bovine SF<br />

served as negative and positive controls, respectively. Test 1: PBS,<br />

rh-PRG4, PRG4, then SF. Test 2: PBS, rh-PRG4, rh-PRG4+HA, then<br />

SF. RESULTS: Biochemical Characterization. rh-PRG4 demonstrated<br />

similar immunoreactivity to PRG4. Western blotting indicated<br />

immunoreactive rh-PRG4 bands had a similar apparent molecular<br />

weight (MW) to those in PRG4. High MW species were observed in NR<br />

samples probed with pAb LPN and mAb 5C11 & 9G3, and lectins PNA<br />

and WGA. Upon reduction, a single high MW species was observed<br />

with pAb J108, mAb 5C11 and lectin PNA. Upon removal of the terminal<br />

sialic acid with neuraminidase in R samples, a reduction in MW of a<br />

J108N immunoreactive species was observed. A further reduction in<br />

MW was observed upon subsequent removal of the O-glycosylations<br />

via O-glycanase. <strong>Cartilage</strong> Lubricating Ability. rh-PRG4 effectively<br />

lowered friction at the cartilage-cartilage biointerface, both alone<br />

and in combination with HA. Lubricants and Tps modulated friction.<br />

μstatic,Neq increased with increasing Tps, whereas <br />

varied only slightly; both varied with test lubricant. Test 1: Both<br />

μstatic,Neq and were highest in PBS and lowest in<br />

SF, with values of PRG4 and rh-PRG4 being intermediate and similar<br />

to each other. Test 2: Both μstatic,Neq and were<br />

again highest in PBS and lowest in SF, and was again intermediate in<br />

rh-PRG4. With subsequent addition of HA, values of μstatic,Neq and<br />

in rhPRG4+HA were further lowered, approaching<br />

that of SF. DISCUSSION: This study demonstrates that full-length<br />

rh-PRG4 can be expressed with structural properties and cartilage<br />

boundary lubricating function similar to that of native PRG4. Western<br />

blot data suggests the rh-PRG4 has the appropriate higher order<br />

structure, with intermolecular disulfide bonds [26], as well as the<br />

expected O-linked glycosylations (α(2,3)NeuAc- β(1,3)Gal-GalNAc),<br />

although further detailed characterization [29] is required. rh-PRG4<br />

also demonstrated equivalent cartilage boundary lubricating ability<br />

to PRG4, both alone and in combination with HA, suggesting rh-<br />

PRG4 is able to both bind/interact with the native articular surface<br />

as well as HA to reduce friction during cartilage-cartilage articulation.<br />

Industrial scale-up of rh-PRG4 expression and purification, will<br />

facilitate future pre-clinical animal model work, and ultimately<br />

clinical trials, to further evaluate the effectiveness of rh-PRG4 as a<br />

biotherapeutic treatment to prevent or slow the progression of OA.<br />

Additionally, rh-PRG4 may be a useful biotherapeutic treatment at<br />

other biointerfaces where lubrication is needed, such as the ocular<br />

surface where PRG4 has been recently discovered and shown to<br />

provide lubricating function [30], either alone or in combination with<br />

existing HA-containing products.<br />

References:<br />

1. S. Ikegawa et al. Cytogenet Cell Genet 90, 291 (2000).<br />

2. D. A. Swann et al. J Biol Chem 256, 5921 (1981).<br />

3. B. L. Schumacher et al. Arch Biochem Biophys 311, 144 (1994).<br />

4. G. D. Jay et al. J Orthop Res 19, 677 (2001).<br />

5. G. D. Jay. Curr Opin Orthop 15, 355 (2004). 6. J. Marcelino et al. Nat<br />

Genet 23, 319 (1999).<br />

7. G. D. Jay et al. Arthritis Rheum 56, 3662 (2007).<br />

8. D. K. Rhee et al. J Clin Invest 115, 622 (2005).<br />

9. G. D. Jay et al. Proc Natl Acad Sci U S A 104, 6194 (2007).<br />

10. T. A. Schmidt et al. Arthritis Rheum 56, 882 (2007).<br />

11. D. A. Swann et al. J Biol Chem 247, 8069 (1972).<br />

12. K. C. Morrell et al. Proc Natl Acad Sci U S A 102, 14819 (2005).<br />

13. J. J. Kwiecinski et al. Osteoarthritis <strong>Cartilage</strong> 19, 1356 (2011).<br />

14. K. A. Elsaid et al. Arthritis Rheum 52, 1746 (2005).<br />

15. E. Teeple et al. J Orthop Res 26, 231 (2008).<br />

16. K. A. Elsaid et al. Arthritis Rheum 58, 1707 (2008).<br />

17. A. C. Gelber et al. Ann Intern Med 133, 321 (2000).<br />

18. T. E. Hill et al. Trans Orthop Res Soc 57, 34 (2011).<br />

19. E. Teeple et al. Am J Sports Med 39, 164 (2011).<br />

20. C. R. Flannery et al. Arthritis Rheum 60, 840 (2009).<br />

21. G. D. Jay et al. Arthritis Rheum 62, 2382 (2010).<br />

22. A. R. Jones et al. J Orthop Res 25, 283 (2007).<br />

23. J. P. Gleghorn et al. J Orthop Res 27, 771 (2009).<br />

24. P. A. Girod et al. Nature methods 4, 747 (2007).<br />

25. M. C. Alvarez et al. Trans Orthop Res Soc 56, 850 (2010).<br />

26. T. A. Schmidt et al. Biochim Biophys Acta 1790, 375 (2009).<br />

27. T. A. Schmidt et al. Osteoarthritis <strong>Cartilage</strong> 15, 35 (2007).<br />

28. J. R. Watterson et al. J Am Acad Orthop Surg 8, 277 (2000).<br />

29. R. P. Estrella et al. Biochem J 429, 359 (2010). 30. S. Morrison et<br />

al. Eye Contact Lens 38, 27 (2011).<br />

Acknowledgments:<br />

Alberta Innovates <strong>–</strong> Health Solutions, Canadian Arthritis Network,<br />

Lubris, Natural Sciences and Engineering Research Council of<br />

<strong>Canada</strong>.


21.1.3<br />

New frontiers in joint lubrication therapy<br />

C.R. Flannery<br />

Cambridge/United States of America<br />

Introduction: Recent advances in our understanding of the<br />

(mechano)biological attributes of key molecular regulators<br />

of cartilage lubrication have led to escalating progress in the<br />

development of therapeutic strategies designed to enhance joint<br />

function in pathological and repair environments. The results from a<br />

number of encouraging preclinical studies, and discussion of some<br />

translational elements applicable to aiding risk mitigation on the<br />

path to the clinic, are presented herein.<br />

Content: Articular joints are highly dependent on effective lubrication<br />

mechanisms to prevent the wear and degeneration of articular<br />

cartilage. Natural lubricants in synovial fluid include hyaluronan<br />

(HA), a viscous, high molecular weight glycosaminoglycan, and<br />

lubricin (also referred to as Proteoglycan 4/PRG4 or Superficial Zone<br />

Protein/SZP), a mucinous glycoprotein which confers specialized<br />

protection at cartilage surfaces (1). In addition to its rheological<br />

properties, recent data indicates that HA, like lubricin, appears to<br />

play an important role in boundary-mode lubrication, helping to<br />

guard cartilage surfaces from wear under conditions of high contact<br />

load and slow sliding speeds (2, 3). The biomolecular composition<br />

of cartilage itself is also critical for reducing friction and shear at<br />

the tissue surface. Thus, high concentrations of water-adsorbing<br />

proteoglycan (aggrecan) molecules entrapped within the cartilage<br />

collagen meshwork allow for interstitial pressurization and fluid<br />

exudation during joint loading cycles (4), thereby facilitating<br />

the generation of a surface hydration layer which contributes to<br />

joint lubrication. Several detrimental processes can contribute<br />

to lubrication deficiencies during joint pathology. For example,<br />

following traumatic injury, a significant decrease in the concentration<br />

of HA in the synovial fluid can occur, as well as alterations in<br />

lubricin expression levels and cartilage surface properties (5-8). In<br />

addition, proteolytic fragmentation of aggrecan by aggrecanases<br />

(ADAMTS proteases) is acutely elevated post-injury, resulting in<br />

significant proteoglycan loss from the articular cartilage (9). Various<br />

therapeutic strategies can therefore be pursued to help restore joint<br />

lubrication and diminish disease progression. Intraarticular (IA)<br />

viscosupplementation with HA has been used in the clinic now for<br />

several decades, and improvements in patient pain and function<br />

outcome measures are well documented (10). More recently, IA<br />

treatment with lubricin has been tested in preclinical animal models,<br />

and found to be effective in providing both chondroprotection<br />

(prevention of cartilage structural damage) and evidence of pain<br />

relief (11, 12). Likewise, chondroprotective effects are observed for<br />

aggrecanase (ADAMTS5) knockout mice (13), and oral administration<br />

of an aggrecanase inhibitor is efficacious in preventing cartilage<br />

aggrecan catabolism in a rat model of post-traumatic osteoarthritis<br />

(14). While attractive for their mode of local delivery, intraarticular<br />

treatment procedures are not without challenges. The clearance<br />

rate of macromolecules from synovial fluid is quite rapid, indicating<br />

that strategies to improve joint residence time could substantially<br />

improve therapeutic effects. Interestingly, lubricin, which possesses<br />

a targeted affinity for cartilage surface binding (15), appears to<br />

display a tri-phasic distribution profile following IA administration.<br />

While there is a rapid early decline in the levels of radiolabelled<br />

recombinant lubricin (LUB:1) from rat knees, detectable counts<br />

remain in the joint for at least 28 days, and can be observed at both<br />

intact and damaged cartilage surfaces by micro-autoradiography (16).<br />

Additional and complimentary strategies may therefore be envisioned<br />

in the further development of joint lubrication therapies, including<br />

treatments designed to enhance lubricant biosynthesis. In the case<br />

of lubricin, biochemical stimuli such as TGF-beta and related family<br />

members can increase its expression (17, 18). Microencapsulation of<br />

therapeutic agents/lubricants into polymeric delivery particles may<br />

thus be employable to enable extended release and delivery within<br />

the joint. Interestingly, it is also worth noting that the levels of both<br />

lubricin and hyaluronan are enhanced by biomechanical forces such<br />

as surface motion, which may thereby contribute to the beneficial<br />

effects of continuous passive motion applied during post-operative<br />

patient rehabilitation (19, 20). Clearly, efficient lubrication will also<br />

be important for the success of cartilage repair and cartilage tissue<br />

engineering procedures. In this regard, the presence of lubricin has<br />

been detected in biopsies of repair tissue from patients treated<br />

with autologous chondrocyte implantation (21). Of the 43 samples<br />

analyzed, most were immunopositive for lubricin, with a high<br />

percentage demonstrating its presence in the surface layer. Protocols<br />

for enhancing lubricin localization at the articular surface, such as<br />

described above, may therefore serve to further improve repair<br />

tissue performance and hence clinical outcomes. In another recent<br />

Extended Abstracts 167<br />

study, chondrocyte differentiation under hypoxic conditions was<br />

found to increase lubricin expression by superficial (but not middle/<br />

deep) zone cells, highlighting the rationale for composing stratified/<br />

zonally-organized cartilaginous constructs (22). Furthermore, the<br />

ability to demonstrate a functional/biophysical correlation (i.e.<br />

improved lubrication) with enhanced lubricant expression, as has<br />

been shown for growth factor-stimulated cartilage explants and<br />

chondrocyte-seeded scaffolds subjected to sliding motion (23, 24),<br />

serves to provide an additional measure of confidence for translation<br />

into the clinic.<br />

References:<br />

1. Hui AY, McCarty WJ, Masuda K, Firestein GS, Sah RL. A systems<br />

biology approach to synovial joint lubrication in health, injury, and<br />

disease. WIREs Syst Biol Med. 2012;4:15-37.<br />

2. Schmidt TA, Gastelum NS, Nguyen QT, Schumacher BL, Sah RL.<br />

Boundary lubrication of articular cartilage: role of synovial fluid<br />

constituents. Arthritis Rheum. 2007;56:882-91.<br />

3. Greene GW, Banquy X, Lee DW, Lowrey DD, Yu J, Israelachvili JN.<br />

Adaptive mechanically controlled lubrication mechanism found in<br />

articular joints. Proc Natl Acad Sci USA. 2011;108:5255-9.<br />

4. Ateshian GA. The role of interstitial fluid pressurization in articular<br />

cartilage lubrication. J Biomech. 2009;42:1163-76.<br />

5. Elsaid KA, Fleming BC, Oksendahl HL, Machan JT, Fadale PD,<br />

Hulstyn MJ, Shalvoy R, Jay GD. Decreased lubricin concentrations and<br />

markers of joint inflammation in the synovial fluid of patients with<br />

anterior cruciate ligament injury. Arthritis Rheum. 2008;58:1707-15.<br />

6. Jones AR, Chen S, Chai DH, Stevens AL, Gleghorn JP, Bonassar<br />

LJ, Grodzinsky AJ, Flannery CR. Modulation of lubricin biosynthesis<br />

and tissue surface properties following cartilage mechanical injury.<br />

Arthritis Rheum. 2009;60:133-42.<br />

7. Wong BL, Chris Kim SH, Antonacci JM, McIlwraith CW, Sah RL.<br />

<strong>Cartilage</strong> shear dynamics during tibio-femoral articulation: effect<br />

of acute joint injury and tribosupplementation on synovial fluid<br />

lubrication. Osteoarthritis <strong>Cartilage</strong>. 2010;18:464-71.<br />

8. Catterall JB, Stabler TV, Flannery CR, Kraus VB. Changes in serum<br />

and synovial fluid biomarkers after acute injury (NCT00332254).<br />

Arthritis Res Ther. 2010;12:R229.<br />

9. Larsson S, Lohmander LS, Struglics A. Synovial fluid level of<br />

aggrecan ARGS fragments is a more sensitive marker of joint disease<br />

than glycosaminoglycan or aggrecan levels: a cross-sectional study.<br />

Arthritis Res Ther. 2009;11:R92.<br />

10. Iannitti T, Lodi D, Palmieri B. Intra-articular injections for the<br />

treatment of osteoarthritis. Focus on the clinical use of hyaluronic<br />

acid. Drugs R D. 2011;11:13-27.<br />

11. Flannery CR, Zollner R, Corcoran C, Jones AR, Root A, Rivera-<br />

Bermudez MA, Blanchet T, Gleghorn JP, Bonassar LJ, Bendele AM,<br />

Morris EA, Glasson SS. Prevention of cartilage degeneration in a rat<br />

model of osteoarthritis by intraarticular treatment with recombinant<br />

lubricin. Arthritis Rheum. 2009;60:840-7.<br />

12. Jay GD, Elsaid KA, Kelly KA, Anderson SC, Zhang L, Teeple E,<br />

Waller K, Fleming BC. Prevention of cartilage degeneration and gait<br />

asymmetry by lubricin tribosupplementation in the rat following ACL<br />

transaction. Arthritis Rheum. 2011;Published online Nov 29.<br />

13. Glasson SS, Askew R, Sheppard B, Carito B, Blanchet T, Ma HL,<br />

Flannery CR, Peluso D, Kanki K, Yang Z, Majumdar MK, Morris EA.<br />

Deletion of active ADAMTS5 prevents cartilage degradation in a<br />

murine model of osteoarthritis. Nature. 2005;434:644-8.<br />

14. Chockalingam PS, Sun W, Rivera-Bermudez MA, Zeng W, Dufield<br />

DR, Larsson S, Lohmander LS, Flannery CR, Glasson SS, Georgiadis<br />

KE, Morris EA. Elevated aggrecanase activity in a rat model of<br />

joint injury is attenuated by an aggrecanase specific inhibitor.<br />

Osteoarthritis <strong>Cartilage</strong>. 2011;19:315-23. 15. Jones AR, Gleghorn JP,<br />

Hughes CE, Fitz LJ, Zollner R, Wainwright SD, Caterson B, Morris EA,<br />

Bonassar LJ, Flannery CR. Binding and localization of recombinant<br />

lubricin to articular cartilage surfaces. J Orthop Res. 2007;25:283-<br />

92.<br />

16. Vugmeyster Y, Wang Q, Xu X, Harrold J, Daugusta D, Li J,


168<br />

Extended Abstracts<br />

Zollner R, Flannery CR, Rivera-Bermudez MA. Disposition of human<br />

recombinant lubricin in naïve rats and in a rat model of post-traumatic<br />

arthritis after intra-articular or intravenous administration. AAPS J.<br />

2012:Published online Jan 7.<br />

17. Jones AR, Flannery CR. Bioregulation of lubricin expression by<br />

growth factors and cytokines. Eur Cell Mater. 2007;13:40-5.<br />

18. Niikura T, Reddi AH. Differential regulation of lubricin/superficial<br />

zone protein by transforming growth factor beta/bone morphogenetic<br />

protein family superfamily members in articular chondrocytes and<br />

synoviocytes. Arthritis Rheum. 2007;56:2312-21.<br />

19. Grad S, Lee CR, Gorna K, Gogolewski S, Wimmer MA, Alini M.<br />

Surface motion upregulates superficial zone protein and hyaluronan<br />

production in chondrocyte-seeded three-dimensional scaffolds.<br />

Tissue Eng. 2005;11:249-56.<br />

20. Nugent-Derfus GE, Takara T, O‘neill JK, Cahill SB, Görtz S, Pong T,<br />

Inoue H, Aneloski NM, Wang WW, Vega KI, Klein TJ, Hsieh-Bonassera<br />

ND, Bae WC, Burke JD, Bugbee WD, Sah RL. Continuous passive<br />

motion applied to whole joints stimulates chondrocyte biosynthesis<br />

of PRG4. Osteoarthritis <strong>Cartilage</strong>. 2007;15:566-74.<br />

21. Roberts S, Menage J, Flannery CR, Richardson JB. Lubricin: its<br />

presence in repair cartilage following treatment with autologous<br />

chondrocyte implantation. <strong>Cartilage</strong>. 2010;1:298-305.<br />

22. Schrobback K, Malda J, Crawford RW, Upton Z, Leavesley DI, Klein<br />

TJ. Effects of oxygen on zonal marker expression in human articular<br />

chondrocytes. Tissue Eng Part A. 2012;Published online Jan 4.<br />

23. Gleghorn JP, Jones AR, Flannery CR, Bonassar LJ. Alteration of<br />

articular cartilage frictional properties by transforming growth<br />

factor beta, interleukin-1beta, and oncostatin M. Arthritis Rheum.<br />

2009;60:440-9.<br />

24. Grad S, Loparic M, Peter R, Stolz M, Aebi U, Alini M. Sliding<br />

motion modulates stiffness and friction coefficient at the surface of<br />

tissue engineered cartilage. Osteoarthritis <strong>Cartilage</strong>. 2012;Published<br />

online Jan 10.<br />

Acknowledgments:<br />

The valued contributions and insights provided by colleagues and<br />

collaborators are gratefully acknowledged.<br />

21.2.1<br />

Rehabilitation after cartilage repair<br />

M. Steinwachs<br />

Freiburg/Germany<br />

Introduction: The native joint cartilage is not able to structurally<br />

repair traumatic or repetitive overload injuries to the articular<br />

cartilage.A higher-grade cartilage injury leads to a shift in the balance<br />

towards cartilage degradation. This results in greater deformation<br />

of the cartilage under the enormous impact forces of sports and<br />

thereby mechanical structural damage of the ECM.<br />

Content: The treatment of specific cartilage defects can be<br />

distinguished between tissue transplants and cell-based regenerative<br />

techniques (7). The tissue transplantation techniques include the<br />

autologous osteochondral transplantation system (OATS) or tissue<br />

engineered cartilage tissue. Studies have identified three stages in<br />

cartilage regeneration (9):<br />

Phase I (Proliferative Phase 0-12 weeks): After cell attachment a<br />

non specific, soft repair tissue is formed. Phase II (Transition Phase<br />

3-6 month): A specific integration into the local environment and<br />

the improvement of the structural quality of the repair cartilage<br />

can be observed. Up to phase II, there generation steps are similar<br />

in both, the ACI as well as in the bone marrow based technique.<br />

Phase III (Remodelling and Maturation Phase 6-24month): The final<br />

adaptation of the cartilage is made to the biomechanical needs of<br />

the various joint compartments (1). This process includes a period of<br />

up to 2 years, as seen in MRI studies. The ACI proved to be superior<br />

in the quality of the regenerate tissue achieved compared to marrow<br />

stimulation techniques resulting in a longer maturation process up<br />

to 2 years (2,4,5, 6,7<br />

The primary objective for rehabilitation following cartilage repair<br />

procedures is the provision of an optimal environment and protection<br />

for overloading of the chondral repair tissue. Return to sport after<br />

Ccrtilage repair range from 8-24 month(1,7). The range in timescale<br />

for return to the preeinjury level of activities reflects the histological<br />

differences in the repair tissue across the cartilage repair procedures<br />

and the speed of the final maturation process after surgery (7).<br />

The precise timelines for the rehabilitation after cartilage repair will<br />

be dependent on a number of patient specific and lesion specific<br />

factors. These factors have been previously identified in differnt<br />

publications like leasion size, defect location etc.<br />

The tradditional rehabilitation program include in the phase I<br />

nonweight bearing<br />

for the first two weeks followed by partial weight bearing (5-10<br />

KG) for the next four weeks. After the sixth postoperativ week the<br />

progression to full weight bearing is necessary (2,5,6,9) There is still<br />

a controversial discussion in literature about the use of continuous<br />

passive motion (CPM). Although the results of partial retrospective,<br />

partial prospective,sometimes randomized or double blinded studies<br />

are incontradiction, there can only be found a trend to better results.<br />

New clinical studies for evidence based guidelines in the handling of<br />

continous passive motion after knee surgery are necessary (8).<br />

Due to the very complex biological and cell biological processes<br />

in general and specific tissue healing, to secure good results, a<br />

standardised rehabilitation depending on the methods, the defect<br />

location and the associated injuries is important.<br />

References:<br />

1) Della Villa S, Kon E, Filardo G, Ricci M, Vincentelli F, Delcogliano<br />

M, et al. Does intensive rehabilitation permit early return to sport<br />

without compromising the clinical outcome after arthroscopic<br />

autologous chondrocyte implantation in highly competitive athletes?<br />

Am J Sports Med. 2010;38(1):68-77.<br />

2) Hambly K, Bobic V, Wondrasch B, Van Assche D, Marlovits<br />

S. Autologous chondrocyte implantation postoperative care<br />

and rehabilitation: science and practice. Am J Sports Med.<br />

2006;34(6):1020-1038<br />

3) Hurst JM, Steadman JR, O’Brien L, Rodkey WG, Briggs KK.<br />

Rehabilitation following microfracture for chondral injury in the<br />

knee. Clin Sports Med. 2010;29(2):257-265, viii.<br />

4) Kreuz PC, Steinwachs M, Erggelet C, Lahm A, Krause S, Ossendorf C,<br />

et al. Importance of sports in cartilage regeneration after autologous<br />

chondrocyte implantation: a prospective study with a 3-year followup.<br />

Am J Sports Med. 2007;35(8):1261-1268.<br />

5) Van Assche D, Caspel DV, Staes F, Saris DB, Bellemans J, Vanlauwe<br />

J, et al. Implementing one standardized rehabilitation protocol<br />

following autologous chondrocyte implantation or microfracture<br />

in the knee results in comparable physical therapy management.<br />

Physio Ther Prac. 2011;27(2):125-136.<br />

6) Steinwachs MR, Kreuz PC, Guhlke-Steinwachs U, Niemeyer P.<br />

Current treatment for cartilage damage in the patellofemoral joint<br />

Orthopade. 2008 Sep;37(9):841-7.<br />

7) Hambly K, Silvers H, Steinwachs M. Rehabilitation after articular<br />

cartilage repair of the knee in the football player. <strong>Cartilage</strong> 2012<br />

8) Kirschner P. CPM <strong>–</strong> continuous passive motion in the treatment of<br />

injured or operated<br />

knee-joints: a metaanaylis of current literature Unfallchirurg<br />

2004107:328<strong>–</strong>340<br />

9) Minas T, Chui R. Autologous Chondrocyte implantation. Am J Knee<br />

Surg 2000; 41-50


21.2.2<br />

Return to sports after articular cartilage repair<br />

K. Mithoefer<br />

Cambridge/United States of America<br />

Introduction: Articular cartilage injury in athletes is frequently<br />

observed and often associated with significant acute or chronic<br />

limitation of the ability to participate in athletic activities. For the<br />

symptomatic, injured athlete, the ability to return to competitive<br />

sports participation presents one of the most important measures<br />

of functional outcome and treatment success. Information on the<br />

ability to return to sport is therefore of critical relevance in this<br />

active population.<br />

Content: The ability to return to sport provides a categorical but<br />

nonspecific outcome measure after treatment of an athletic injury.<br />

Return to sports participation alone does not provide any discrete<br />

variables of the athletic function and performance. Due to the<br />

complexity of the process of returning to sports participation a<br />

detailed analysis of the return to athletic activity is recommended.<br />

Post-recovery sports activity should be categorized based on the<br />

competitive level of the sports activity and compared to the preinjury<br />

level of play. In addition, objective performance parameters<br />

should be recorded to assess athletic functional outcome. The<br />

presence of any residual symptoms during sports participation<br />

should be noted. Besides the ability to return to sport, continued<br />

sports participation over time and risk for re-injury present important<br />

measures of long-term functional outcome. Independent of physical<br />

and performance parameters, psychological and social aspects of<br />

the athlete may affect return to sport and need to be included in<br />

a thorough functional evaluation of the athlete’s return or failure<br />

to return to sport. Health care providers treating injured athletes<br />

are frequently required to make decisions regarding the timing of<br />

exercise progression, the resumption of functional activities and<br />

return to competitive play. The decision when the athlete can safely<br />

return to sport after recovering from a performance-limiting injury<br />

often presents a particular challenge to the athlete’s physician,<br />

physical therapist, and athletic trainer. Quickly and safely returning<br />

the athlete to sport can be achieved by adopting an outcomesbased<br />

approach to treatment progression, in which the athlete must<br />

reach specific benchmarks before advancing activity levels. This<br />

strategy allows for quantification of an athlete’s functional ability,<br />

comparison with pre-injury performance status and verification that<br />

an appropriate level of rehabilitation has been achieved. Based on<br />

the method of data collection, functional outcome measures utilized<br />

in the process of returning the athlete to sport can be divided into<br />

subjective (patient-reported) and objective (clinician-reported)<br />

measures. Subjective measures typically include questionnaires<br />

and patient-based outcome scores. Objective measures include<br />

clinical examination, radiological studies, and testing of the athlete’s<br />

function and performance. Subjective and objective outcome<br />

parameters provide complementary information and are best used<br />

in combination to provide the most comprehensive assessment of<br />

the athlete’s functional ability. The criteria for return to sport may<br />

vary depending on the injury location, type of sport, and athlete’s<br />

performance level. In summary, return to sports after injury in<br />

the athletic population presents a critical but complex issue that<br />

requires a systematic and individualized approach to assure the<br />

safe return of the athlete to competition at pre-injury performance<br />

level and successful continued participation without re-injury. The<br />

rate of return to sport after articular cartilage repair in the athletic<br />

population is comparable between currently available surgical<br />

repair techniques but differences exist in the ability to continue<br />

active sports participation over time.<br />

References:<br />

1. Della Villa S, Kon E, Filardo G, Ricci M, Vincentelli F, Delcogliano<br />

M, Marcacci M. Does intensive rehabilitation permit early return to<br />

sport without compromising the clinical outcome after arthroscopic<br />

autologous chondrocyte transplantation in highly competitive<br />

athletes? Am J Sports Med. 2010 Jan;38(1):68-77.<br />

2. Gudas R, Kelesinskas RJ, Kimtys V, Stankevicius E, Toliusis<br />

V, Benotavicius G, Smailys A: A prospective randomized clinical<br />

study of mosaic osteochondral autologous transplantation versus<br />

microfracture for the treatment of osteochondral defects in the knee<br />

joint in young athletes. Arthroscopy 2005; 21:1066-75.<br />

3. Irrgang JJ, Pezzullo, D. Rehabilitation following surgical procedures<br />

to address articular cartilage lesions in the knee. J Orthop Sports<br />

Phys Ther. 1998;28:232-240.<br />

Extended Abstracts 169<br />

4. Kreuz PC, Steinwachs M, Erggelet C, Lahm A, Krause S, Ossendorf C,<br />

et al. Importance of sports in cartilage regeneration after autologous<br />

chondrocyte implantation. Am J Sports Med 2007; 35: 1261-68.<br />

5. Maletius W, Messner K: The long-term prognosis for severe<br />

damage to the weightbearing cartilage in the knee: A 14-year clinical<br />

and radiographic follow-up in 28 young athletes. Acta Orthop Scand<br />

1996; 165-168.<br />

6. Mithoefer K, Gill TJ, Cole BJ, Williams RJ, Mandelbaum BR. Clinical<br />

outcome and return to competition after microfracture in the athlete’s<br />

knee: An evidenc-based syetematic review. <strong>Cartilage</strong> 2010; 1:113-20.<br />

7. Mithoefer K, Hambly K, Della Villa S, Silvers H, Mandelbaum<br />

BR. Return to sports participation after articular cartilage repair in<br />

the knee: scientific evidence. Am J Sports Med 2009; 37 Suppl 1:<br />

167S-76S.<br />

8. Mithöfer K, Minas T, Peterson L, Yeon H, Micheli LJ: Functional<br />

outcome of articular cartilage repair in adolescent athletes. Am J<br />

Sports Med 2005; 33:1147-1153.<br />

9. Mithöfer K, Peterson L, Mandelbaum BR, Minas T: Articular<br />

cartilage repair in soccer players with autologous chondrocyte<br />

transplantation: Functional outcome and return to competition. Am J<br />

Sports Med 2005; 33(11):1639-46.<br />

10. Mithoefer K, Williams RJ, Warren RF, Wickiewicz TL, Marx<br />

RG. High-impact athletics after knee articular cartilage repair: A<br />

prospective evaluation of the microfracture technique. Am J Sports<br />

Med. 2006;34:1413-8.<br />

11. Reinold MM, Wilk KE, Macrina LC, Dugas JR, Cain EL. Current<br />

concepts in the rehabilitation following articular cartilage repair<br />

procedures in the knee. J Orthop Sports Phys Ther. 2006;36:774-<br />

794.<br />

12. Roberston CM, Williams RJ, Warren RF, Rodeo SA, Wickiewicz TL.<br />

Return to sports after fresh osteochondral allograft transplantation<br />

in the knee. Abstract 8970 presented at the 2010 annual meeting<br />

of the American Orthopedic <strong>Society</strong> of Sports Medicine (AOSSM),<br />

Providence, RI, USA, July 17, 2010.<br />

13. Van Assche D, Van Caspel D, Vanlauwe J, Bellemans J, Saris<br />

DB, Luyten FP, Staes F. Physical activity levels after characterized<br />

chondrocyte implantation versus microfracture in the knee and the<br />

relationship to objective functional outcome with 2-year follow-up<br />

Am J Sports Med. 2009 Nov;37 Suppl 1:42S-49S.<br />

14. Wondrasch B, Zak L, Welsch GH, Marlovits S. Effect of accelerated<br />

weightbearing after matrix-associated autologous chondrocyte<br />

implantation on the femoral condyle on radiographic and clinical<br />

outcome after 2 years: A prospective, randomized controlled pilot<br />

study. Am J Sports Med. 2009;37:88S-96S.<br />

21.2.3<br />

Challenges in Standardizing Rehabilitation in <strong>Cartilage</strong> <strong>Repair</strong><br />

RCTs<br />

M.S. Shive<br />

Quebec/<strong>Canada</strong><br />

Introduction: Current orthopaedic efforts towards improving<br />

evidence-based treatment algorithms in cartilage repair should<br />

also recognize post-treatment rehabilitation as a critical covariate<br />

in the analysis and understanding of outcome success. However,<br />

the standardization and control of patient rehabilitation offers<br />

unique challenges, particularly in multicenter studies of large or<br />

varied geographic scale and even more so for those intended to<br />

meet regulatory requirements and conducted under Good Clinical<br />

Practice (GCP). Currently, there is little scientific evidence comparing<br />

rehabilitation protocols following different cartilage repair therapies.<br />

This, compounded with ‘custom’ programs developed by orthopaedic<br />

surgeons with little or no scientific basis, or programs established<br />

by therapists themselves, leaves those trying to design new studies<br />

with little scientific support. However, some clinical evidence has<br />

begun to surface focused on rehabilitation. Clinical, radiological<br />

and biomechanical outcomes following only MACI cartilage repair<br />

treatment were studied in patients randomized to 2 different<br />

rehabilitation approaches, with either conservative or accelerated<br />

weight bearing, bringing some new insight into weight bearing to


170<br />

Extended Abstracts<br />

those using MACI.[1] And perhaps the only published randomized<br />

clinical trial (RCT) which controlled and studied rehabilitation and<br />

subsequent outcomes was reported by Van Assche et al [2], who<br />

compared functional outcomes under standardized rehabilitation<br />

protocols for microfracture versus ACI. Further awareness has been<br />

raised by broader discussions as found recently in the <strong>International</strong><br />

<strong>Cartilage</strong> <strong>Repair</strong> Newsletter [3] dedicated to the topic, and<br />

highlighting the fact that rehabilitation is a true adjunct to cartilage<br />

repair. And to standardize and study rehabilitation in cartilage repair<br />

RCTs, multiple challenges must be overcome.<br />

Content: Specific Challenges to Overcome: 1) Rehabilitation<br />

Protocol. The design of new cartilage repair studies should carefully<br />

weigh the choice of rehabilitation protocols, their implementation<br />

and the type of data that will be collected. Study protocols should<br />

dictate the selection of the appropriate therapeutic modalities<br />

and post-operative timeframe, such as the period of restricted<br />

weightbearing, or the use (or non-use) of continuous passive<br />

motion (CPM). Furthermore, clinical trials which compare cartilage<br />

repair therapies that differ in their repair mechanisms- as with an<br />

osteochondral graft device compared to arthroscopic microfracture-<br />

might call for different protocols altogether. 2) Studied parameters<br />

and relationship to outcomes. Special consideration should be made<br />

as to the role that rehabilitation plays within a study protocol and<br />

its relationship to study endpoints. Choosing specific rehabilitation<br />

parameters and the extent of data collection can represent an<br />

excellent, albeit challenging, opportunity to understand how a<br />

rehabilitation protocol influences repair outcomes. Even obvious<br />

variables like patient compliance, the progression of range of<br />

motion and weightbearing and physical knee characteristics<br />

could be informative, as well as more complex data obtained from<br />

functional tests of gait or proprioception. The extent of the data and<br />

the method of collection should be established with the therapists<br />

prior to treatment. Use of electronic data capture facilitates this<br />

collection and permits real-time tracking of both therapists and<br />

the progression of patients. 3) Standardization of rehabilitation.<br />

The selection and training of study therapists is an essential step<br />

in successfully implementing a standardized rehabilitation program.<br />

<strong>International</strong> studies offer the challenge of multi-lingual protocols<br />

and datasets and may need to employ therapists with differing<br />

educational backgrounds but who have appropriate experience.<br />

Training of therapists should not be left to documentation as direct<br />

contact permits Q&A and explanation of subtleties, if any. Frequent<br />

communication regarding patients is helpful, especially with regards<br />

to study treatment, lesion size and location or other unique study<br />

components. Furthermore, the logistics and financial management,<br />

if needed, of such a relationship with the clinic or hospital providing<br />

the rehabilitation services, needs to be established well in advance<br />

of patient treatment. 4) Patient compliance. Patients should be well<br />

informed regarding the importance of protocol compliance (eg.<br />

attending sessions, weightbearing requirements) to both the health<br />

of their knee as well as the clinical study. Encouragement should<br />

not only come from the clinical site and the investigator, but also<br />

from the participating therapists. The objective of an unbiased<br />

study rehabilitation program should try to avoid the situation where<br />

compliance becomes a function of the efficacy of treatment, or of the<br />

willingness or ability to pay. 5) Regulatory unknowns. Regulatory<br />

agencies are expecting to see standardized rehabilitation data<br />

(duration, type, frequency of weight bearing, methods etc) conducted<br />

under GCPs in approval submissions. However, recent FDA Guidance<br />

for Industry [4] on cartilage repair clinical trials is not clear regarding<br />

the expected correlation of rehabilitation parameters to treatment<br />

outcomes and the extent of statistical analysis, if any, which must<br />

be carried out. There is, nonetheless a stated concern that the<br />

inability to standardize and track rehabilitation may introduce<br />

bias and as such, any potential influence on outcomes should be<br />

examined. Furthermore, supporting documentation and rationales<br />

are expected. This important issue needs to be clarified at an agency<br />

level- particularly if poor rehabilitation compliance or progression<br />

is viewed as a study protocol deviation with direct consequences<br />

on final endpoint statistical analyses. As the cartilage repair field<br />

advances with new therapies, more randomized controlled trials<br />

will be needed to improve evidence based treatment algorithms. It<br />

is critical that the appropriate energy and resources are allocated<br />

to implement well thought-out and standardized rehabilitation<br />

programs, not only to normalize cartilage repair outcome data, but<br />

also to focus much needed attention on the specific rehabilitation<br />

modalities currently used but for which there is little scientific<br />

evidence. Consequently, cartilage repair patients will benefit from<br />

rehabilitation protocols with valid scientific basis.<br />

References:<br />

[1] Ebert JR, Robertson WB, Lloyd DG, Zheng MH, Wood DJ, Ackland<br />

T. Traditional vs accelerated approaches to post-operative<br />

rehabilitation following matrix-induced autologous chondrocyte<br />

implantation (MACI): comparison of clinical, biomechanical and<br />

radiographic outcomes. Osteoarthritis & <strong>Cartilage</strong>. 16(10):1131-40,<br />

2008<br />

[2] Van Assche D, Staes F, Van Caspel D, Vanlauwe J, Bellemans J,<br />

Saris DB, Luyten FP. Autologous chondrocyte implantation versus<br />

microfracture for knee cartilage injury: a prospective randomized<br />

trial, with 2-year follow-up. Knee Surg Sports Traumatol Arthrosc;<br />

18(4):486-95, 2010<br />

[3] “Special Focus: Rehabilitation. Global Concepts for Successful<br />

Joint Restoration”. <strong>International</strong> <strong>Cartilage</strong> <strong>Repair</strong> <strong>Society</strong> Newsletter,<br />

Winter Issue 14, 2011<br />

[4] US FDA Guidance for Industry: Preparation of IDEs and INDs for<br />

Products Intended to <strong>Repair</strong> or Replace Knee <strong>Cartilage</strong>, December<br />

2011<br />

Acknowledgments:<br />

The author would like to acknowledge the BST-CarGel (Piramal<br />

Healthcare, formerly BioSyntech) clinical development team which<br />

included over 60 dedicated physiotherapists who participated in the<br />

BST-CarGel multicenter RCT<br />

22.1<br />

Contribution of other tissues to cartilage degeneration<br />

C.B. Little<br />

St Leonards/Australia<br />

Introduction: The maintenance of any tissue is a balance between<br />

anabolism and catabolism, both of which are inevitably occurring<br />

simultaneously as part of normal homeostasis. In musculoskeletal<br />

tissues such as cartilage and bone where the extracellular matrix<br />

makes up such a large proportion of the tissue volume and is key<br />

to the mechanical properties, this balance is particularly important.<br />

In diseases such as osteoarthritis (OA) despite evidence for<br />

increased biosynthesis of cartilage matrix components at least<br />

early in the disease, the catabolic processes predominate and<br />

matrix breakdown and cartilage loss occurs. It is clear that just as<br />

the capacity to synthesize cartilage matrix and (re)generate and<br />

repair cartilage resides with the resident cells (chondrocytes), so<br />

much of the catabolism of cartilage in disease occurs as the result<br />

of secretion matrix degrading enzymes by the same cells. Targeting<br />

chondrocyte anabolic and catabolic mechanisms has therefore been<br />

a major focus of OA and cartilage repair research.<br />

Content: Despite the central role of chondrocytes in maintenance<br />

and turnover of cartilage, it is becoming increasingly evident<br />

that diseases of the joint such as OA, do not represent disorders<br />

solely of cartilage, but rather they are maladies of the entire “joint<br />

organ”. In OA, pathological change is seen in synovium, joint<br />

capsule, subchondral bone (SCB), and where present intra-articular<br />

ligaments and menisci, as well as cartilage. There is the opportunity<br />

for extensive cross-talk between all of these joint tissues by both<br />

soluble mediators and biomechanical signaling pathways. As a<br />

result of this potential joint tissue interaction, different “scientific<br />

schools” have evolved, each with its favorite tissue(s) placed at the<br />

center of the joint pathology cascade and subsequently as the focus<br />

of anti-catabolic therapeutics. The cartilage-centric view of the joint,<br />

posits that the primary pathological change is in cartilage and this<br />

leads to secondary pathology in other joint tissues. Thus, cartilage<br />

damage can incite a cascade of events including altered loading on<br />

the subjacent subchondral bone inducing its remodeling, release<br />

of cartilage degradation products/fragments inciting inflammation<br />

in the synovium, the synovitis-associated effusion decreasing joint<br />

lubrication and biomechanical stability, and together with secretion<br />

of growth factors from synovial macrophages initiating osteophyte<br />

development. The validity of this model is supported by in vivo<br />

studies where partial thickness scarification of the load-bearing<br />

femoral condyle cartilage in dogs leads to progressive subchondral<br />

bone remodeling, osteophytosis and synovitis changes similar to<br />

that seen with transection of the anterior cruciate ligament (1-3).


It is of course obvious, that the entry point into the above outlined<br />

vicious cycle of joint degeneration could be in any of the involved<br />

tissues. Indeed in vivo models of OA and its characteristic cartilage<br />

degradation have been described where the primary initiating event<br />

is in the ligament or meniscus, synovium, or subchondral bone<br />

(reviewed in (4-6)). What these various models demonstrate is the<br />

strong interrelationship between all joint tissues, and highlight<br />

that to focus on one tissue in isolation and without reference to<br />

its neighbors/partners is fraught with risk. Longitudinal studies of<br />

human knee joints using sensitive MRI imaging techniques, have<br />

demonstrated significant association between pathology in the<br />

various joint tissues. Importantly, there are significantly increased<br />

risk/odds of incident and/or rapid progressive cartilage erosion<br />

(in knees without or with pre-existing OA, respectively) with the<br />

presence of subchondral bone lesions, degenerative meniscal<br />

change and synovitis with joint effusion (7-10). These studies suggest<br />

that changes in these other joint tissues are not just secondary<br />

to cartilage degradation, but may be important in initiating and<br />

driving the chondral change. Whether the mechanisms behind the<br />

influence of these other joint tissues on cartilage degradation are<br />

mechanical, soluble signaling molecules (cytokines, growth factors),<br />

direct contribution to degradative enzyme burden in the joint or a<br />

combination remains to be determined. We have undertaken various<br />

in vitro and in vivo studies to address these questions.<br />

We have studied the relationship between cartilage and bone<br />

changes in a surgical model of post-traumatic (pt)OA using wild<br />

type (WT) and genetically-modified (GM) mice. <strong>Cartilage</strong> erosion,<br />

subchondral bone sclerosis and osteophyte size were scored in<br />

serial histological sections of knee joints from over 400 age matched<br />

male WT mice 2-16 weeks after surgical destabilization of the medial<br />

meniscus (DMM). Not surprisingly there is a significant temporal<br />

progression of cartilage and bone pathology after meniscal injury.<br />

There is a significant correlation between cartilage damage and<br />

both subchondral bone sclerosis and osteophyte size at all times,<br />

but particularly in early and progressive phases of joint disease.<br />

Ordinal logistic regression analysis demonstrates that the odds of<br />

worse cartilage damage are significantly greater when increased<br />

SC bone density (2.6 x) or osteophytes (3.7 x) are present, either<br />

separately or together and when corrected for (i.e. independent<br />

of) time post surgery and differences within strains of mice. This<br />

association supports the hypothesis of a causal relationship<br />

between subchondral bone sclerosis and osteophytosis and<br />

cartilage degradation. However, in none of the 12 different GM mice<br />

strains examined were the same associations maintained, and none<br />

of the GMs has a significant effect (better or worse) on all three of<br />

the pathology outcomes. While pathology in bone and cartilage are<br />

associated in ptOA, the data from GM mice indicates this parallel<br />

pathology and not causally linked, and disease in one tissue can be<br />

modulated independent of the other. This has important implications<br />

for OA therapy, and on the potential effects on any cartilage repair<br />

strategy.<br />

In the knee joint the meniscus plays a significant mechanical role in<br />

dissipating and transferring loads, as well as contributing to joint<br />

stability. Disruption of the ability of the meniscus to appropriately<br />

undertake these mechanical functions (through excision, transection<br />

or severing its attachment to the tibia) results in all species examined<br />

(including humans), all the hallmark pathologies of OA including<br />

progressive cartilage destruction. While excision of the meniscus<br />

likely has its major impact through altering joint mechanics, injury<br />

to or degenerative change in the meniscus may also contribute to<br />

cartilage breakdown directly through release of catabolic enzymes.<br />

There is a distinct zonal difference in the catabolic events that occur<br />

in the meniscus, the inner cartilaginous zone having significant<br />

ADAMTS-driven proteolysis while in the outer fibrous zone MMP<br />

activity predominates (11). The expression ADAMTS4, ADAMTS5,<br />

MMP13 was significantly higher in meniscus than cartilage from<br />

the same joint in response to IL-1 and TNF stimulation. The outer<br />

meniscus secreted active MMP-2 and MMP-13 ± cytokine stimulation.<br />

The meniscus is thus a significant source of key enzymes involved<br />

in cartilage breakdown, and may have a major impact on both<br />

progressive cartilage breakdown in OA, and the success of cartilage<br />

repair strategies.<br />

The synovium is a potential source of degradative enzymes and<br />

catabolic cytokines. Whether inflammation (synovitis) in OA is a<br />

secondary event to cartilage degradation or may play a primary<br />

role in initiation and progression of cartilage breakdown is unclear.<br />

Long-term treatment with NSAIDs and corticosteroids do not have<br />

a disease modifying effect in human OA. GM mice with deletion of<br />

prostaglandin E synthase, Cox1 or Cox2 do not show any diminution<br />

of ptOA cartilage degradation (12, 13). Clinical trials of IL-1 or TNF<br />

blocking strategies in established human OA have not shown<br />

Extended Abstracts 171<br />

significant benefit, however, IL-1beta deficient mice do have reduced<br />

cartilage loss in ptOA (14), and treatment with IL-1ra abrogates<br />

cartilage damage in animal models of ptOA (15). It may be that there<br />

is a window of opportunity to target inflammation after joint injury to<br />

protect against long term OA and cartilage damage. However there<br />

is very limited information on the changes in joint inflammation that<br />

occur in OA, and it may be that the principal inflammatory cytokine<br />

or cell type, and thus treatment target changes with time. We have<br />

evaluated the cellular and expression changes in the synovium that<br />

occur with time after joint injury and between OA-inducing (DMM<br />

surgery) versus non-inducing (sham) trauma. Increased IL-1 and TNF<br />

expression occur rapidly in both DMM and sham but return t baseline<br />

by 7-14 days. In contrast there were distinct temporal patterns of<br />

activated macrophage, CD4 and CD8 lymphocyte accumulation in the<br />

synovium and importantly significant differences between sham and<br />

DMM surgeries. Importantly this was a local event as no systemic<br />

changes were observed. This exciting data suggests that cellular<br />

inflammatory/immune events occur early in OA and may drive the<br />

disease process and progression. Current studies are targeting<br />

different cell populations in the joint at distinct times after injury to<br />

determine whether this may be a viable treatment to prevent the<br />

onset and halt the progression of OA.<br />

References:<br />

1. Frost-Christensen LN, Mastbergen SC, Vianen ME, Hartog<br />

A, DeGroot J, Voorhout G, et al. Degeneration, inflammation,<br />

regeneration, and pain/disability in dogs following destabilization<br />

or articular cartilage grooving of the stifle joint. Osteoarthritis<br />

<strong>Cartilage</strong>. 2008;16(11):1327-35.<br />

2. Marijnissen AC, van Roermund PM, Verzijl N, Tekoppele JM, Bijlsma<br />

JW, Lafeber FP. Steady progression of osteoarthritic features in the<br />

canine groove model. Osteoarthritis <strong>Cartilage</strong>. 2002;10(4):282-9.<br />

3. Sniekers YH, Intema F, Lafeber FP, van Osch GJ, van Leeuwen JP,<br />

Weinans H, et al. A role for subchondral bone changes in the process<br />

of osteoarthritis; a micro-CT study of two canine models. BMC<br />

Musculoskelet Disord. 2008;9:20.<br />

4. Little C, Smith M. Animal models of osteoarthritis. Current<br />

Rheumatology Reviews 2008;4:175-82.<br />

5. Little CB, Fosang AJ. Is cartilage matrix breakdown an appropriate<br />

therapeutic target in osteoarthritis--insights from studies of aggrecan<br />

and collagen proteolysis? Curr Drug Targets. 2010;11(5):561-75.<br />

6. Smith M, Little C. Experimental Models of Osteoarthritis. In:<br />

Moskowitz R, Altman R, Hochberg M, Buckwalter J, Goldberg V,<br />

editors. Osteoarthitis: Diagnosis and Medical/Surgical Management.<br />

Philadelphia: Lipincott Williams & Wilkins; 2007. p. 107-25. .<br />

7. Felson DT. Developments in the clinical understanding of<br />

osteoarthritis. Arthritis Res Ther. 2009;11(1):203.<br />

8. Hill CL, Hunter DJ, Niu J, Clancy M, Guermazi A, Genant H, et al.<br />

Synovitis detected on magnetic resonance imaging and its relation<br />

to pain and cartilage loss in knee osteoarthritis. Ann Rheum Dis.<br />

2007;66(12):1599-603.<br />

9. Hunter DJ, Zhang YQ, Niu JB, Tu X, Amin S, Clancy M, et al. The<br />

association of meniscal pathologic changes with cartilage loss in<br />

symptomatic knee osteoarthritis. Arthritis Rheum. 2006;54(3):795-<br />

801.<br />

10. Roemer FW, Zhang Y, Niu J, Lynch JA, Crema MD, Marra MD, et<br />

al. Tibiofemoral joint osteoarthritis: risk factors for MR-depicted fast<br />

cartilage loss over a 30-month period in the multicenter osteoarthritis<br />

study. Radiology. 2009;252(3):772-80.<br />

11. Fuller ES, Smith MM, Little CB, Melrose J. Zonal differences in<br />

meniscus matrix turnover and cytokine response. Osteoarthritis<br />

<strong>Cartilage</strong>. 2012;20(1):49-59.<br />

12. Glasson S, Blanchet T, Ma H, Hopkins B, Diane P, Carito B, et al.<br />

Evaluation of twelve knock-out mice following surgical induction of<br />

osteoarthritis. Trans Orth Res Soc. 2006;31:216.<br />

13. Yamakawa K, Kamekura S, Kawamura N, Saegusa M, Kamei<br />

D, Murakami M, et al. Association of microsomal prostaglandin E<br />

synthase 1 deficiency with impaired fracture healing, but not with<br />

bone loss or osteoarthritis, in mouse models of skeletal disorders.<br />

Arthritis Rheum. 2008;58(1):172-83.<br />

14. Glasson SS. In vivo osteoarthritis target validation utilizing<br />

genetically-modified mice. Curr Drug Targets. 2007;8(2):367-76.<br />

15. Calich AL, Domiciano DS, Fuller R. Osteoarthritis: can anti-cytokine<br />

therapy play a role in treatment? Clin Rheumatol. 2010;29(5):451-5.


172<br />

Acknowledgments:<br />

Extended Abstracts<br />

Portions of the work presented were funded by grants received from<br />

the National Health and Medical Research Council of Australia, the<br />

Hillcrest Foundation through Perpetual Philanthropies, Arthritis<br />

Australia, Pfizer Inc, and Sydney Medical School Foundation.<br />

22.2<br />

<strong>Cartilage</strong> repair; what does it mean and how to assess cartilage<br />

repair outcome?<br />

L.E. Dahlberg<br />

Malmö/Sweden<br />

Introduction: Often, cartilage repair is considered restoring<br />

macroscopic defects. Defects may originate as a result of an acute<br />

injury or because of osteoarthritis (OA). However, defects may also be<br />

considered as molecular loss in an intact matrix. Where there may be<br />

a rationale to restore defects after an acute injury by e.g., stimulating<br />

matrix production by means of cartilage cell transplantation, there is no<br />

convincing evidence for restoring cartilage loss in OA joints to improve<br />

symptoms or decrease further OA deterioration. In OA it seems more<br />

rationale to use the repair capacity of cartilage chondrocytes within<br />

the matrix to support the restoration of degraded and lost matrix<br />

molecules before macroscopic loss occur. This abstract, as well as my<br />

talk, will therefore focus on the latter form of cartilage repair.<br />

Content: In OA, it is generally agreed that as joint space narrowing and<br />

osteophytes are identified by radiography, cartilage matrix changes<br />

involved in the pathogenesis of OA have occurred for a long time. The<br />

fact that OA joint symptoms commonly come about in patients without<br />

radiographic OA changes supports this and suggests that in the interest<br />

of the patient, the OA diagnosis should be based on symptoms rather<br />

than x-ray findings (1). Pain mechanisms in OA are not well understood,<br />

but clearly the cartilage matrix molecular status in symptomatic patients<br />

without radiographic changes needs further attention. Risk for knee<br />

OA is associated with meniscectomy, obesity, muscle weakness and<br />

major injury whereas hip OA is commonly associated with congenital<br />

and developmental defects such as acetabular dysplasia and possibly<br />

obesity (2, 3). Patients with inreased OA risk may serve as OA models.<br />

The cartilage matrix consists of two main macromolecules, type II<br />

collagen and the large, aggregating proteoglycan, aggrecan. With its<br />

highly negatively charged glycosaminoglycans (GAG), aggrecan can<br />

bind up to 50 times its weight in water resulting in a swelling pressure<br />

that is normally constrained by the tensile strength of the collagen<br />

fibrillar network (4). This interaction is the key mechanism behind the<br />

visco-elastic properties of articular cartilage that provides joints with<br />

the necessary resistance to mechanical loading (5).<br />

The hallmark of OA, cartilage loss, suggests a metabolic imbalance with<br />

less repair and increased enzymatic degradation and loss of matrix<br />

molecules in the pathogenesis. In a cartilage of low quality, depleted<br />

of aggrecan and other matrix molecules, more stress is transmitted to<br />

the collagen fiber-network. That may eventually result in fatigue, fiber<br />

damage and cartilage oedema which will further dilute the matrix and<br />

impair the biomechanical properties.<br />

Clearly, we need methods that identify in vivo molecular joint cartilage<br />

alterations related to early OA pathogenesis in order to enable the<br />

development of treatments with the ability to slow down matrix<br />

degradation and stimulate molecular repair. Similarly, matrix repair<br />

in acutely injured subjects by means of transplantation needs to be<br />

monitored at the molecular level and not only via macroscopic grading<br />

systems. In this respect MRI is a valuable tool. In the last decade<br />

several different techniques have emerged that enable the monitoring<br />

of molecular cartilage quality; dGEMRIC, gagCEST, T1rho, sodium-MRI,<br />

different T1 and T2 parameters (6, 7, 8, 9, 10). dGEMRIC, gagCEST,<br />

T1rho and sodium-MRI are considered GAG-related whereas T1 and T2<br />

measurements are less specific and influenced by water and collagen<br />

content and other variables. gagCEST and sodium-MRI are still at an<br />

experimental level whereas there are considerable clinical experience<br />

with dGEMRIC although the technique still needs further validation.<br />

Recent in vivo and in vitro dGEMRIC studies have increased our<br />

understanding of cartilage biology, specifically matrix changes related<br />

to joint biomechanics in health and disease, as load, over-load,<br />

exercise and x-ligament and meniscus injuries. dGEMRIC estimates<br />

cartilage GAG content using a negatively charged contrast agent that,<br />

after intravenous injection, distributes into the cartilage inversely to<br />

cartilage negative charge (6).<br />

More and more, we understand the cartilage contrast-distribution in<br />

dGEMRIC which has helped us to interpret the MRI signal in different<br />

clinical cohorts. In vivo, there is no equilibrium between surrounding<br />

fluid contrast concentration and that in the cartilage but a continued<br />

wash-in and wash-out of the contrast medium which is in contrast to<br />

the in vitro experimental situation. In in vitro examinations, variables<br />

determining contrast diffusion as well as relationship between<br />

Gadolinium concentration and T1Gd, usually regarded as the dGEMRIC<br />

index, can be determined. Recent studies examining depth-wise<br />

contrast concentration in vivo and in vitro show the importance of<br />

having a strict protocol regarding the timing of MRI examinations after<br />

contrast-injection and also that the cartilage thickness needs to be<br />

included in the protocol (11). Determining cartilage GAG by dGEMRIC<br />

and in the synovial fluid by a colorimetric assay, may give information<br />

on the relationship between metabolic and structural changes (12).<br />

A cross-sectional dGEMRIC study showed that the index was higher in<br />

athletes than in sedentary people which suggest an adaptive capacity<br />

of human knee cartilage (13). An exercise study in healthy subjects<br />

showed improved cartilage quality after running (14). To examine the<br />

influence of exercise to the cartilage in meniscetcomized subjects,<br />

a randomized trial showed that a 4 month exercise intervention<br />

significantly increased the dGEMRIC index (15). Interestingly, exercise<br />

is also a core treatment of OA patients and meniscectomy is a risk factor<br />

for OA. However, at the same time OA is often considered a “wear and<br />

tear” disease which may seem contradictory to treatment guidelines.<br />

In acutely injured cruciate ligament subjects, dGEMRIC index generally<br />

decreases (12) indicating a generalized cartilage matrix impairment<br />

with GAG loss. After meniscectomy, known to increase OA risk,<br />

dGEMRIC index is low in the meniscectomized compartment (16).<br />

Accordingly, a study of cruciate ligament injuried patients shows<br />

that compartments that had suffered a meniscus injury had a lower<br />

dGEMRIC index compared to those without meniscus injury (17). In<br />

patients with early cartilage disease, the dGEMRIC index is lower in<br />

diseased than in normal appearing cartilage (18, 19). At follow-up,<br />

patients having a low dGEMRIC index at baseline had increased risk<br />

of developing radiographic OA 6 years later (20). In patients with hip<br />

dysplasia, another group at OA risk, the dGEMRIC index was lower than<br />

in asymptomatic subjects, despite the absence of radiographic OA<br />

changes. Surgical treatment of dysplasia (periacetabular osteotomy)<br />

is associated with a poor outcome if the patient had a low preoperative<br />

dGEMRIC index (21).<br />

Other known risk factors for OA are obesity and muscle weakness. To<br />

identify possible correlates between molecular GAG changes and joint<br />

disease, these risk factors have been related to the dGEMRIC index. A<br />

lower quadriceps muscle strength and worse muscular performance<br />

is associated with inferior cartilage quality (dGEMRIC index) (16).<br />

Recently, Anandacoomarasamy et al. in an obese cohort study (22),<br />

where only one third of had clinical OA, subjects were scheduled for<br />

gastric banding or exercise and diet programs. Results show weight<br />

loss to be associated with improved dGEMRIC index and cartilage<br />

quantity medially in the knee.<br />

These studies provide evidence that structural changes related to<br />

GAG is involved in the OA process and that such alterations may be<br />

identified with dGEMRIC. Furthermore, they support the content that<br />

cartilage is a dynamic tissue and that molecular loss that may be<br />

related to OA can be repaired by intrinsic mechanisms. However to<br />

enable this, monitoring techniques that identifies cartilage events at<br />

the molecular level need to be utilized.<br />

References:<br />

1 Ericsson YB, Roos EM, Dahlberg L. Muscle strength, functional<br />

performance and self-reported outcome four years after arthroscopic<br />

partial meniscectomy in middle-aged patients. Arthritis Care Res.<br />

2006;55:946-952.<br />

2 Felson DT, Lawrence RC, Dieppe PA, et al. Osteoarthritis: new insights.<br />

Part 1: the disease and its risk factors. Ann Intern Med. 2000;133:635-<br />

46.<br />

3 Jacobsen S, Sonne-Holm S. Hip dysplasia: a significant risk factor<br />

for the development of hip osteoarthritis. A cross-sectional survey.<br />

Rheumatology (Oxford). 2005;44:211-8.<br />

4 Venn M, Maroudas A. Chemical composition and swelling of normal<br />

and osteoarthrotic femoral head cartilage. I. Chemical composition.<br />

Ann Rheum Dis. 1977;36:121-9.<br />

5 Mow VC, Lai M. Biorheology of swelling tissue. Biorheology.<br />

1990;27:110-9.


6 Dahlberg LE, Lammentausta, Tiderius CJ, Nieminen MT. In Vivo<br />

Monitoring of Joint <strong>Cartilage</strong> <strong>–</strong> Lessons to Be Learned by Delayed<br />

Gadolinium Contrast-enhanced Magnetic Resonance Imaging of<br />

<strong>Cartilage</strong>. European Musculoskeletal review. In press<br />

7 Wheaton AJ, Casey FL, Gougoutas AJ, Dodge GR, Borthakur A, Lonner<br />

JH, et al. Correlation of T1rho with fixed charge density in cartilage.<br />

J.Magn.Reson.Imaging. 2004; 20:519-25.<br />

8 Wheaton AJ, Borthakur A, Shapiro EM, Regatte RR, Akella SV, Kneeland<br />

JB, et al. Proteoglycan loss in human knee cartilage: quantitation with<br />

sodium MR imaging--feasibility study. Radiology. 2004; 231:900-5.<br />

9 Burstein D, Velyvis J, Scott KT, Stock KW, Kim YJ, Jaramillo D, Boutin<br />

RD, Gray ML. Protocol issues for delayed Gd(DTPA)(2-)-enhanced MRI<br />

(dGEMRIC) for clinical evaluation of articular cartilage. Magn Reson<br />

Med. 2001;45:36-41.<br />

10 Tiderius CJ, Olsson LE, de Verdier H, Leander P, Ekberg O, Dahlberg L.<br />

Gd-DTPA2)-enhanced MRI of femoral knee cartilage: a dose-response<br />

study in healthy volunteers. Magn Reson Med. 2001;46:1067-71.<br />

11 Hawezi Z, Tiderius CJ, Svensson J, Dahlberg LE, Lammentausta E.<br />

Temporal dynamics of Gd-enhanced T1 relaxation time in deep and<br />

superficial femoral articular cartilage. 17th Annual Scientific Meeting,<br />

<strong>International</strong> <strong>Society</strong> for Magnetic Resonance in Medicine 16:3963,<br />

2009. Honolulu, Hawaii.<br />

12 Tiderius CJ, Olsson LE, Nyquist F, Dahlberg L. <strong>Cartilage</strong><br />

glycosaminoglycan loss in the acute phase after an anterior cruciate<br />

ligament injury: delayed gadolinium-enhanced magnetic resonance<br />

imaging of cartilage and synovial fluid analysis. Arthritis Rheum.<br />

2005;52:120-7.<br />

13 Tiderius CJ, Svensson J, Leander P, Ola T, Dahlberg L. dGEMRIC<br />

(delayed gadolinium-enhanced MRI of cartilage) indicates adaptive<br />

capacity of human knee cartilage. Magn Reson Med. 2004;51:286-90.<br />

14 Van Ginckel A, Baelde N, Almqvist KF, Roosen P, McNair P, Witvrouw<br />

E. Functional adaptation of knee cartilage in asymptomatic female<br />

novice runners compared to sedentary controls. A longitudinal analysis<br />

using delayed Gadolinium Enhanced Magnetic Resonance Imaging of<br />

<strong>Cartilage</strong> (dGEMRIC). Osteoarthritis <strong>Cartilage</strong>. 2010 Dec;18(12):1564-9.<br />

15 Roos EM, Dahlberg L. Positive effects of moderate exercise<br />

on glycosaminoglycan content in knee cartilage: a four-month,<br />

randomized, controlled trial in patients at risk of osteoarthritis. Arthritis<br />

Rheum. 2005;52:3507-14.<br />

16 Ericsson YB, Tjörnstrand J, Tiderius CJ, Dahlberg LE. Relationship<br />

between cartilage glycosaminoglycan content (assessed with dGEMRIC)<br />

and OA risk factors in meniscectomized patients. Osteoarthritis<br />

<strong>Cartilage</strong>. 2009;17:565-70.<br />

17 Neuman P, Tjörnstrand J, Svensson J, Ragnarsson C, Roos H, Englund<br />

M, Tiderius CJ, Dahlberg LE. Longitudinal assessment of femoral knee<br />

cartilage quality using contrast enhanced MRI (dGEMRIC) in patients<br />

with anterior cruciate ligament injury--comparison with asymptomatic<br />

volunteers. Osteoarthritis <strong>Cartilage</strong>. 2011 Aug;19(8):977-83.<br />

18 Tiderius CJ, Olsson LE, Leander P, Ekberg O, Dahlberg L. Delayed<br />

gadolinium-enhanced MRI of cartilage (dGEMRIC) in early knee<br />

osteoarthritis. Magn Reson Med. 2003;49:488-92.<br />

19 Tiderius CJ, Jessel R, Kim YJ, Burstein D. Hip dGEMRIC in asymptomatic<br />

volunteers and patients with early osteoarthritis: the influence of<br />

timing after contrast injection. Magn Reson Med. 2007;57:803-5.<br />

20 Owman H, Tiderius CJ, Neuman P, Nyquist F, Dahlberg LE. Association<br />

between findings on delayed gadolinium-enhanced magnetic<br />

resonance imaging of cartilage and future knee osteoarthritis. Arthritis<br />

Rheum. 2008;58:1727-30.<br />

21 Kim YJ, Jaramillo D, Millis MB, Gray ML, Burstein D. Assessment<br />

of early osteoarthritis in hip dysplasia with delayed gadoliniumenhanced<br />

magnetic resonance imaging of cartilage. J Bone Joint Surg<br />

Am. 2003;85-A:1987-92.<br />

22 Anandacoomarasamy A, Leibman S, Smith G, Caterson I, Giuffre B,<br />

Fransen M, Sambrook PN, March L.Weight loss in obese people has<br />

structure-modifying effects on medial but not on lateral knee articular<br />

cartilage. Ann Rheum Dis. 2012 Jan;71(1):26-32.<br />

Extended Abstracts 173<br />

Acknowledgments:<br />

Supported by grants from the Swedish Research Council, King Gustaf<br />

V’s 80-year Fund, Swedish National Center for Research in Sports, The<br />

Herman Järnhardt Foundation, The Alfred Österlund Foundation and<br />

The Skåne University Hospital Funds.<br />

24.1.1<br />

Change of thinking in treating cartilage defects in the hip<br />

M. Philippon<br />

Vail/United States of America<br />

Introduction: Chondral Injuries occur in conjunction with many hip<br />

disorders, both traumatic and atraumatic. These injuries can be acute,<br />

chronic or degenerative and partial or full thickness defects.<br />

Content: Recently, literature showed that chondral defects with<br />

identified in 79% of hip arthroscopies.(Arthroscopy Journal. 2008)<br />

There is also a strong association between chondral defects and<br />

femoroacetabular impingement. Full-thickness of acetabular cartilage<br />

occurred with an average alpha angle of 60 degrees while hips with no<br />

delamination had an average alpha angle of 51 degrees. The average<br />

alpha angles was also associated with chondral defect size. Treatment<br />

Currently chondral defects of the hip are most commonly treated<br />

by debridement or microfracture. Microfracture is well-established<br />

treatment for fill thickness chondral lesions in the knee. Perforations<br />

in the subchondral bone bring marrow elements into the chondral<br />

defect that are capable of differentiating into fibrocartilaginous tissue.<br />

Advances in the field of hip arthroscopy make it possible to use this<br />

technique for treatment of chondral injuries of the hip. Indication for<br />

the use of microfracture include focal and contained lesions, 2-4 cm in<br />

size, full thickness loss of articular cartilage in weight-bearing areas,<br />

and unstable chondral flaps overlying intact subchondral bone. Contra-<br />

Indications include partial thickness defects and patient unwillingness<br />

to comply with rigorous rehabilitation protocol. Proper technique<br />

and rehabilitation are critical to the success of this procedure. New<br />

techniques have been reported recently in the literature. Fibrin glue<br />

has been used to treat delaminated acetabular articular cartilage.<br />

More research is needed to determine the survivorship of this repair<br />

over time. A recent study also reported on the use of autologous<br />

chondrocyte transplantation compared to debridement. This study<br />

showed that ACT have improved outcomes compared to debridement<br />

in large chondral lesions. Chondral Grafts are also being reported in the<br />

literature for large chondral defects. These grafts include mosiacplasty<br />

and biological plugs. Currently our practice is to use these grafts when<br />

bone loss is present on the femoral head. We then microfracture in<br />

between plugs and fill the donor hole with a biological plug.<br />

References:<br />

Crawford K, Philippon MJ, et al. Microfracture of the hip in athletes.<br />

Clin Sports Med. 2006 ;25:327-35<br />

Field RE, Rajakulendran K,et al. Arthroscopic grafting of chondral defects<br />

and subchondral cysts of the acetabulum. Hip Int. 2011;21:479-86<br />

Fontana A, Bistolfi A, et al. Arthroscopic Treatment of Hip Chondral<br />

Defects: Autologous Chondrocyte Transplantation Versus Simple<br />

Debridement-A Pilot Study. Arthroscopy. 2011 Dec 3.<br />

Johnston TL, Schenker ML, et al. Relationship between offset angle alpha<br />

and hip chondral injury in femoroacetabular impingement. Arthroscopy.<br />

2008;24:669-75.<br />

Philippon MJ, Briggs, KK, et al. Outcomes Following Hip Arthroscopy<br />

for femoroacetabular Impingement with Associated Chondrolabral<br />

Dysfunction Minimum 2 year Follow-up. JBJS Br. 2009;91:16-23.<br />

Philippon MJ, Kuppersmith DA, et al. Arthroscopic findings following<br />

traumatic hip dislocation in 14 professional athletes. Arthroscopy.<br />

2009;25:169-74.<br />

Philippon MJ, Schenker ML, et al. Can microfracture produce repair tissue<br />

in acetabular chondral defects? Arthroscopy. 2008;24:46-50.<br />

Philippon MJ, Schenker ML, et al. Revision Hip Arthroscopy. Am J Sports<br />

Med.2007;35:1918-1921.<br />

Stafford GH, Bunn JR, et al.Arthroscopic repair of delaminated acetabular<br />

articular cartilage using fibrin adhesive. Results at one to three years.<br />

Hip Int. 2011;21:744-50.


174<br />

24.1.2<br />

ACI in the hip joint<br />

J.B. Richardson<br />

Oswestry/United Kingdom<br />

Extended Abstracts<br />

Introduction: Autologous chondrocyte implantation (ACI) has been<br />

used most commonly as a treatment for cartilage defects in the<br />

knee and there are few studies of its use in other joints. Can ACI<br />

work in the hip? We have used hip arthroscopy for over 15 years and<br />

identified a significant proportion of patients with chondral defects<br />

but relatively normal radiology. Increasing use of hip arthroscopy<br />

will identify more patients with localised chondral defects of both<br />

the femoral head and the acetabulum. Can these problems be<br />

treated by autologous chondrocyte implantation?<br />

Content: Assessing chondral defects in the hip requires assessment<br />

of loss of function, accurate mapping of size and location of defects,<br />

and identification of predisposinbg factors. These three dimensions<br />

are useful for evaluation of any joint with a chondral defect. We<br />

consider the development of arthritis to be a process of loss of<br />

function of the joint as an organ. Thus there is not one particular<br />

point at which a joint becomes ‘arthritic’, but a gradual process that<br />

develops over time. Some of this process is pre-clinical and thus<br />

joint narrowing or osteophyte formation often develops prior to<br />

symptoms. These pre-clinical features can be assessed by imaging,<br />

but may not lead to treatment. We propose chondral defects to be part<br />

of this process, and also are not always symptomatic. Indeed many<br />

heal with repair tissue, as is seen in a full thickness ACI harvest site.<br />

This new tissue we propose to call novocartilage. It is characterised<br />

by being well attached to underlying bone, to have type 2 collagen<br />

and variable proteoglycan content but has no ingrowth of vessels or<br />

nerves. This is a true repair tissue which may remodell to articular<br />

cartilage. It may have differing amounts of hyaline characteristics<br />

as assessed by polarised light microscopy. We believe this tissue is<br />

an end in itself as is callus healing a fracture en route to remodelled<br />

bone. Renovocartilage may be sufficient for purpose to slsoow joint<br />

function as callus can be sufficient for purpose, although it consists<br />

of woven bone. The scientist may seek regeneration of the original<br />

tissue, but the clinician may be satisfied with a functioning repair<br />

tissue. The process of organ failure that is seen as a chondral defect<br />

of the knee may be quantified by a self-assessed score of function for<br />

the knee that has been weighted and validated as an interval score<br />

(Smith et al., 2009). In the hip we advocate a modified Harris Hip<br />

score which has allowed evaluation of 5000 BHR hip resurfacings<br />

(Khan et al, 2009). Mapping by a geometric projection allows the<br />

arthoscopic site to be correlated with the MRI findings (Gokhale et al,<br />

2008). Why develop biological repair for the hip? Function following<br />

hip resurfacing or hip replacement can be excellent. This sets a high<br />

standard for a biological therapy. However the high function leads<br />

to wear and eventual failure of most devices. Metal-polythene hip<br />

replacements have had an excellent outcome in the older patient,<br />

but relatively high failure rates of around 12% by 10 years in the<br />

under-50 year-olds. Higher rates of failure in osteoarthritis leave<br />

only 50% surviving by 20 years. A patient having hip replacement<br />

for osteoarthritis at the age of 30 is likely to have several revisions<br />

over the next 50 years, and each revision is marked by loss of bone.<br />

Metal-on-metal bearings were re-introduced as a solution for hip<br />

resurfacing and here for most designs have been very successful<br />

in the young active male patients. We have identified femoral head<br />

size less than 46mm and female gender as pre-operative factors<br />

that identify earlier failure of the implant. Stemmed metal-metal hip<br />

designs may suffer from high local levels of metal debris as the trunion<br />

is an additional source of metal debris. New ceramic designs and<br />

cross-linked polythene containing vitamin E may provide improved<br />

solutions for the future, but all synthetic implants carry a risk of<br />

infection that is difficult to eradicate. There is therefore an argument<br />

for developing biological reconstructions that maintain bone stock<br />

and avoid the problems of synthetic bearing surfaces. The cost<br />

that can be supported can be considered not just the cost of a joint<br />

replacement but the cost of a difficult revision post-phoned in later<br />

life. This life-time perspective is similar for the hip, ankle and knee. A<br />

surgeon’s perspective may focus on the duration of survival for each<br />

implant, but the patient generally seeks high levels of function. This<br />

supports a routine self-assessed measure of function of the affected<br />

joint at every clinic with ‘hot-audit’ providing a chart for each patient<br />

in the clinic. Renovo-cartilage: an end in itself? 1. Pain relief which<br />

should be prolonged to 10 years 2. Improved function measured on<br />

a self-assessment scale validated for chondral defects (Sith et al.,<br />

2009) 3. Absence of secondary changes : a) osteophyte formation,<br />

b) bone marrow oedema. 4. Preferably evidence of: a) Good<br />

attachment to underlying bone b) Lateral integration c) Sufficient<br />

thickness d) Low surface friction e) Resistance to fluid movement<br />

In the hip the development of different patterns of cartilage loss is<br />

not well documented. The femoral head and the acetabulum appear<br />

to have separate patterns, and are usefully considered separately.<br />

Cysts are a difficult problem to successfully bone-graft in the<br />

hip, presumably because of the very high pressures that develop<br />

under load compared to other joints. Bone loss can be addressed<br />

in the femoral head by mosaicplasty combined with ACI. Avascular<br />

necrosis of the femoral head has been treated by mesenchymal<br />

stem cell insertion in France, and in Tromsoe, Norway by ACI alone.<br />

Our preferred technique has been the use of a plug of autologous<br />

bone from the greater trochanter, either 15mm or 20mm in diameter.<br />

This plug is shaped to fit the normal contour of the femoral head<br />

and then ACI performed over the top. We have treated several<br />

patients with femoral head chondral or osteochondral defects of six<br />

cm square average area. In all cases surgical dislocation of the hip<br />

was necessary for surgery. We have observed some excellent results<br />

(Akimau, 2006) but in our experience patients with cyst formation<br />

do not respond as well. Initially we used the ipsilateral knee as a<br />

source of cartilage for culture but in later cases used the hip. The<br />

normal knee will have symptoms for several months following<br />

biopsy, but this does not hinder the rehabilitation of the hip. These<br />

hips have been treated between 2 and 10 years ago. Five patients<br />

have progressed to hip replacement relatively quickly. We believe<br />

this is partly due to the high levels of pain-free function that hip<br />

replacement can offer the younger patient. All these patients had<br />

cyst formation pre-operatively and we therefore caution against<br />

undertaking ACI in the presence of cyst formation. We do not believe<br />

it is technically feasible to perform sutured ACI in the acetabulum<br />

by open surgery. The acetabulum is however ideally treated by<br />

arthroscopic ACI. Fontana et al reported in 2011 to have good results<br />

with arthroscopic matrix induced autologous in 15 patients. In their<br />

study 15 patients with an average 2cm acetabular defect had an<br />

improvement of over 40 points in the Harris Hip score with a 6 year<br />

follow-up. A similar group of patients treated with debridement had<br />

no significant improvement in symptoms. This group were strongly<br />

of the opinion that a non-sutured matrix would not stay in place on a<br />

defect of the femoral head. In conclusion it would appear that ACI is<br />

a possible option for femoral chondral and osteochondral defects by<br />

open procedure, and arthroscopic ACI a good option for acetabular<br />

defects.<br />

References:<br />

AKIMAU, P, BHOSALE, A, HARRISON, P.E. ROBERTS, S, MCCALL,<br />

I.W., JB RICHARDSON, J.B. ASHTON B.A. Autologous chondrocyte<br />

implantation with bone grafting for osteochondral defect due to<br />

posttraumatic osteonecrosis of the hip. Acta Orthopaedica. 2006;<br />

77 (2): 333-336.<br />

GOKHALE S, KHAN M, KUIPER JH, RICHARDSON JB, DAVIES JP An<br />

arthroscopic hip documentation form Arthroscopy. 2008; 24 (7):<br />

839 <strong>–</strong> 842.<br />

KHAN M, EDWARDS D, RICHARDSON JB Birmingham Hip Arthroplasty:<br />

Five to Eight Years Prospective Multicentre Centre Results Journal of<br />

Arthroplasty. 2009; 24(7): 1044 <strong>–</strong> 1050. H J SMITH, JB RICHARDSON,<br />

A TENNANT Modification and validation of the Lysholm Knee Scale<br />

to assess articular cartilage damage. Osteoarthritis and <strong>Cartilage</strong>.<br />

2009; 17:53-58.<br />

Fontana, A., Bistolfi, Crova, M., Rosso, F., Massazza, G. Arthroscopic<br />

Treatment of Hip Chondral Defects: Autologous Chondrocyte<br />

Transplantation Versus Simple Debridement—A Pilot Study<br />

Acknowledgments:<br />

With many thanks to Brian Ashton, Paul Harrison, Sally Roberts, Jan<br />

Herman Kuiper, Iain McCall and all in the Oscell team ()


24.1.3<br />

<strong>Cartilage</strong> repair in the hip joint<br />

R.M. Mardones<br />

Santiago/Chile<br />

Introduction: Chondral lesions of the hip represent a diagnostic<br />

challenge and can be an elusive source of pain. Treatment may<br />

present difficulties due to localization and spherical form of the joint<br />

and is most commonly limited to excision, debridement, thermal<br />

chondroplasty and microfractures.<br />

Content: Femoroacetabular impingement is frequently associated<br />

with chondral damage. The abnormal contact between femoral<br />

neck and acetabular rim results in labral detachment and chondral<br />

damage. In the hip the types of chondral lesions differs from other<br />

joints; the cam type femoroacetabular impingement is frequently<br />

associated with chondrolabral junction damage with the subsequent<br />

cartilage detachment. Delamination is a characteristic chondral<br />

lesion of the hip (wave sign), in which the cartilage detaches from<br />

the subchondral bone leading to a “bag lesion” and chondral<br />

flaps. Outerbridge is the most used chondral lesion classification<br />

system although delamination was not originally described it could<br />

be considered as a type III. Konan et al recently described a new<br />

classification system for hip chondral lesions, including the wave<br />

sign, delamination and chondrolabral lesions considering extension<br />

and location.<br />

The frequency of chondral lesions in hip arthroscopy for<br />

femoroacetabular impingement is high, up to 67.3% of the patients,<br />

as described by Nepple et al. Risk factors for the presence of a<br />

chondral lesion are: male, tonnis 1 or 2 and an alpha angle over<br />

50º.<br />

The arthroscopic treatment of chondral lesions of the hip is limited to<br />

excision (rim trimming and femoral neck osteoplasty), debridement,<br />

chondroplasty and microfractures. Rim trimming and femoral neck<br />

osteoplasty can lead to excise the chondral lesion if located in the<br />

overcoverage area. When the chondral damage extends beyond<br />

resection area the treatment of choice will be chosen according to<br />

Outerbridge or Konan classifications, as follows:<br />

- Type I or II with thermal chondroplasty<br />

- Delamination with fibrin glue or microfractures<br />

- Type III or IV (full thickness chondral lesion) with microfractures<br />

Thermal chondroplasty has shown to be a safe technique for closed<br />

chondral lesions leading to morphological changes with better<br />

structural characteristics than mechanical debridement.<br />

Delamination represents a treatment challenge among chondral<br />

lesions. Excising such an area of chondral instability seems an<br />

unnecessary surgical manoeuvre, particularly if the articular cartilage<br />

itself may contain a significant number of viable chondrocytes. The<br />

main objective is the reattachment of the cartilage to the underlying<br />

subchondral bone. This could be achieved with transchondral<br />

microfractures, forming an adherent retrolabral clot or with the use of<br />

an adhesive such as fibrin glue. Tzaveas and Villar report on a series<br />

of 19 patients treated with fibrin adhesive showing improvement in<br />

pain and function at 6 months and one year after surgery.<br />

The indications for microfracture of the hip are similar to the knee<br />

and include focal and contained lesions, tipically less than 2 to 4 cm2<br />

in size, (Outerbridge III or IV) including delamination. Microfracture<br />

is a marrow-stimulating procedure that brings undifferentiated<br />

stem cells from a subchondral perforation into the chondral defect.<br />

A clot formed in the microfractured area provides an environment<br />

for both pluripotent marrow cells and mesenchymal stem cells to<br />

differentiate into stable fibrocartilaginous tissue. Several studies<br />

had shown good midterm results with this technique; however we<br />

know that this fibrocartilaginous tissue does not have the required<br />

mechanical properties and eventually will fail, leading to advanced<br />

chondral damage and osteoarthritis.<br />

Animal and clinical studies have demonstrated that the use of a<br />

Platelet Rich Plasma clot or bone marrow mesenchymal stem cells<br />

over the microfractured area could lead to a better quality hyalinelike<br />

fibrocartilage with better mechanical properties.<br />

Gadolinium enhanced arthrography MRI is limited in its ability<br />

to reliably show the presence and extent of these lesions. More<br />

recently, however, delayed gadolinium enhanced MRI of cartilage<br />

(dgemric) has shown to be useful to delineate chondral damage<br />

Extended Abstracts 175<br />

within the hip.<br />

Our surgical technique is described below. In our clinical practice the<br />

treatment of choice for hip condral lesions is the use of a Platelet Rich<br />

Pplasma clot and bone marrow concentrate over the microfractured<br />

area with excellent results shown by delayed gadolinium enhanced<br />

MRI cartilage (dGEMRIC).<br />

Surgical Technique: After rim trimming and labrum refixation;<br />

cartilage assessment is made. If chondral lesion exists, we proceed to<br />

harvest autologous bone marrow stem cells, which are centrifugated<br />

obtaining 2 to 4 cc of autologous bone marrow <strong>–</strong> mesenchymal stem<br />

cells concentrate (average 14 millions of nucleated cells/cc3). At<br />

the same time, 50 cc of peripheral blood is taken and centrifugated<br />

twice, in order to obtain 4 cc of PRP (6 to 9x), ready to be activated<br />

with autologous thrombin. Treatment of chondral lesion is made<br />

as described by Steadman in the knee, with debridement of all<br />

remaining unstable cartilage, followed for the removal of the calcified<br />

plate. After preparation of the bed, multiple holes in the exposed<br />

subchondral bone plate are made, leaving about 3 to 4 mm between<br />

each. Once microfracture is complete, traction is release and we<br />

focus on the femoral osteoplasty, obtaining free range of motion<br />

with no abnormal contact between acetabular rim and femoral neckhead<br />

junction. At the end of the procedure, traction is reinstalled<br />

and we proceed to the final part of the procedure. After activation of<br />

Platelet Rich Plasma and clot formation, a slotted cannula is inserted<br />

via the anterior portal. Platelet Rich Plasma clot is inserted through<br />

the cannula and positioned over the microfractured area. A 21-gauge<br />

trocar is then inserted passing through previously located clot and<br />

autologous bone marrow <strong>–</strong> mesenchymal stem cells concentrate is<br />

instilled under PRP clot. Traction is then released and the procedure<br />

is finished.<br />

Rehabilitation protocol: Passive motion device is maintained for 8<br />

hours. Two crutches with partial weight bearing are indicated for 6<br />

to 8 weeks. Progressive physical activities are allowed.<br />

Preliminary Results: At the time, 13 patients with chondral lesion<br />

of the hip had been treated with microfractures and autologous<br />

bone marrow <strong>–</strong> mesenchymal stem cells concentrate transplanted<br />

on a Platelet Rich Plasma clot. All patients symptoms improved over<br />

the follow-up period of 8 months (4 to 12 months). Average Hip<br />

Outcome, Vail Hip and Modified Harris Hip scores for all patients<br />

showed significant improvement at 3 and 6 months. Dgemric of 4<br />

patients at 6 months postoperatively revealed complete defect fill<br />

and complete surface congruity with native cartilage.<br />

Conclusions: Surgical technique of a novel treatment for chondral<br />

lesions of the hip is presented. Autologous bone marrow <strong>–</strong><br />

mesenchymal stem cells concentrate transplanted on a Platelet Rich<br />

Plasma clot may be an effective approach to promote the repair<br />

of articular cartilage defects of the hip. Further study is currently<br />

underway to identify medium and long-term effects in cartilage<br />

repair and outcomes.<br />

References:<br />

1. Yen YM, Kocher MS. Chondral lesions of the hip: microfracture and<br />

chondroplasty. Sports Med Arthrosc. 2010 Jun;18(2):83-9.<br />

2. Nepple JJ, Carlisle JC, Nunley RM, Clohisy JC. Clinical and<br />

radiographic predictors of intra-articular hip disease in arthroscopy.<br />

Am J Sports Med. 2011 Feb;39(2):296-303.<br />

3. Konan S, Rayan F, Meermans G, Witt J, Haddad FS. Validation of<br />

the classification system for acetabular chondral lesions identified<br />

at arthroscopy in patients with femoroacetabular impingement. J<br />

Bone Joint Surg Br. 2011 Mar;93(3):332-6.<br />

4. Kaplan LD, Chu CR, Bradley JP, Fu FH, Studer RK. Recovery of<br />

chondrocyte metabolic activity after thermal exposure. Am J Sports<br />

Med. 2003 May-Jun;31(3):392-8.<br />

5. Johnston TL, Schenker ML, Briggs KK, Philippon MJ.<br />

Relationship between offset angle alpha and hip chondral injury in<br />

femoroacetabular impingement. Arthroscopy. 2008 Jun;24(6):669-<br />

75.<br />

6. Philippon MJ, Schenker ML, Briggs KK, Maxwell RB. Can<br />

microfracture produce repair tissue in acetabular chondral defects?<br />

Arthroscopy. 2008 Jan;24(1):46-50.


176 174<br />

24.1.2<br />

Extended Abstracts<br />

7. Tzaveas AP, Villar RN. Arthroscopic repair of acetabular chondral<br />

delamination with fibrin adhesive. Hip Int. 2010 Jan-Mar;20(1):115-9.<br />

ACI in the hip joint<br />

J.B. 8. Lotto Richardson ML, Wright EJ, Appleby D, Zelicof SB, Lemos MJ, Lubowitz<br />

Oswestry/United JH. Ex vivo comparison Kingdom of mechanical versus thermal chondroplasty:<br />

assessment of tissue effect at the surgical endpoint. Arthroscopy.<br />

Introduction: 2008 Apr;24(4):410-5. Autologous chondrocyte implantation (ACI) has been<br />

used most commonly as a treatment for cartilage defects in the<br />

knee 9. Edwards and there RB, are Lu few Y, Cole studies BJ, of Muir its use P, Markel in other MD. joints. Comparison Can ACI<br />

work of radiofrequency in the hip? We have treatment used hip and arthroscopy mechanical for over debridement 15 years and of<br />

identified fibrillated cartilage a significant in an proportion equine model. of patients Vet Comp with Orthop chondral Traumatol. defects<br />

but 2008;21(1):41-8.<br />

relatively normal radiology. Increasing use of hip arthroscopy<br />

will identify more patients with localised chondral defects of both<br />

the 10. Voss femoral JR, Lu head Y, Edwards and the RB, acetabulum. Bogdanske Can JJ, these Markel problems MD. Effects be<br />

treated of thermal by autologous energy on chondrocyte implantation?<br />

viability. Am J Vet Res. 2006<br />

Oct;67(10):1708-12.<br />

Content: Assessing chondral defects in the hip requires assessment<br />

of 11. loss McIlwraith of function, CW, accurate Frisbie DD, mapping Rodkey of WG, size and Kisiday location JD, Werpy of defects, NM,<br />

and Kawcak identification CE, Steadman of predisposinbg JR. Evaluation factors. of intra-articular These three mesenchymal<br />

dimensions<br />

are stem useful cells to for augment evaluation healing of any of joint microfractured with a chondral defect. defects. We<br />

consider Arthroscopy. the 2011 development Nov;27(11):1552-61. of arthritis to be a process of loss of<br />

function of the joint as an organ. Thus there is not one particular<br />

point 12. Frisbie at which DD, a Oxford joint becomes JT, Southwood ‘arthritic’, L, but Trotter a gradual GW, process Rodkey that WG,<br />

develops Steadman over JR, Goodnight time. Some JL, of McIlwraith this process CW. Early is pre-clinical events in and cartilage thus<br />

joint repair narrowing after subchondral or osteophyte bone microfracture. formation often Clin develops Orthop Relat prior Res. to<br />

symptoms. 2003 Feb;(407):215-27.<br />

These pre-clinical features can be assessed by imaging,<br />

but may not lead to treatment. We propose chondral defects to be part<br />

of 13. this Steadman process, JR, and Rodkey also are WG, not Rodrigo always symptomatic. JJ. Microfracture: Indeed surgical many<br />

heal technique with repair and rehabilitation tissue, as is seen to treat in a full chondral thickness defects. ACI harvest Clin Orthop site.<br />

This Relat new Res. tissue 2001 Oct;(391 we propose Suppl):S362-9.<br />

to call novocartilage. It is characterised<br />

by being well attached to underlying bone, to have type 2 collagen<br />

and 14. Steadman variable proteoglycan JR, Rodkey WG, content Briggs but KK. has Microfracture no ingrowth of to vessels treat full- or<br />

nerves. thickness This chondral is a true defects: repair tissue surgical which technique, may remodell rehabilitation, to articular and<br />

cartilage. outcomes. It J Knee may have Surg. differing 2002;15(3):170-6. amounts of hyaline characteristics<br />

as assessed by polarised light microscopy. We believe this tissue is<br />

an 15. end Milano in itself G, Sanna as is callus Passino healing E, Deriu a fracture L, Careddu en route G, to Manunta remodelled L,<br />

bone. Manunta Renovocartilage A, Saccomanno may MF, be Fabbriciani sufficient for C. The purpose effect to of slsoow platelet joint rich<br />

function plasma combined as callus can with be microfractures sufficient for purpose, on the treatment although of it chondral consists<br />

of defects: woven an bone. experimental The scientist study may in seek a sheep regeneration model. of Osteoarthritis<br />

the original<br />

tissue, <strong>Cartilage</strong>. but 2010 the Jul;18(7):971-80.<br />

clinician may be satisfied with a functioning repair<br />

tissue. The process of organ failure that is seen as a chondral defect<br />

of 16. the Gobbi knee A, may Karnatzikos be quantified G, Scotti by a C, self-assessed Mahajan V, Mazzucco score of L, function Grigolo for B.<br />

the One-Step knee that <strong>Cartilage</strong> has been <strong>Repair</strong> weighted with Bone and Marrow validated Aspirate as an interval Concentrated score<br />

(Smith Cells and et Collagen al., 2009). Matrix In the in hip Full-Thickness we advocate Knee a modified <strong>Cartilage</strong> Harris Lesions: Hip<br />

score Results which at 2-years has allowed follow-up. evaluation <strong>Cartilage</strong>, of July 5000 2011;2(3):286-299<br />

BHR hip resurfacings<br />

(Khan et al, 2009). Mapping by a geometric projection allows the<br />

arthoscopic 17. De Girolamo site L, to Bertolini be correlated G, Cervellin with the M, MRI Sozzi findings G, Volpi (Gokhale P. Treatment et al,<br />

2008). of chondral Why develop defects biological of the knee repair with for the one hip? step Function matrix-assisted following<br />

hip technique resurfacing enhanced or hip by replacement autologous can concentrated be excellent. bone This marrow: sets a high in<br />

standard vitro characterisation for a biological of mesenchymal therapy. However stem cells the high from function iliac crest leads and<br />

to subchondral wear and eventual bone. Injury. failure 2010 of Nov;41(11):1172-7.<br />

most devices. Metal-polythene hip<br />

replacements have had an excellent outcome in the older patient,<br />

but 18. Haleem relatively A, high El Singergy failure rates A, Sabry of around D, Atta 12% H, Rashed by 10 years L, Chu in C, the El<br />

under-50 Shewy M, year-olds. Azzam A, Higher Abdel rates Aziz of M. failure The Clinical in osteoarthritis Use of Human leave<br />

only Culture<strong>–</strong>Expanded 50% surviving Autologous by 20 years. Bone A patient Marrow having Mesenchymal hip replacement Stem<br />

for Cells osteoarthritis Transplanted at on the Platelet-Rich age of 30 is Fibrin likely to Glue have in the several Treatment revisions of<br />

over Articular the next <strong>Cartilage</strong> 50 years, Defects: and each A Pilot revision Study is and marked Preliminary by loss of Results. bone.<br />

Metal-on-metal <strong>Cartilage</strong> 2010;1(4):253-261.<br />

bearings were re-introduced as a solution for hip<br />

resurfacing and here for most designs have been very successful<br />

in Acknowledgments:<br />

the young active male patients. We have identified femoral head<br />

size less than 46mm and female gender as pre-operative factors<br />

that Catalina identify Larrain earlier M.D. failure of the implant. Stemmed metal-metal hip<br />

designs may suffer from high local levels of metal debris as the trunion<br />

is Alexander an additional Tomic source M.D. of metal debris. New ceramic designs and<br />

cross-linked polythene containing vitamin E may provide improved<br />

solutions Matías Salineros for the M.D. future, but all synthetic implants carry a risk of<br />

infection that is difficult to eradicate. There is therefore an argument<br />

for developing biological reconstructions that maintain bone stock<br />

and avoid the problems of synthetic bearing surfaces. The cost<br />

that can be supported can be considered not just the cost of a joint<br />

replacement but the cost of a difficult revision post-phoned in later<br />

life. This life-time perspective is similar for the hip, ankle and knee. A<br />

surgeon’s perspective may focus on the duration of survival for each<br />

implant, but the patient generally seeks high levels of function. This<br />

supports a routine self-assessed measure of function of the affected<br />

joint at every clinic with ‘hot-audit’ providing a chart for each patient<br />

in the clinic. Renovo-cartilage: an end in itself? 1. Pain relief which<br />

should be prolonged to 10 years 2. Improved function measured on<br />

a self-assessment scale validated for chondral defects (Sith et al.,<br />

2009) 3. Absence of secondary changes : a) osteophyte formation,<br />

b) bone marrow oedema. 4. Preferably evidence of: a) Good<br />

attachment to underlying bone b) Lateral integration c) Sufficient<br />

thickness d) Low surface friction e) Resistance to fluid movement<br />

In the hip the development of different patterns of cartilage loss is<br />

24.2.1<br />

not well documented. The femoral head and the acetabulum appear<br />

to have separate patterns, and are usefully considered separately.<br />

Cysts ICRS consensus are a difficult on animal problem models to successfully bone-graft in the<br />

hip, M.B. presumably Hurtig because of the very high pressures that develop<br />

under Guelph/<strong>Canada</strong> load compared to other joints. Bone loss can be addressed<br />

in the femoral head by mosaicplasty combined with ACI. Avascular<br />

necrosis Introduction: of the The femoral ICRS white head paper has been on recommendations treated by mesenchymal for use of<br />

stem animal cell models insertion in cartilage in France, repair and in included Tromsoe, a Norway diverse group by ACI with alone. a<br />

Our broad preferred knowledge technique of model has systems been the used use in their of a locale; plug of In autologous Japan and<br />

bone most from of Europe the greater it is not trochanter, possible either to use 15mm dogs, or in 20mm the southwestern<br />

in diameter.<br />

This USA, plug central is shaped and South to fit the America normal healthy contour goats of the and femoral sheep head are<br />

and readily then available, ACI performed while horses over the are easily top. We available have treated in some several parts<br />

patients of the USA with and femoral Europe. head Using chondral an or iterative osteochondral consensus defects building of six<br />

cm approach square agreement average area. was In achieved all cases about surgical the relative dislocation strengths of the and hip<br />

was weaknesses necessary of for the surgery. model systems. We have observed some excellent results<br />

(Akimau, 2006) but in our experience patients with cyst formation<br />

do Also, not we respond found that as well. events Initially in the we orthopaedic used the ipsilateral industry was knee driving as a<br />

source changes of in cartilage the research for culture environment. but in later Regulatory cases agency used the guidelines hip. The<br />

normal are a moving knee target will have as higher symptoms standards for for several repair and months replacement following of<br />

biopsy, articular but surfaces this does are imposed. not hinder Part the of rehabilitation this is due to of the the early hip. failure These<br />

hips of prostheses have been which treated has between triggered 2 and government-led 10 years ago. investigations<br />

Five patients<br />

have and class progressed action to suits hip replacement in several countries. relatively quickly. Amalgamation We believe and<br />

this restructuring is partly of due large to the pharmaceutical high levels and of pain-free biotechnology function companies that hip<br />

replacement in the last few can years offer constitutes the younger a contraction patient. All of these the Research patients had and<br />

cyst Development formation industry, pre-operatively triggering and concerns we therefore that there caution are against fewer<br />

undertaking entities capable ACI in of the making presence the of multi-million cyst formation. dollar We investment do not believe to<br />

it bring is technically a new product feasible to market. to perform Reduced sutured risk taking ACI in and the emphasis acetabulum on<br />

by small open changes surgery. to existing The acetabulum products seems is however to be favored ideally strategies. treated by<br />

arthroscopic ACI. Fontana et al reported in 2011 to have good results<br />

with Content: arthroscopic Our mandate matrix was induced to establish autologous a consensus in 15 patients. for In animal their<br />

study models 15 studies patients and with promote an average more logical 2cm acetabular research methods. defect had To do an<br />

improvement this we initiated of over a review 40 points of models in the and Harris strategic Hip score steps with that a 6 led year to<br />

follow-up. successful A registration similar group of new of patients therapies. treated Though with the debridement success of these had<br />

no models significant was unassailable, improvement failures in symptoms. were equally This group instructive were because strongly<br />

of they the demonstrated opinion that a non-sutured the need for matrix easily would overlooked not stay components<br />

in place on a<br />

defect of a successful of the femoral project head. including: In conclusion adequate it would integrative appear that repair, ACI is<br />

a reliable possible fixation, option for accurate femoral clinically-relevant chondral and osteochondral imaging and defects interim by<br />

open assessments procedure, in long and term arthroscopic studies ACI including a good arthroscopic option for acetabular biopsies,<br />

defects. etc.. It was our hope that logical recommendations might stop the<br />

wastefulness that has been pervasive in the cartilage R&D culture.<br />

A good example of this is the still-widespread use of immature rabbits<br />

References:<br />

for cartilage repair experiments, which due to their brisk intrinsic<br />

AKIMAU,<br />

repair response<br />

P, BHOSALE,<br />

have little<br />

A,<br />

predictive<br />

HARRISON,<br />

value<br />

P.E.<br />

for<br />

ROBERTS,<br />

patients. Investors<br />

S, MCCALL,<br />

and<br />

I.W.,<br />

others<br />

JB<br />

outside<br />

RICHARDSON,<br />

our industry<br />

J.B. ASHTON<br />

could be easily<br />

B.A. Autologous<br />

misled by such<br />

chondrocyte<br />

studies.<br />

implantation<br />

Though many<br />

with<br />

examples<br />

bone grafting<br />

exist, Wakitani<br />

for osteochondral<br />

et al in 1994<br />

defect<br />

and Grande<br />

due to<br />

posttraumatic<br />

et al (1995) demonstrated<br />

osteonecrosis<br />

that<br />

of the<br />

adherent<br />

hip. Acta<br />

cells<br />

Orthopaedica.<br />

from bone marrow<br />

2006;<br />

77<br />

in collagen<br />

(2): 333-336.<br />

1 gel or polyglycolic acid could restore large defects<br />

in the rabbit by 24 weeks. Recently, Chang (2011) demonstrated<br />

GOKHALE<br />

that empty<br />

S,<br />

PLGA<br />

KHAN<br />

scaffolds<br />

M, KUIPER<br />

with<br />

JH,<br />

continuous<br />

RICHARDSON<br />

passive<br />

JB, DAVIES<br />

motion<br />

JP<br />

were<br />

An<br />

arthroscopic<br />

equally successful.<br />

hip documentation<br />

If near perfect<br />

form<br />

repair<br />

Arthroscopy.<br />

in rabbit cartilage<br />

2008; 24<br />

can<br />

(7):<br />

be<br />

839<br />

driven<br />

<strong>–</strong> 842.<br />

by number of conditions, rather than completely condemn<br />

the use of laboratory animals, we adopted guidelines for maturity<br />

KHAN that were M, EDWARDS not based D, on RICHARDSON chronological JB Birmingham age or skeletal Hip development.<br />

Arthroplasty:<br />

Five A fully to Eight mature Years “cartilage Prospective organ” Multicentre should contain: Centre zonal Results distribution Journal of<br />

Arthroplasty. of chondrocytes, 2009; a 24(7): continuous 1044 <strong>–</strong> tidemark 1050. H J with SMITH, a calcified JB RICHARDSON, cartilage<br />

A layer, TENNANT and regional Modification specialization and validation of biochemical of the Lysholm and biomechanical<br />

Knee Scale<br />

to properties. assess articular In the rabbit, cartilage Wei damage. and Messners Osteoarthritis (1995, 1997) and <strong>Cartilage</strong>. excellent<br />

2009; work demonstrated 17:53-58. age-related repair in rabbits is substantially<br />

different than larger species until rabbits are 8 months old. Thus,<br />

Fontana, since most A., workers Bistolfi, are Crova, unable M., Rosso, or unwilling F., Massazza, to obtain G. rabbits Arthroscopic of this<br />

Treatment age, the utility of of Hip this Chondral model system Defects: is limited Autologous to use as Chondrocyte<br />

a screening<br />

Transplantation tool in preliminary Versus studies. Simple Our Debridement—A society should encourage Pilot Study our peers,<br />

reviewers and editors to combine preliminary studies in rabbits or<br />

laboratory animals with a large animal study for publication.<br />

Acknowledgments:<br />

Due to the concern about durability and maturation of the repair<br />

With tissue many or neocartilage, thanks to Brian long Ashton, term studies Paul Harrison, of a year Sally or more Roberts, in two Jan<br />

Herman species are Kuiper, recommended. Iain McCall and Though all in Good the Oscell Laboratory team ()<br />

Practices are<br />

not absolutely required, most regulatory authorities require near-<br />

GLP conditions with a gap analysis for cartilage repair studies. The<br />

cost of such studies is substantial and time consuming, since larger<br />

species require specialized care and housing conditions. Adequate<br />

statistical power will require groups of 10 or more per variable<br />

with equivalent controls. Bilateral treatments are not allowed in<br />

many jurisdictions but have the advantage of controlling for interanimal<br />

variability in repair response. Unilateral models provide<br />

more opportunity for controlled weight bearing and assessment of<br />

lameness. The consensus document reviews the evidence-based<br />

medicine around use of the larger species since the mini-pig, sheep,<br />

goat and horse have all contributed to new product registrations. The<br />

thick cartilage of the horse, suitability for arthroscopic procedures<br />

including interim biopsies, growing availability of biomarkers and<br />

reagents and similarity to the cartilage volume of the human knee


have made this an attractive model for those who have specialized<br />

facilities, trained personnel and approval from animal use<br />

committees. Despite the relatively thin cartilage of the sheep and<br />

goat, which makes fixation of scaffolds and retention of implanted<br />

cells or tissue more difficult, experience and expertise can overcome<br />

these obstacles. The incidence of subchondral cysts and persistent<br />

remodeling after deep subchondral bone drilling or microfracture in<br />

the sheep, goat and horse remains enigmatic and can interfere with<br />

assessment of cartilage repair (Orth et al 2012). Unfortunately none<br />

of the larger species can simulate the extremely thin subchondral<br />

bone plate and low trabecular volume of the metaphysis in the<br />

human knee.<br />

The consensus document also contains comprehensive<br />

recommendations for outcome measures. Though histology at<br />

the time of sacrifice remains the gold standard of cartilage repair<br />

studies, functional imaging at interim time points and biopsies to<br />

allow biochemical, immunohistology and mechanical testing can<br />

play important roles in long term studies.<br />

The cost of bringing new cartilage repair products to market is<br />

daunting and in our present world economy demands efficiencies.<br />

The consensus document represents our current understanding<br />

and should guide us away from wasteful inconclusive studies and<br />

towards progress and regulatory approvals.<br />

References:<br />

Hurtig M, et al. Pre-clinical studies for cartilage repair:<br />

Recommendations from the international cartilage repair society.<br />

<strong>Cartilage</strong> 2011 2: 137-153.<br />

Wakitani S, et al. Mesenchymal cell-based repair of large, fullthickness<br />

defects of articular; cartilage. J Bone Joint Surg Am. 1994<br />

Apr;76(4):579-92.<br />

Grande DA, et al. <strong>Repair</strong> of articular cartilage defects using<br />

mesenchymal stem cells. Tissue Eng. 1995;1(4):345-53.<br />

Wei XC, et al. Maturation-dependent durability of spontaneous<br />

cartilage repair in rabbit knee joint. J Biomed Mater Res. 1999;46:539-<br />

548.<br />

Wei XC, et al. Maturation-dependent repair of untreated<br />

osteochondral defects in the rabbit knee joint. J Biomed Mater Res.<br />

1997;34:63-72.<br />

Orth P, et al. Effect of subchondral drilling on the microarchitecture<br />

of subchondral bone: Analysis in a Large Animal Model at 6 Months.<br />

Am J Sports Med.2012 Jan 5.<br />

Acknowledgments:<br />

Michael D. Buschmann, Lisa A. Fortier, Caroline D. Hoemann, Ernst B.<br />

Hunziker, Jukka S. Jurvelin, Pierre Mainil-Varlet, C. Wayne McIlwraith,<br />

Robert L. Sah, Robert A. Whiteside.<br />

Extended Abstracts 177<br />

24.2.2<br />

Rabbit cartilage repair models: a review<br />

C.D. Hoemann<br />

<strong>Montreal</strong>/<strong>Canada</strong><br />

Introduction: The rabbit has both promising and limiting features as an<br />

orthopedic model for cartilage repair. Rabbit articular cartilage is ~10x<br />

thinner than human, and osteochondral defects repair quite rapidly,<br />

~3 to 5-fold faster than large animals and humans. The fast repair<br />

rate accelerates hypothesis testing and formulation screening, which<br />

is economical, but potentially raises other challenges in scale-up to<br />

large animals. Adolescent rabbits (3-6 months old) have a high rate of<br />

spontaneous osteochondral repair. Acute defects in otherwise healthy<br />

rabbits (and all other species) do not mimic the clinical condition where<br />

cartilage lesions have variable cartilage degradation, subchondral bone<br />

changes, chronicity, and concomitant factors (ACL/meniscal tear, knee<br />

mal-alignment), in patients with variable health (BMI, age, smoking,<br />

medications, prior trauma, osteoporosis, activity level) and mental<br />

status (compliance, rehab). Yet since the 1970’s, significant progress has<br />

been made in cartilage repair therapies through data generated in rabbit<br />

models. This paper looks into the historical use of rabbit preclinical<br />

models.<br />

Content: Methods A literature review was carried out on rabbit cartilage<br />

repair studies, with particular attention to models that supported the<br />

use of new clinical treatments. Experiments dating from the 1970’s were<br />

compared for rabbit age, number, method of surgical defect creation<br />

and defect site, treatment used, timepoints analyzed, methodologies<br />

used for quantitative outcome measures. Evolving methodologies<br />

over the past 50 years were noted. Results In the 1970’s, rabbits were<br />

a testing ground for analyzing surgical procedures already being used<br />

in the clinic including drilling, shaving, and abrasion. Studies in rabbits<br />

confirmed that partial thickness lesions fail to heal spontaneously,<br />

that debridement alone is insufficient to stimulate cartilage repair,<br />

and that subchondral drilling elicits a repair tissue that grew out of the<br />

bone marrow, “resurfaced” the lesion and contained variable levels<br />

of glycosaminoglycan 2-4. In 1975 it was first reported that a sutured<br />

patch of ear-derived perichondrium with the cambium layer facing out<br />

produced a cartilage-like tissue 4 to 8 weeks post-surgery in a rabbit<br />

cartilage defect 5. In the early 1980’s biochemical assays showed that<br />

bone marrow-derived tissue outgrowth is initially 60% collagen type<br />

I and 40% collagen type II 6. Researchers began to experiment with<br />

periosteal grafts, post-operative joint immobilization and continuous<br />

passive motion (CPM) 7, 8. Some of these early rabbit studies<br />

employed N=50 to hundreds of rabbits, sometimes with up to 20%<br />

of operated rabbits having a serious adverse event (death, infection,<br />

condylar fracture, patellar dislocation)6-8. In the late 80’s, the first<br />

patient was treated with in vitro passaged autologous chondrocytes<br />

injected below a periosteal graft (ACI) 9, and the first rabbit study<br />

using autologous chondrocytes and a periosteal patch to treat patellar<br />

defects was also published, with a 6-week endpoint 10. In the 1990’s<br />

many studies were still using immature rabbits to analyze the effect<br />

of periosteal grafts, ACI, bone marrow-derived mesenchymal stem<br />

cells, fibrin glue, and various biomaterial and synthetic carriers11-14.<br />

However, in osteochondral defects with no further treatment, a hyalinelike<br />

tissue could be consistently elicited in 3-month old rabbits, and<br />

much less frequently, in 8-month-old rabbits, up to 48 weeks postoperative<br />

15. A time series of marrow-derived repair (3 mm diameter,<br />

3 mm deep drill holes) showed that very good tissues developed at 4<br />

to 8 weeks that subsequently degenerated at 1 year post-surgery16.<br />

Occasional outliers with excellent repair at 1 year were also seen to<br />

arise from drilling and no further treatment16. These observations are<br />

very important because it suggests that if enough adolescent rabbits<br />

are included in a study, with defects that penetrate the subchondral<br />

bone, at least one excellent repair is likely to be observed, especially<br />

if a biological treatment is ineffective or the implant is not retained.<br />

The first quantitative histology scoring methods were developed in the<br />

late 1980’s and 90’s and are still used today 8, 17. Early timepoints<br />

to verify residency of delivered cells or tissues were still lacking. By<br />

the 2000’s skeletal maturity began to be recognized as an important<br />

feature for extrapolating to the human condition. Rabbit periosteum<br />

was found to thin out and have a dramatic loss of stem cell precursors<br />

with aging 18. In a rabbit model with free caged activity, the periosteal<br />

patch was retained in most defects for 4 days but rarely after 7<br />

days19. Many more scaffolds were being tested, as well as cytokines<br />

and different cell sources 20-22. One-shot growth factor delivery<br />

previously optimized in vitro with cultured cells was giving negative<br />

results in vivo in some rabbit models23. Experiments with mini-pumps<br />

to deliver recombinant human FGF2 to the synovial cavity in adolescent<br />

rabbits showed promising early repair that was quite heterogeneous<br />

24. Data began to hint at pragmatics involved in developing biologics<br />

with rabbit preclinical models, where xenogenic formulations were<br />

being tested with unknown cross-reactivity. Other scaffolds began


178 174<br />

24.1.2<br />

Extended Abstracts<br />

to be used in the clinic (HYAFF, collagen matrices ChondroGuide and<br />

MACI, PLGA acellular plugs, alginate, BST-CarGel, carbon fiber pads),<br />

without ACI in the or hip with joint cells (ACI, characterized chondrocyte implantation or<br />

CCI, J.B. Richardson<br />

bone marrow-derived mesenchymal cells) and biologics (plateletrich<br />

Oswestry/United plasma, PRP). Kingdom All of these treatments used rabbit models as their<br />

testing ground10, 12-14, 20, 22, 25-28. In 2007, the FDA recognized<br />

the Introduction: lack of regulation Autologous and emitted chondrocyte draft implantation guidelines that (ACI) are has now been final<br />

for used preclinical most commonly models leading as a treatment to new cartilage for cartilage repair defects products, in and the<br />

indicated knee and that there rabbits are few were studies useful of for its formulation use in other screening joints. Can 29, ACI 30.<br />

Rabbit work in studies the hip? started We have using used micro-computed hip arthroscopy tomography for over 15 to years analyze and<br />

subchondral identified a significant bone below proportion repairing rabbit of patients cartilage-bone with chondral defects defects 31-33<br />

Since but relatively 2010, it normal was shown radiology. that augmented Increasing microfracture use of hip arthroscopy treatments<br />

that will identify displace more chondrogenic patients foci with close localised to the chondral subchondral defects bone of plate both<br />

give the femoral a better head hyaline and quality the acetabulum. of cartilage repair Can these tissue problems at later time be<br />

points treated 34. by autologous Osteoclasts chondrocyte were implicated implantation? in marrow-derived cartilage<br />

repair 35, and reasonable models of microfracture were attained in<br />

mature Content: rabbits36. Assessing Geriatric chondral rabbits defects (24-months in the hip requires old) were assessment shown to<br />

have of loss a of further function, dampened accurate response mapping to of marrow size and stimulation location of compared defects,<br />

to and adult identification rabbits (12-months of predisposinbg old)35 factors. confirming These the three importance dimensions of<br />

this are useful age dependence. for evaluation New of approaches any joint with to a stimulate chondral subchondral<br />

defect. We<br />

repair consider using the biologics, development different of arthritis cell types, to be and a new process solid of scaffolds loss of<br />

and function carriers of the have joint been as tried. an organ. In response Thus there to the is FDA not guidelines, one particular the<br />

ICRS point mandated at which a working joint becomes groups ‘arthritic’, to write recommendation but a gradual process papers that for<br />

preclinical develops over models time. and Some histological of this outcome process measures is pre-clinical 37, 38. and These thus<br />

recently joint narrowing published or guidelines osteophyte re-affirmed formation the often importance develops of prior skeletal to<br />

maturity symptoms. in These preclinical pre-clinical models, features although can some be assessed teams are by still imaging, using<br />

immature but may not rabbits. lead to Immature treatment. rabbits We propose could be chondral still potentially defects to useful be part in<br />

some of this types process, of studies and also looking are not at acute always implant symptomatic. residency. Indeed Conclusions many<br />

Rabbit heal with models repair are tissue, useful as for is seen proof-of-concept in a full thickness data and ACI as harvest a stepping site.<br />

stone This new to large tissue animal we propose pivotal studies. to call novocartilage. The overlying recurrent It is characterised outcome<br />

of by rabbit being studies well attached is the heterogeneous to underlying repair bone, response. to have type There 2 is collagen still no<br />

clear and variable explanation proteoglycan for animal-to-animal content but variation. has no ingrowth Bilateral of models vessels with or<br />

head-to-head nerves. This is comparison a true repair of treatment tissue which versus may control remodell defect to articular can use<br />

fewer cartilage. animal It may numbers have due differing to higher amounts power of hyaline multivariate characteristics and paired<br />

statistics. as assessed The by high polarised rate of light spontaneous microscopy. repair We in believe adolescent this tissue animals is<br />

is an not end reproduced in itself as is in callus mature healing or geriatric a fracture rabbits. en Therefore, route to remodelled skeletally<br />

mature bone. Renovocartilage animals (i.e., ≥7 may months be sufficient old) should for purpose be used to to slsoow screen joint the<br />

efficacy function of as formulations callus can be whose sufficient indication for purpose, for use although is in adult it consists human<br />

patients. of woven Standardized bone. The scientist surgical may defects seek and regeneration outcome measures of the original could<br />

help tissue, facilitate but the objective clinician comparison may be satisfied between with studies a functioning and accelerate repair<br />

progress tissue. The in the process field. of organ failure that is seen as a chondral defect<br />

of the knee may be quantified by a self-assessed score of function for<br />

the knee that has been weighted and validated as an interval score<br />

References:<br />

(Smith et al., 2009). In the hip we advocate a modified Harris Hip<br />

score which has allowed evaluation of 5000 BHR hip resurfacings<br />

1. (Khan Masoud et al, I, Shapiro 2009). F, Mapping Kent R, Moses by a geometric A. (1986) J. projection Orthop. Res. allows 4(2):221- the<br />

231. arthoscopic site to be correlated with the MRI findings (Gokhale et al,<br />

2008). Why develop biological repair for the hip? Function following<br />

2. hip Mankin, resurfacing H. J. (1974) or hip N. replacement Engl. J. Med. 291:1285-1292<br />

can be excellent. This sets a high<br />

standard for a biological therapy. However the high function leads<br />

3. to Meachim, wear and G., eventual and Roberts, failure C. (1971) of most J Anat devices. 109:317-327 Metal-polythene hip<br />

replacements have had an excellent outcome in the older patient,<br />

4. but Mitchell, relatively N., and high Shepard, failure N. rates (1976) of J. around Bone Joint 12% Surg. by - 10 Am. years 58:230-233 in the<br />

5.Engkvist, under-50 year-olds. O., Johansson, Higher S. H., rates Ohlsen, of L., failure and Skoog, in osteoarthritis T. (1975) Scand. leave J.<br />

Plast. only 50% Reconstr. surviving Surg. 9:203-206 by 20 years. A patient having hip replacement<br />

for osteoarthritis at the age of 30 is likely to have several revisions<br />

6. over Furukawa, the next T., 50 Eyre, years, D. and R., Koide, each S., revision and Glimcher, is marked M. by J. (1980) loss of J. bone. Bone<br />

Joint Metal-on-metal Surg.- Am. 62:79-89 bearings were re-introduced as a solution for hip<br />

resurfacing and here for most designs have been very successful<br />

7. in Amiel, the young D., Coutts, active R. male D., Abel, patients. M., Stewart, We have W., Harwood, identified F., femoral and Akeson, head<br />

W. size H. less (1985) than J. Bone 46mm Joint and Surg.- female Am. 67:911-920 gender as pre-operative factors<br />

that identify earlier failure of the implant. Stemmed metal-metal hip<br />

8. designs O’Driscoll, may S. suffer W., Keeley, from high F. W., local and levels Salter, of R. metal B. (1988) debris J. Bone as the Joint trunion Surg.-<br />

Am. is an 70:595-606 additional source of metal debris. New ceramic designs and<br />

cross-linked polythene containing vitamin E may provide improved<br />

9. solutions Brittberg, for M., the Lindahl, future, A., but Nilsson, all synthetic A., Ohlsson, implants C., Isaksson, carry a O., risk and of<br />

Peterson, infection that L. (1994) is difficult N. Engl. to J. eradicate. Med. 331:889-895 There is therefore an argument<br />

for developing biological reconstructions that maintain bone stock<br />

10. and Grande, avoid D. the A., problems Pitman, M. of I., synthetic Peterson, bearing L., Menche, surfaces. D., and The Klein, cost M.<br />

(1989) that can J. Orthop. be supported Res. 7:208-218 can be considered not just the cost of a joint<br />

replacement but the cost of a difficult revision post-phoned in later<br />

11. life. Moran, This life-time M. E., Kim, perspective H. K. W., and is Salter, similar R. for B. (1992) the hip, J. Bone ankle Joint and Surg.-Br. knee. A<br />

7:659-667 surgeon’s perspective may focus on the duration of survival for each<br />

implant, but the patient generally seeks high levels of function. This<br />

12. supports Wakitani, a routine S., Goto, self-assessed T., Pineda, S. J., measure Young, R. of G., function Mansour, of the J. M., affected Caplan,<br />

A. joint I., and at every Goldberg, clinic V. with M. (1994) ‘hot-audit’ J. Bone providing Joint Surg.- a chart Am. 76A:579-592<br />

for each patient<br />

in the clinic. Renovo-cartilage: an end in itself? 1. Pain relief which<br />

13. should Brittberg, be prolonged M., Nilsson, to 10 A., years Lindahl, 2. Improved A., Ohlsson, function C., and measured Peterson, on L.<br />

(1996) a self-assessment Clin. Orthop. & scale Rel. Res., validated 326:270-283 for chondral defects (Sith et al.,<br />

2009) 3. Absence of secondary changes : a) osteophyte formation,<br />

14. b) Freed, bone L. marrow E., Marquis, oedema. J. C., Nohria, 4. Preferably A., Emmanual, evidence J., Mikos, of: A. a) G., Good and<br />

Langer, attachment R. (1993) to underlying J. Biomed. Mater. bone Res. b) Lateral 27:11-23 integration c) Sufficient<br />

thickness d) Low surface friction e) Resistance to fluid movement<br />

15. In the Wei, hip X. C., the and development Messner, K. (1999) of different J. Biomed. patterns Mater. of Res. cartilage 46:539-548 loss is<br />

not 16. Shapiro, well documented. F., Koide, S., The and femoral Glimcher, head M. J. (1993) and the J. Bone acetabulum Joint Surg. appear - Am.<br />

to 75A:532-553 have separate patterns, and are usefully considered separately.<br />

Cysts are a difficult problem to successfully bone-graft in the<br />

hip, 17. Pineda, presumably S., Pollack, because A., Stevenson, of the very S., high Goldberg, pressures V., and that Caplan, develop A.<br />

under (1992) Acta load Anatomica compared 143:335-340 to other joints. Bone loss can be addressed<br />

in the femoral head by mosaicplasty combined with ACI. Avascular<br />

necrosis 18. O’Driscoll, of the S. W. femoral M., Saris, head D. B. has F., Ito, been Y., and treated Fitzimmons, by mesenchymal<br />

J. S. (2001) J.<br />

stem Orthop. cell Res. insertion 19:95-103 in France, and in Tromsoe, Norway by ACI alone.<br />

Our preferred technique has been the use of a plug of autologous<br />

bone 19. Aroen, from A., the Heir, greater S., Loken, trochanter, S., Reinholt, either F. 15mm P., and or Engebretsen, 20mm in diameter. L. (2005)<br />

This Acta Orthopaedica plug is shaped 76:220-224 to fit the normal contour of the femoral head<br />

and then ACI performed over the top. We have treated several<br />

patients 20. Grigolo, with B., femoral Roseti, L., head Fiorini, chondral M., Fini, or M., osteochondral Giavaresi, G., Nicoli defects Aldini, of six N.,<br />

cm Giardino, square R., average and Facchini, area. A. In (2001) all cases Biomaterials surgical 22:2417-2424<br />

dislocation of the hip<br />

was necessary for surgery. We have observed some excellent results<br />

(Akimau, 21. Hoemann, 2006) C. D., but Sun, in our J., Legare, experience A., McKee, patients M. D., with and cyst Buschmann, formation M.<br />

do D. (2005) not respond Osteoarthritis as well. <strong>Cartilage</strong> Initially 13:318-329 we used the ipsilateral knee as a<br />

source of cartilage for culture but in later cases used the hip. The<br />

normal 22. Hoemann, knee C. will D., have Sun, J., symptoms McKee, M. for D., Chevrier, several A., months Rossomacha, following E.,<br />

biopsy, Rivard, G. but E., this Hurtig, does M., not and hinder Buschmann, the rehabilitation M. D. (2007) of the Osteoarthritis hip. These<br />

hips <strong>Cartilage</strong> have 15:78-89 been treated between 2 and 10 years ago. Five patients<br />

have progressed to hip replacement relatively quickly. We believe<br />

this 23. Holland, is partly T. A., due Bodde, to the E. W. high H., Cuijpers, levels of V. pain-free M. J. I., Baggett, function L. S., that Tabata, hip<br />

replacement Y., Mikos, A. G., can and offer Jansen, the younger J. A. (2007) patient. Osteoarthritis All these <strong>Cartilage</strong> patients 15:187- had<br />

cyst 197 formation pre-operatively and we therefore caution against<br />

undertaking ACI in the presence of cyst formation. We do not believe<br />

it<br />

24.<br />

is<br />

Yamamoto,<br />

technically<br />

T.,<br />

feasible<br />

Wakitani,<br />

to<br />

S.,<br />

perform<br />

Imoto, K.,<br />

sutured<br />

Hattori,<br />

ACI<br />

T., Nakaya,<br />

in the acetabulum<br />

H., Saito, M.,<br />

by<br />

and<br />

open<br />

Yonenobu,<br />

surgery.<br />

K. (2004)<br />

The<br />

Osteoarthritis<br />

acetabulum<br />

<strong>Cartilage</strong><br />

is however<br />

12:636-641<br />

ideally treated by<br />

arthroscopic ACI. Fontana et al reported in 2011 to have good results<br />

with<br />

25. Solchaga,<br />

arthroscopic<br />

L. A., Temenoff,<br />

matrix induced<br />

J. S., Gao,<br />

autologous<br />

J. Z., Mikos,<br />

in<br />

A.<br />

15<br />

G.,<br />

patients.<br />

Caplan, A.<br />

In<br />

I.,<br />

their<br />

and<br />

study<br />

Goldberg,<br />

15<br />

V.<br />

patients<br />

M. (2005)<br />

with<br />

Osteoarthritis<br />

an average<br />

<strong>Cartilage</strong><br />

2cm acetabular<br />

13:297-309<br />

defect had an<br />

improvement<br />

26. Fragonas, E.,<br />

of over<br />

Valente,<br />

40 points<br />

M., Pozzi-Mucelli,<br />

in the Harris<br />

M.,<br />

Hip<br />

Toffanin,<br />

score with<br />

R., Rizzo,<br />

a 6 year<br />

R.,<br />

follow-up.<br />

Silvestri, F.,<br />

A<br />

and<br />

similar<br />

Vittur,<br />

group<br />

F. (2000)<br />

of patients<br />

Biomaterials<br />

treated<br />

21:795-801<br />

with debridement had<br />

no significant improvement in symptoms. This group were strongly<br />

of 27. the Willers, opinion C., that Chen, a non-sutured J., Wood, D., matrix and would Zheng, not M. stay H. (2005) in place Tissue on a<br />

defect Engineering of the 11:1065-1076 femoral head. In conclusion it would appear that ACI is<br />

a possible option for femoral chondral and osteochondral defects by<br />

open 28. Marchand, procedure, C., Chen, and arthroscopic G., Tran-Khan, ACI N., a Sun, good J., option Chen, H., for Buschmann, acetabular<br />

defects. M. D., and Hoemann, C. D. Tissue Engineering, Pt A (in press)<br />

29. FDA, U. S. A. (2007) Guidance for Industry: Preparation of IDEs and<br />

References:<br />

INDs for Products Intended to <strong>Repair</strong> or Replace Knee <strong>Cartilage</strong>. Draft<br />

Guidance (ed., CBER, U.S. Department of Health and Human Services),<br />

AKIMAU, Rockville, MD P, BHOSALE, A, HARRISON, P.E. ROBERTS, S, MCCALL,<br />

I.W., JB RICHARDSON, J.B. ASHTON B.A. Autologous chondrocyte<br />

implantation 30. FDA, U. S. A. with (2011) bone Guidance grafting for Industry: for osteochondral Preparation of defect IDEs and due INDs to<br />

posttraumatic for Products Intended osteonecrosis to <strong>Repair</strong> or of Replace the hip. Knee Acta <strong>Cartilage</strong>. Orthopaedica. (ed., CBER, 2006; U.S.<br />

77 Department (2): 333-336. of Health and Human Services), Rockville, MD<br />

GOKHALE 31. Maehara, S, H., KHAN Sotome, M, KUIPER S., Yoshii, JH, T., RICHARDSON Torigoe, I., Kawasaki, JB, DAVIES Y., Sugata, JP An<br />

arthroscopic Y., Yuasa, M., Hirano, hip documentation M., Mochizuki, form N., Kikuchi, Arthroscopy. M., Shinomiya, 2008; 24 K., and (7):<br />

839 Okawa, <strong>–</strong> 842. A. (2010) J. Orthop. Res. 28:677-686<br />

KHAN 32. Chen, M, EDWARDS H. M., Sun, D, J., RICHARDSON Hoemann, C. JB D., Birmingham Lascau-Coman, Hip Arthroplasty:<br />

V., Wei, O. Y.,<br />

Five McKee, to Eight M. D., Years Shive, Prospective M. S., and Buschmann, Multicentre M. Centre D. (2009) Results J. Orthop. Journal Res. of<br />

Arthroplasty. 27:1432-1438 2009; 24(7): 1044 <strong>–</strong> 1050. H J SMITH, JB RICHARDSON,<br />

A TENNANT Modification and validation of the Lysholm Knee Scale<br />

to 33. assess Marchand, articular C., Chen, cartilage H. M., damage. Buschmann, Osteoarthritis M. D., and Hoemann, and <strong>Cartilage</strong>. C. D.<br />

2009; (2011) Tissue 17:53-58. Engineering Part C-Methods 17:475-484<br />

Fontana, 34. Chevrier, A., Bistolfi, A., Hoemann, Crova, C. M., D., Rosso, Sun, J., F., and Massazza, Buschmann, G. Arthroscopic<br />

M. D. (2011)<br />

Treatment Osteoarthritis of <strong>Cartilage</strong> Hip Chondral 19:136-144 Defects: Autologous Chondrocyte<br />

Transplantation Versus Simple Debridement—A Pilot Study<br />

35. Chen, G., Sun, J., Lascau-Coman, V., Chevrier, A., Marchand, C., and<br />

Hoemann, C. D. (2011) <strong>Cartilage</strong> 2:173-185<br />

Acknowledgments:<br />

36. Chen, H., Hoemann, C. D., Sun, J., Chevrier, A., McKee, M. D., Shive, M.<br />

With S., Hurtig, many M., thanks and Buschmann, to Brian Ashton, M. D. (2011) Paul J. Harrison, Orthop. Res. Sally 29:1178-1184 Roberts, Jan<br />

Herman Kuiper, Iain McCall and all in the Oscell team ()<br />

37. Hoemann, C. D., Kandel, R., Roberts, S., Saris, D. B. F., Creemers, L.,<br />

Mainil-Varlet, P., Methot, S., Hollander, A. P., and Buschmann, M. D. (2011)<br />

<strong>Cartilage</strong> 2:153-172<br />

38. Hurtig, M. B., Buschmann, M. D., Fortier, L. A., Hoemann, C. D.,<br />

Hunziker, E. B., Jurvelin, J. S., Mainil-Varlet, P., McIlwraith, C. W., Sah, R. L.,<br />

and Whiteside, R. A. (2011) <strong>Cartilage</strong> 2:137-152<br />

Acknowledgments:<br />

Salary support is acknowledged from the Fonds de la Recherche sur la<br />

Santé du Quebec. I’d like to thank Julie Tremblay for helping retrieve<br />

articles and Michael Buschmann for critical reading of the paper.


24.2.3<br />

Equine models: a review<br />

D.D. Frisbie, W. Mcilwraith<br />

Fort Collins/United States of America<br />

Introduction: Articular cartilage injuries of the knee and ankle are<br />

common and a number of different methods have been developed<br />

in an attempt to improve their repair. There are two clinical aims of<br />

cartilage repair the first, restoration of joint function and the second,<br />

prevention or at least delay of the onset of osteoarthritis. These goals<br />

can potentially be achieved through replacement of damaged or lost<br />

articular cartilage with tissue capable of functioning under normal<br />

physiological environments for an extended period. Further, it may not<br />

require the return or restoration of the original tissue, a goal that has<br />

eluded researchers and clinicians.1<br />

Content: The ability to screen novel procedures for human clinical use<br />

is done by preclinical studies using animal models. While a multitude<br />

of models exists, this abstract will focus on the horse as a model<br />

describing current techniques as well as the horses advantages and<br />

disadvantages. The majority of equine cartilage resurfacing models<br />

have been focused on the equine knee or “stifle” as it is commonly<br />

called. The anatomy is similar to the human with the exception of<br />

three patellar ligaments instead of one. Comparative studies have<br />

demonstrated that the horse has articular cartilage of similar thickness<br />

to that in the human knee, in fact much closer than other species<br />

commonly used in preclinical trials.2 Further, this joint in the horse<br />

is commonly clinically affected with cartilage lesions allowing equine<br />

clinicians experience with not only research but clinical outcomes<br />

as well; these experiences can aid in the interpretation of preclinical<br />

data. This clinical disease in the horse also provides similar diagnostic,<br />

surgical and follow-up methods as those utilized in many human<br />

preclinical and clinical trails. Other species used in preclinical trials<br />

do not have a similar clinically recognized cartilage disease process.<br />

Defect models in the femoropatellar, femorotibial, and tibiotalar joints<br />

have been developed. Work in defining the critical sized defect in the<br />

stifle has also been advantageous to the use of the horse. A minimum<br />

9mm diameter defect has been determined to be a critical size in<br />

the equine stifle.3 Many studies have used a 15mm diameter defect,<br />

which represent a clinically relevant size in the human knee. The ability<br />

to differentially determine the calcified cartilage and subchondral<br />

bone plate have allowed further refinement of defect creation both<br />

through open and arthroscopic methods. The thickness of the articular<br />

cartilage also allows the reliable suturing of membranes or constructs<br />

to cartilage surrounding the defect. The size of the equine joint allows<br />

multiple defects to be placed in the same joint increasing the defect<br />

numbers without additional animals being needed. As mentioned<br />

previously defects can be created and treated either using an<br />

arthrotomy or arthroscopically. This can be an advantage in replicating<br />

the final human application. A distinct disadvantage of the horse is<br />

the ability to provide compression bandaging of the stifle area as is<br />

often done in human patients. In addition, another disadvantage is the<br />

lack of a non-weight bearing period post-operatively. The latter issue<br />

can be over come based on defect location. More specifically, defects<br />

in the more distal region of the joint surface will only be loaded in<br />

more intense exercise which can be controlled based on housing and<br />

exercise protocols. For example a defect can be non-weight bearing for<br />

the most part if a distal location is chosen and the horse is confined in<br />

a stall or small area. The defect can then be reliably loaded by using a<br />

high-speed treadmill to control the exercise speed and duration. The<br />

number of horses can be kept to a minimum in many cases by using the<br />

horse as its own control as well as evaluating the outcomes at various<br />

time points (i.e., second look arthroscopies or biopsy). Performing a<br />

repair procedure in one limb can do this and using the opposite limb<br />

as a control or having two defects in the same joint allowing for the<br />

investigator to use statistical evaluations that account for inter-patient<br />

variability therefore, fewer animals are required to achieve sufficient<br />

power. The extensive number of outcome parameters that can be<br />

utilized is a significant strength of using the equine model. Because<br />

of the relatively large size of defects that can be made in the horse,<br />

more outcome parameters can be measured on each repair response<br />

than is possible in other animal models. Potential assessments include<br />

clinical examination for lameness and synovial effusion as well as<br />

response to flexion; pretreatment and post-treatment radiographs;<br />

MRI; synovial fluid and serum biomarkers; routine synovial fluid<br />

analysis; sequential arthroscopies; optical coherence tomography;<br />

gross postmortem examination; histopathological, histochemical,<br />

and immunohistological analyses; biochemical analysis for type II<br />

collagen/type I collagen as well as aggrecan and glycosaminoglycan<br />

content; real-time quantitative PCR evaluation for mRNA expression<br />

in the tissue; and biomechanical evaluation. Several studies in which<br />

biopsies were taken of repair tissue at 4 or 6 months have indicated no<br />

detrimental long-term implications for the repair tissue from biopsies.<br />

Extended Abstracts 179<br />

There is no perfect model for objectively evaluating the repair in<br />

human articular cartilage defects. However, the need for preclinical<br />

studies using animal models in evaluating a new technique for repair<br />

is important and mandated by licensing bodies. The authors believe<br />

that there has been a positive evolution of model selection from it<br />

being based on cost and convenience to more critically evaluating how<br />

well an animal model simulates the human situation. It is recognized<br />

that some laboratories are content with small animal models and<br />

that these models will continue to be used. On the other hand, it<br />

needs to be recognized that complete removal of calcified cartilage<br />

with retention of a subchondral bone plate is important to model the<br />

human clinical scenario, and this is not possible in some smaller animal<br />

models. The horse is a large animal and requires special animal care<br />

capabilities as well as expertise. However, because of the ability to do<br />

follow-up arthroscopic evaluations, the increased amount of tissue<br />

for evaluation, and the fewer number of animals to have sufficient<br />

statistical power, the costs are necessarily disparate from studies<br />

with smaller laboratory animals. It is also important to recognize the<br />

need for long-term studies because of experience with failure between<br />

8 and 12 months, both in quality of the tissue as well as integration.<br />

It should also be remembered that one should strive for the closest<br />

approximation between preclinical research results in a given model<br />

and its extrapolation to the human situation and that the horse itself is<br />

also an end-goal animal for potential therapeutic cartilage repair.<br />

References:<br />

1. McIlwraith CW, Fortier LA, Frisbie DD, Nixon AJ. Equine models of articular<br />

cartilage repair. <strong>Cartilage</strong> 2011;2:317-326.<br />

2. Frisbie DD, Cross MW, McIlwraith CW. A comparative study of articular<br />

cartilage thickness in the stifle of animal species used in human pre-clinical<br />

studies compared to articular cartilage thickness in the human knee. Vet<br />

Comp Orthop Traumatol 2006;19:142-6.<br />

3. Hurtig MB, Fretz PB, Doige CE, Schnurr DL. Effects of lesions size and<br />

location on equine articular cartilage repair. Can J Vet Res. 1988;52:137-46.<br />

24.3.1<br />

Does unloading of joint surfaces affect cartilage healing in<br />

patients with end-stage ankle osteoarthritis; an overview of the<br />

literature<br />

A.N. Amendola 1 , M. Nguyen 1 , C. Saltzman 2<br />

1 Iowa City/United States of America, 2 Salt Lake City, Ut/United<br />

States of America<br />

Introduction: End stage osteoarthritis (OA) is a debilitating disease<br />

which causes pain, stiffness and hinders mobility in all joints. In<br />

contrast to hip and knee osteoarthritis which is of primary origin<br />

in middle aged and elderly individuals, ankle osteoarthritis is<br />

usually occurs secondary to trauma, affecting mainly in a younger<br />

population (1). Post traumatic ankle osteoarthritis is the most<br />

common form of ankle OA, progressive and usually leads surgical<br />

methods of management (2-5). However, research related to<br />

optimizing treatment of ankle osteoarthritis remains limited. Ankle<br />

fusion and total ankle replacement have been the mainstays of<br />

treatment for end stage osteoarthritis of the ankle, but both of these<br />

options have significant limitations (6-12). Recently, joint distraction<br />

has emerged as a promising treatment for ankle osteoarthritis.<br />

Unlike fusion or replacement surgery, distraction does not “burn<br />

any bridges” and patients who fail ankle distraction can still later be<br />

eligible for arthrodesis or total ankle arthroplasty. Studies utilizing<br />

thin wire and small pin external fixators including retrospective<br />

and prospective studies from the Netherlands and recently, the<br />

United States have demonstrated significant short term follow-up<br />

improvements for patients following joint distraction surgery. Long<br />

term follow-up outcomes, however are largely unknown (13-18).<br />

Adjunctive measures such as early range of motion to improve ankle<br />

distraction efficacy, are also under investigation.<br />

Content: Overview of the Literature: Osteoarthritis and Distraction<br />

The use of distraction to treat painful arthritis of the ankle was first<br />

introduced by Judet and Judet in 1978 (20). Sixteen patients with painful<br />

ankle arthritis had hinged ankle distraction (4-8 mm of distraction with<br />

motion for 6-12 weeks). At a mean follow up of 16 months, thirteen<br />

were retrospectively reported to have improved symptoms, and eight<br />

had restoration of unlimited walking ability. A subsequent study of<br />

patients with hip arthrosis showed that articulated distraction of the<br />

hip yielded good results in 42 of 59 (71%) patients who were younger<br />

than 45 years but poor results in patients older than 45 years and


180<br />

Extended Abstracts<br />

in patients with inflammatory arthritis (19). The treatment of ankle<br />

OA with distraction, using thin wire external fixators, has been<br />

reported in both retrospective and prospective clinical series from<br />

the Netherlands (13-19). Van Valburg et al. documented that ankle<br />

distraction resulted in intermittently fluctuating joint hydrostatic<br />

pressures, (17) and that similar fluctuations in vitro resulted in<br />

increased proteoglycan synthesis in osteoarthritic cartilage (16-17). A<br />

study of ankle distraction in 11 patients with post-traumatic arthritis<br />

showed that all patients had less pain at follow-up of 20 months (13).<br />

Persistent ankle swelling and crepitus has been noted after removal<br />

of the external fixator, but average function improved after 1 year after<br />

surgery. In a more recent prospective study of 57 patients followed<br />

for an average of 2.8 years after ankle distraction, significant clinical<br />

improvement was noted in three fourths of the patients, improvement<br />

increased over time, and joint distraction had significantly better<br />

results than ankle joint debridement alone (22). A subsequent review<br />

by the same researchers at minimum 7 years of follow-up evaluation<br />

after ankle distraction for osteoarthritis showed that 16 of 22 (73%)<br />

patients had significant improvement of all clinical parameters and<br />

6 (27%) patients had failed treatment (18). Following these early<br />

papers others have published on the outcome of distraction. Tellisi<br />

et al. completed a retrospective review of 25 patients, in which they<br />

found improvement of AOFAS score and SF-36 at 30 month f/u and<br />

satisfactory results in 91% of the patients (24). Others have reported<br />

on their clinical experience as well (22,23,26) in limited numbers with<br />

similar outcomes. Distraction and joint Changes There is also effort<br />

of characterizing changes following ankle distraction using imaging.<br />

Lamm et al. conducted a preliminary MRI study to characterize joint<br />

changes following distraction (26). In more in-depth studies, Intema<br />

et al. reported on the subchondral bone changes using CT in 26<br />

patients treated with distraction with and without motion. While<br />

overall density decreased, density in cystic lesions actually increased<br />

(normalization of bone density). In addition, a correlation was found<br />

between clinical improvement and the resolution of subchondral<br />

bone cysts (25). In a subsequent study (27) on knee distraction,<br />

at one year demonstrated increase in joint space, MRI increase in<br />

cartilage thickness, increased synthesis of collagen type 2, improved<br />

function and decreased pain. This was reported recently at the AAOS<br />

2012 at 2 year follow up. Joint Motion and Distraction Joint motion has<br />

been accepted as an essential adjunctive component in the biological<br />

restoration of articular cartilage from injury. This has largely followed<br />

from previous clinical and experimental studies that confirmed the<br />

deleterious effects of prolonged immobilization on musculoskeletal<br />

tissues, including cartilage. The only study assessing the effect of<br />

motion with distraction in a clinical study is currently accepted for<br />

publication (28). A prospective RCT was carried out comparing<br />

motion with distraction versus distraction alone for a 3 month<br />

period. Thirty-six patients were followed for a minimum of 2 years.<br />

Results demonstrated significant improvement in pain and function<br />

in both groups, but greater improvement in the motion group (28).<br />

Distraction and <strong>Cartilage</strong> Regeneration In terms of regenerating<br />

cartilage in clinical patients with ankle OA, very little direct published<br />

information is available. In a rabbit model with articular injury,<br />

some direct evidence exists for cartilage healing with the effect of<br />

distraction. Kajiwara et al. demonstrated in a rabbit model that with<br />

a combination of subchondral drilling, joint motion and distraction by<br />

an articulated external fixator, repair of a fresh osteochondral defect<br />

in the weight bearing area occurred. Although distraction for 4 weeks<br />

was not a long enough period to repair the defect, distraction for 8<br />

and 12 weeks resulted in a good outcome (29). Nishino et al. applied<br />

joint distraction and motion to rabbit model articular defect, and with<br />

graduated loading improved regeneration of cartilage (30).<br />

References:<br />

1. Saltzman CL, Kamp J, Cook TA. The leather ankle lacer. Iowa Orthop<br />

J. 1995; 15: 204-8.<br />

2. Pagenstert GI, Hinterman B, Barg A, Leuman A, Valderrabano V.<br />

Realignment surgery as alternative treatment of varus and valgus ankle<br />

osteoarthritis. Clin Orthop Relat Res. 2007; 462:156-68.<br />

3. Takakura Y, Tanaka Y, Kumai T, Tamai S. Low tibial osteotomy for<br />

osteoarthritis of the ankle. Results of a new operation in 18 patients. J<br />

Bone Joint Surg Br. 1995; 77(1): 50-4.<br />

4. Tanaka Y, Takakura Y, Hayashi K. Taniguchi A, Kumai T, Sugimoto K.<br />

Low tibial osteotomy for varus-type osteoarthritis of the ankle. J Bone<br />

Joint Surg Br. 2006; 88(7): 909-13.<br />

5. Baltzer AW, Arnold JP. Bone-cartilage transplantation from the<br />

ipsilateral knee for chondral lesions of the talus. Arthroscopy. 2005;<br />

21(2):159-66.<br />

6. Glazebrook MA, Arsenault K, Dunbar M. Evidence-based classification<br />

of complications in total ankle arthroplasty. Foot Ankle Int. 2009;<br />

30(10):945-9.<br />

7. Haddad SL, Coetzee JC, Estok R, Fahrbach K, Banel D, Nalysnyk L.<br />

Intermediate and long-term outcomes of total ankle arthroplasty and<br />

ankle arthrodesis. A systematic review of the literature. J Bone Joint<br />

Surg Am. 2007; 89(9):1899-905.<br />

8. Saltzman CL, Amendola A, Anderson R, Coetzee JC, Gall RJ, Haddad<br />

SL, Herbst S, Lian G, Sanders RW, Scioli M, Younger AS. Surgeon<br />

training and complications in total ankle arthroplasty. Foot Ankle Int.<br />

2003; 24(6):514-8.<br />

9. Coester LM, Saltzman CL, Leupold J, Pontarelli W.. Long-term results<br />

following ankle arthrodesis for post-traumatic arthritis. J Bone Joint<br />

Surg Am. 2001; 83-A(2):219-28.<br />

10. Lee KB, Cho SG, Hur CL, Yoon TR. Perioperative complications of<br />

HINTEGRA total ankle replacement: Our initial 50 cases. Foot Ankle Int.<br />

2008; 29(10):978-84.<br />

11. Spirt AA, Assal M, Hansen Jr. ST. Complications and failure after total<br />

ankle arthroplasty. J Bone Joint Surg Am. 2004; 86-A(6):1172-8.<br />

12. Morrey BF, Wiedeman, Jr GP. Complications and long-term results<br />

of ankle arthrodeses following trauma. J Bone Joint Surg Am. 1980;<br />

62(5):777-84.<br />

13. van Valburg AA, vanRoermund PM, Lammens J, VanMelkebeek J,<br />

Verbout AJ, Lafeber EP, Bijlsma JW. Can Ilizarov joint distraction delay<br />

the need for an arthrodesis of the ankle? A preliminary report. J Bone<br />

Joint Surg Br. 1995; 77(5):720-5.<br />

14. van Roermund PM, Lafeber FP. Joint distraction as treatment for<br />

ankle osteoarthritis. Instr Course Lect. 1999; 48:249-54.<br />

15. Marijnissen AC, Vincken KL, Viergever MA, vanRoy HL, VanRoermund<br />

PMLefeber FP, Bijlsma JW. Ankle images digital analysis (AIDA): Digital<br />

measurement of joint space width and subchondral sclerosis on<br />

standard radiographs. Osteoarthritis <strong>Cartilage</strong>. 2001; 9(3):264-72.<br />

16. Marijinssen AC. Osteoarthritis and joint distraction: Models,<br />

mechanisms and long-term effects. PhD Thesis, Rheumatology<br />

and Cinical Immunology, University Medical Center, Utrecht, The<br />

Netherlands, 2001.<br />

17. van Valburg AA, van Roy HL, Lafeber FP, Bijlsma JW. Beneficial<br />

effects of intermittent fluid pressure of low physiological magnitude<br />

on cartilage and inflammation in osteoarthritis. An in vitro study. J<br />

Rheumatol. 1998; 25(3):515-20.<br />

18. Ploegmakers JJ, vanRoemund RP, van Melkebeek J, Lammens J,<br />

Bijlsma JW, Lafeber FP, Marijnissen AC. Prolonged clinical benefit from<br />

joint distraction in the treatment of ankle osteoarthritis. Osteoarthritis<br />

<strong>Cartilage</strong>. 2005; 13(7):582-8.<br />

19. Aldegheri, R.; Trivella, G.; and Saleh, M.: Articulated distraction<br />

of the hip. Conservative surgery for arthritis in young patients. Clin<br />

Orthop. 1994; 301:94-101.<br />

20. Judet R, Judet T. The use of a hinge distraction apparatus after<br />

arthrolysis and arthroplasty (author’s transl). Rev Chir Orthop<br />

Reparatrice Appar Mot. 1978; 64(5):353-65.<br />

21. Marijnissen AC, Van Roermund PM, Van Melkebeek J, et al. Clinical<br />

benefit of joint distraction in the treatment of severe osteoarthritis<br />

of the ankle: Proof of concept in an open prospective study and in a<br />

randomized controlled study. Arthritis Rheum. 2002; 46:2893-2902.<br />

22. Paley D, Lamm BM. Ankle joint distraction. Foot Ankle Clin. 2005;<br />

10(4):685-98, ix.<br />

23. Paley D, et al. Distraction arthroplasty of the ankle--how far can you<br />

stretch the indications? Foot Ankle Clin. 2008; 13(3):471-84, ix.<br />

24. Tellisi N, et al. Joint preservation of the osteoarthritic ankle using<br />

distraction arthroplasty. Foot Ankle Int. 2009; 30(4):318-25.<br />

25. Intema F, et al. Subchondral bone remodeling is related to<br />

clinical improvement after joint distraction in the treatment of ankle<br />

osteoarthritis. Osteoarthritis <strong>Cartilage</strong>. 2011; 19(6):668-75.<br />

26. Lamm BM, Gourdine-Shaw M. MRI evaluation of ankle distraction:<br />

A preliminary report. Clin Podiatr Med Surg. 2009; 26(2):185-91.<br />

27. Intema F, et al. Tissue structure modification in knee osteoarthritis<br />

by use of joint distraction: An open 1-year pilot study. Ann Rheum Dis.<br />

2011; 70(8):1441-6.<br />

28. Saltzman CL, et al. Prospective randomized controlled trial of motion<br />

vs. distraction in the treatment of ankle OA. Accepted for Publication, J<br />

Bone Joint Surg Am. 2012 (in Print).<br />

29. Kajiwara R, Ishida O, Kawasaki K, Adachi N, Yasunaga Y, Ochi M.<br />

Effective repair of a fresh osteochondral defect in the rabbit knee joint<br />

by articulated joint distraction following subchondral drilling. J Orthop<br />

Res. 2005; 23:909-915.<br />

30. Nishino T, Ishii T, Chang F, Yanai T, Watanabe A, Ogawa T, Mishima<br />

H, Nakai K, Ochiai N. Effect of gradual weight-bearing on regenerated<br />

articular cartilage after joint distraction and motion in a rabbit model. J<br />

Orthop Res. 2010; 28(5):600-6.


24.3.2<br />

<strong>Cartilage</strong> and subchondral bone repair by joint distraction in<br />

clinical and experimental models of joint degeneration<br />

P.M. Van Roermund<br />

Utrecht/Netherlands<br />

Introduction: Osteoarthritis is a progressive disabling joint disease and is<br />

accompanied by stiffness, crepitus and swelling of the joint. Characteristic<br />

changes in the structure of the joint are damage in the articular cartilage,<br />

changes in the subchondral bone structure, osteophyte formation, and<br />

synovial inflammation. Untill now, the multi factorial etiology and pathophysiology<br />

of such changes in OA are still unclear making osteoarthritis<br />

to a disease that still cannot be cured. Traditional options of treatment<br />

involve joint arthroplasty or arthrodesis meaning that patients will not<br />

only loose the pain but also their joint. Especially in young patients,<br />

long term effects following arthroplasty and arthrodesis may have<br />

deleterious effects such are the need for repeated revision surgery and the<br />

development of osteoarthritis in adjacent joints. Joint distraction is based<br />

on the assumption that mechanical unloading of the damaged articular<br />

surfaces in combination with intermittent intra-articular fluid pressures ma<br />

y stimulate cartilage- and subchondral bone repair. Therefore, distraction,<br />

or arthrodiastasis of an OA ankle joint or knee joint has being used as a<br />

joint salvage procedure for the patient in whom fusion or joint replacement<br />

is not appropriate.<br />

Content: Van Valburg et al reported 1the first results of OA ankle joint<br />

distraction using a distraction period of three months. Acceptance of this<br />

technique is increasing 2-6. Although the mechanism of action remains<br />

unknown, clinical studies, although limited, have resulted in significant<br />

patient benefit in the short-term and long-term treatment of arthritis in<br />

the ankle joint. 2-7 . Encouraging short term effects have been reported<br />

also following distraction of severely painful OA knee joints in relatively<br />

young patients8. Studying the possible mechanisms of joint distraction<br />

requires structural parameters representing the changes in the cartilage<br />

and peri -articular bone. Radiographs during joint loading and measuring<br />

the distance between the two cartilage-bone interfaces still serve as the<br />

gold standard to demonstrate indirectly (diminishing) cartilage thickness.<br />

An increase in joint space width has been found in a number of patients<br />

following distraction of an OA ankle joint 9 or an OA knee joints 8 Radiographs<br />

demonstrate also structural changes in surface geometry and subchondral<br />

sclerosis before, during and following joint distraction of OA ankle 10 and<br />

knee joints. 8 A decrease in peri-articular bone density have been found in<br />

high density areas following OA ankle joint distraction 9 using standardized<br />

radiographs in the AIDA (Ankle Images Digital Analysis) method. Similar<br />

findings have been reported after OA knee joint distraction using the KIDA<br />

(knee Images Digital Analysis) method .8 In addition, serial standardized<br />

CT measurements of OA ankle joints s after distraction showed an increase<br />

in density in cystic regions near the joint which strongly correlated with<br />

clinical improvement in pain and disability. 8 MRI is able to quantify cartilage<br />

and to show changes in its morphology, volume and even composition. 18<br />

Following a two months OA knee joint distraction period, MRI revealed a<br />

significant increase in cartilage thickness and a decrease of denuded bone<br />

areas8. Biomarker levels showed a trend towards increased collagen type<br />

II synthesis and a decreased breakdown. 8Further studies are needed<br />

to evaluate the exact value of MRI measurements. Unfortunately, actual<br />

repair of articular cartilage remains difficult to study in humans. Several<br />

models of inducing OA in canine or rabbit knee joints with subsequent<br />

distraction have been developed in literature such are by ACL transsection<br />

11, or in combination with total meniscectomy,12 papain injection13, , or<br />

by making osteochondral defect in the femur 14or tibia15, or by damaging<br />

the weigth bearing cartilage of both femoral condyles: the groove model.<br />

16. The characteristics of the experimentally induced canine knee joint OA<br />

in ACLT/ medial meniscectomy - and Groove model reflect greatly those<br />

of human OA making these models suitable for studying human OA.12<br />

An 8 weeks joint distraction of an OA knee joint in beagles induced by<br />

ACLT resulted in a normalization of the proteoglycan (PG) turn over of<br />

the articular cartilage directly after treatment. However, histology did not<br />

show cartilage repair11. The study was repeated using the groove model.<br />

A significant less loss of PG content and collagen damage was found in the<br />

distracted OA canine knee joints compared to the not treated OA group. In<br />

addition, both the macroscopic and histological grade of cartilage damage<br />

was found to be less in the distraction group. 17 Further research and<br />

analysis will be necessary to understand he patho-physiologcal changes<br />

which may occur in OA joints during and following joint distraction.<br />

References:<br />

1) van Valburg AA, van Roermund PM, Lammens J, van Melkebeek J, Verbout<br />

AJ, Lafeber EP, Bijlsma JW. Can Ilizarov joint distraction delay the need for<br />

an arthrodesis of the ankle? A preliminary report. J Bone Joint Surg Br. 1995<br />

Sep;77(5):720-5.<br />

Extended Abstracts 181<br />

2) Kluesner AJ, Wukich DK. Ankle arthrodiastasis. Clin Podiatr Med Surg.<br />

2009 Apr;26(2):227-44.<br />

3) Lamm BM, Gourdine-Shaw M. MRI evaluation of ankle distraction: a<br />

preliminary report<br />

Clin Podiatr Med Surg. 2009 Apr;26(2):185-91.<br />

4) Tellisi N, Fragomen AT, Kleinman D, O’Malley MJ, Rozbruch SR. Joint<br />

preservation of the osteoarthritic ankle using distraction arthroplasty.<br />

Foot Ankle Int. 2009 Apr;30(4):318-25. Erratum in: Foot Ankle Int. 2009<br />

Jun;30(6):vi<br />

5)Paley D, Lamm BM, Purohit RM, Specht SC<br />

Distraction arthroplasty of the ankle--how far can you stretch the indications?<br />

Foot Ankle Clin. 2008 Sep;13(3):471-84, ix.<br />

6) Morse KR, Flemister AS, Baumhauer JF, DiGiovanni BF Distraction<br />

arthroplasty. Foot Ankle Clin. 2007 Mar;12(1):29-39<br />

7) Chiodo CP, McGarvey W. Joint distraction for the treatment of ankle<br />

osteoarthritis.Foot Ankle Clin. 2004 Sep;9(3):541-53, ix. Review<br />

8) Intema F, Van Roermund PM, Marijnissen AC, Cotofana S, Eckstein F,<br />

Castelein RM, Bijlsma JW, Mastbergen SC, Lafeber FP Tissue structure<br />

modification in knee osteoarthritis by use of joint distraction: an open 1-year<br />

pilot study. Ann Rheum Dis. 2011 Aug;70(8):1441-6. Epub 2011 May 12.<br />

9) Marijnissen AC, Vincken KL, Viergever MA, van Roy HL, Van Roermund<br />

PM, Lafeber FP, Bijlsma JW. Ankle images digital analysis (AIDA): digital<br />

measurement of joint space width and subchondral sclerosis on standard<br />

radiographs.Osteoarthritis <strong>Cartilage</strong>. 2001 Apr;9(3):264-72<br />

10) Intema F, Thomas TP, Anderson DD, Elkins JM, Brown TD, Amendola A,<br />

Lafeber FP, Saltzman CL Subchondral bone remodeling is related to clinical<br />

improvement after joint distraction in the treatment of ankle osteoarthritis<br />

Osteoarthritis <strong>Cartilage</strong>. 2011 Jun;19(6):668-75. Epub 2011 Feb 13.<br />

11) van Valburg AA, van Roermund PM, Marijnissen AC, Wenting MJ, Verbout<br />

AJ, Lafeber FP, Bijlsma JW. Joint distraction in treatment of osteoarthritis<br />

(II): effects on cartilage in a canine model. Osteoarthritis <strong>Cartilage</strong>. 2000<br />

Jan;8(1):1-8<br />

12) Intema F, Hazewinkel HA, Gouwens D, Bijlsma JW, Weinans H, Lafeber<br />

FP, Mastbergen SC.In early OA, thinning of the subchondral plate is directly<br />

related to cartilage damage: results from a canine ACLT-meniscectomy<br />

model.Osteoarthritis <strong>Cartilage</strong>. 2010 May;18(5):691-8. Epub 2010 Feb 6<br />

13) Karadam B, Karatosun V, Murat N, Ozkal S, Gunal No beneficial effects<br />

of joint distraction on early microscopical changes in osteoarthrotic knees. A<br />

study in rabbits.Acta Orthop. 2005 Feb; 76(1):95-8<br />

14) Kajiwara R, Ishida O, Kawasaki K, Adachi N, Yasunaga Y, Ochi M Effective<br />

repair of a fresh osteochondral defect in the rabbit knee joint by articulated<br />

joint distraction following subchondral drilling. J Orthop Res. 2005 Jul;<br />

23(4):909-15.<br />

15) Yanai T, Ishii T, Chang F, Ochiai N. <strong>Repair</strong> of large full-thickness articular<br />

cartilage defects in the rabbit: the effects of joint distraction and autologous<br />

bone-marrow-derived mesenchymal cell transplantation. J Bone Joint Surg<br />

Br. 2005 May;87(5):721-9<br />

16) Mastbergen SC, Marijnissen AC, Vianen ME, van Roermund PM, Bijlsma<br />

JW, Lafeber FP The canine ‚groove‘ model of osteoarthritis is more than<br />

simply the expression of surgically applied damage. Osteoarthritis <strong>Cartilage</strong>.<br />

2006 Jan;14(1):39-46. Epub 2005 Sep 26.<br />

17) Intema F, DeGroot J, Elshof B, Vianen ME, Yocum S, Zuurmond<br />

A, Mastbergen SC, Lafeber FP The canine bilateral groove model of<br />

osteoarthritis. J Orthop Res. 2008 Nov;26(11):1471-7.<br />

18) Lamm BM, Gourdine-Shaw M. MRI evaluation of ankle distraction: a<br />

preliminary report.Clin Podiatr Med Surg. 2009 Apr;26(2):185-91<br />

Acknowledgments:<br />

• Prof. F Lafeber<br />

• Dr. ACA Marijnissen<br />

• Dr. F Intema<br />

• Dr. A van Valburg<br />

• Dr.SC Mastbergen


182<br />

This author index lists the names of all<br />

authors and co-authors of the all congress<br />

abstracts (Podium Presentations, Poster<br />

Presentations & submitted Extended<br />

Abstracts by the invited faculty). The<br />

numbers in the index refer to the final<br />

programme number and the letter “P”<br />

before the final programme number refers<br />

to the poster section.<br />

A<br />

Abarca, E.: P61<br />

Abazari, A.: P219, P226<br />

Abbushi, A.: P171<br />

Abe, S.: 11.2.7<br />

Aberl, J.: 11.2.1<br />

Abramson, S.: 25.1.5<br />

Abratte, C. M.: 25.4.5<br />

Abubacker, S.: 21.1.1<br />

Acharya, K.: 9.4.9<br />

Ackland, T. R.: 25.3.5, P95, P247<br />

Acuña, M.: 9.4.4<br />

Adachi, N.: P101<br />

Adams, Jr., S.: P34, 25.4.1<br />

Adesida, A. B.: 9.3.3, 9.4.1, P176,<br />

P180, P182, P219<br />

Adkisson, H. D.: P142<br />

Afizah, H.: P18<br />

Agar, G.: P188<br />

Ahmad, R.: P104<br />

Aicher, W. K.: 9.2.8<br />

Ajibade, D.: P109<br />

Al-abbasi, K.: P226<br />

Alba-sanchez, I.: P84<br />

Alblas, J.: 16.4.6<br />

Albrecht, C.: P80<br />

Aldrian, S.: 11.4.6, P80<br />

Alevrogiannis, S.: P111<br />

Alini, M.: 16.2.3, P147, P271<br />

Almazan, A.: 9.4.4<br />

Almqvist, K. F.: 9.2.5, 9.4.6,<br />

11.3.3, 16.2.9,<br />

19.2.2, P6, P58,<br />

P217<br />

Alorjani, M.: P87<br />

Altschuler, N.: P26<br />

Álvarez, E.: P135<br />

Amanatullah, D.: 9.2.7, P119<br />

Amendola, A. N.: 24.3.1<br />

Amiel, D.: 11.1.9<br />

Anderson, A. B.: 16.3.3<br />

Anderson, D. G.: P265<br />

Ando, W.: 25.4.9, P221<br />

Andreatta, B.: P70<br />

Annala, T.: P98<br />

Anton, M.: P147, P272<br />

Apostolidis, K.: 16.1.3<br />

Apprich, S.: 11.4.5, 19.1.1,<br />

P164, P166, P187<br />

Arévalo, F. S.: P47<br />

Arai, R.: P162<br />

Arakaki, K.: P62<br />

Arbel, R.: P52, P114, P177,<br />

P188<br />

Arbes, S.: 11.4.6<br />

Archer, C.: P147, P271<br />

Ariganello, M. B.: 9.3.6<br />

Arno, S.: 9.4.2, 25.1.5, P214<br />

Arnold, M. P.: 9.4.5, 11.4.7<br />

Arnold, R. M.: 25.2.4<br />

Ascani, C.: P267<br />

Athanasiou, K.: 9.3.2, P35, P73<br />

Au, A. Y.: 11.2.4, P212<br />

Audenaert, E. A.: 9.2.5<br />

Aviv, M.: P273<br />

B<br />

Bünger, C.: P23<br />

Baches Jorge, P.: P33<br />

Bader, R.: P131<br />

Authors‘ Index<br />

Bahrs, C.: 9.2.8<br />

Bait, C.: P263<br />

Bal, B.: P51<br />

Balakumar, B.: P38<br />

Ball, S. T.: 11.1.9<br />

Bara, J. J.: 25.4.2<br />

Baradar Khoshfetrat, A.: P54<br />

Barbero, A.: P89<br />

Bardana, D.: 11.1.6<br />

Barr, L.: P157<br />

Barron, V.: P102<br />

Barry, F.: 18.1, P102<br />

Bartels, W.: 11.4.2<br />

Barten-van Rijbroek, A. D.: P16, P25, P77<br />

Bartlett, W.: P87<br />

Baskaran, H.: 16.4.4<br />

Bassett, E.: 9.2.6<br />

Battaglia, M.: P86, P93<br />

Beaufils, P.: 11.3.2<br />

Beaule, P.: P202<br />

Becher, C.: P194<br />

Beekhuizen, M.: 11.3.5, P77, P151,<br />

P156, P198<br />

Beekman, P.: P178<br />

Beer, Y.: P188<br />

Begum, L.: P91, P117<br />

Beier, F.: 2.1.1, 25.1.6<br />

Bekkers, J. E.: 9.3.8, 11.3.5, 11.4.2,<br />

P55, P79, P151, P156<br />

Bell, C.: 9.4.2, 25.1.5, P214<br />

Bellemans, J.: 11.3.2<br />

Beltran, L.: 9.4.2<br />

Ben Shalom, N.: P215<br />

Bendele, A.: P270<br />

Benderdour, M.: 11.2.5, P130<br />

Benders, K. E.: 9.3.4, 11.1.2<br />

Benink, R.: 11.4.2<br />

Bennett, G. W.: 16.3.3<br />

Bentley, G.: P87<br />

Berbig, R.: 9.4.5<br />

Berninger, M.: P82<br />

Bernsen, M.: 16.4.5<br />

Berti, L.: P5<br />

Bhattacharjee, A.: P262<br />

Bichara, D. A.: P227, P265<br />

Bilagi, P.: P192, P193<br />

Bird, J.: P186<br />

Bisschop, A.: 9.4.7<br />

Blanchet, T.: P206<br />

Blanke, M.: P164<br />

Blati, M.: 25.1.6<br />

Blatz, B. W.: P138<br />

Blumberg, N.: P188<br />

Bodugoz Senturk, H.: 11.3.7, P112, P227<br />

Bolduc Beaudoin, S.: 9.3.6<br />

Bommer, C.: 16.2.6<br />

Bonassar, L.: 3.1.3, 11.4.9, P184,<br />

P206<br />

Bonner, K.: P142<br />

Bonner, T. F.: P76<br />

Boot, W.: 9.3.4<br />

Bos, P.: 16.4.5<br />

Boswell, S.: 15.1.1<br />

Bot, A. G.: P151, P198<br />

Bothos, J.: 16.2.9<br />

Bourin, P.: 16.1.7<br />

Bourquin, F.: P231<br />

Boux, E.: P197, P243<br />

Bradica, G.: P160<br />

Brady, K.: P258<br />

Bragdon, C. R.: P227<br />

Brage, M. E.: P9<br />

Braithwaite, G.: P112<br />

Breitenkamp, K.: P36<br />

Brenner, J.: 11.1.6<br />

Briggs, K. K.: P200, P209, P229<br />

Briggs, T.: P87<br />

Brinchmann, J. E.: 16.4.7, 25.4.3,<br />

25.4.8, P90, P254<br />

Brittberg, M.: 2.2.3, 16.2.9, P133,<br />

P143<br />

Brix, M.: P116, P166<br />

Brophy, R. H.: P230<br />

Brown, D. S.: 11.4.3, 25.3.7<br />

Brown, J.: P234<br />

Bryant, T.: P82<br />

Bryne, J. C.: 25.4.8<br />

Buckwalter, J.: 5.10<br />

Buda, R. E.: 25.3.4, P10, P44,<br />

P86, P93, P141, P232,<br />

P236<br />

Bugbee, W. D.: 11.1.9, 15.3.3, 21.3.2,<br />

P3, P4, P9<br />

Burga, R. A.: 11.3.9<br />

Buschmann, M. D.: 16.3.4, P30, P189<br />

Bussiere, C.: P100<br />

Butler, A. P.: 11.4.8<br />

Butler, R. S.: P76<br />

Byers, B. A.: 16.2.9<br />

C<br />

Cárdenas-blanco, A.: P202<br />

Caballero-Santos, R.: P107<br />

Caborn, D.: 7.2<br />

Call, G.: P18<br />

Campbell, K. A.: 9.4.2<br />

Canseco, J.: P21<br />

Caplan, A.: 16.4.4<br />

Carey, J.: P230<br />

Carli, A.: 16.3.9<br />

Caron, M. M.: 9.2.2, 11.2.3, P118,<br />

P120, P121<br />

Caroom, C.: 16.4.9<br />

Carpenter, L.: 16.4.2<br />

Carrington, R.: P87<br />

Carubbi, C.: P59<br />

Casalis, P.: P171<br />

Castiglione, E.: P160<br />

Catanzano, A.: 9.3.7<br />

Caterson, B.: 3.2.3<br />

Cavallo, C.: 25.3.4<br />

Cavallo, M.: P5, P9, P10, P86,<br />

P93, P141, P232<br />

Cazelais, P.: 16.3.9<br />

Cervellin, M.: P263<br />

Chae, B. C.: P115<br />

Chan, J.: P204<br />

Chang, W.: P63, P96<br />

Changoor, A.: 11.4.4, 25.3.3<br />

Chavez, D.: P88<br />

Chen, A. C.: 11.1.9<br />

Chen, G.: 16.3.2<br />

Chen, H.: 16.3.4, 16.3.9, P189<br />

Chen, W.: P20<br />

Chen, X.: P146<br />

Chen, S.: P145<br />

Cheriyan, T.: 11.1.8<br />

Chesnutt, J.: P85<br />

Cheverud, J. M.: 16.1.4<br />

Chevrier, A.: 16.3.2, 16.3.4, P189<br />

Chiang, H.: P63, P96, P145<br />

Chiari, C.: P116<br />

Chien, J.: P174<br />

Chilbule, S.: P38<br />

Chinitz, N.: 9.3.7<br />

Cho, J.: P53, P57<br />

Choi, B.: P49, P53, P57, P169<br />

Choi, Y.: P268<br />

Chorev, Y.: P26<br />

Chou, C.: 16.4.4<br />

Christensen, B. B.: P23<br />

Chu, C.: 25.1.8, P127<br />

Chubinskaya, S.: 2.1.3, 9.1.2, 16.1.5, P1<br />

Chung, C. B.: P43<br />

Ciemniewska-Gorzela, K.: P181, P195,<br />

P238, P249, P250<br />

Cift, H. T.: P224, P225<br />

Clarke, R.: P128<br />

Colbrunn, R. W.: P76<br />

Cole, B. J.: 2.3.1, 15.1.1, 25.3.8,<br />

P1, P142, P241<br />

Colen, S.: P213<br />

Collas, P.: 25.4.8<br />

Colliec-jouault, S.: 9.3.9, P256<br />

Colombet, P.: 11.3.2<br />

Colombini, A.: 25.4.4


Colwell, Lambrecht, C. W.: S.: 9.2.5, P43 P6, P58<br />

Conaghan, Laouar, L.: P.: 9.3.3, P78 P176, P219<br />

Concaro, Laprell, H.: S.: P143 11.3.2<br />

Condello, Lara, J.: V.: P40, P135 P177, P188<br />

Cook, Lascau-coman, J. L.: V.: P51 16.3.2, 16.3.4, 16.3.9<br />

Coolsen, Lattermann, M. M.: C.: 9.2.2, 25.2.2, P120, 25.3.2, P121<br />

Cortes, S.: P88 25.3.9, P230, P253<br />

Cortese, Laursen, F.: J.: P40, P216 P237<br />

Cotofana, Lavigne, P.: S.: 25.1.2 3.3.3, P32<br />

Coyle, Lavoie, S.: J.: P270 P32<br />

Crawford, Law, G. K.: D. C.: P219 11.4.3, P85<br />

Creemers, Le, D. Q.: L.: P23 9.3.8, 11.1.2, 11.3.5,<br />

Lecona, H.: 11.4.2, P22 P55, P77, P79,<br />

Lee, E. H.: P151, P18 P156, P198<br />

Cremers, Lee, H. H.: A.: 25.1.8, 9.4.8, 11.2.3, P127 P118,<br />

Lee, J.: P120, P115, P115 P121<br />

Croutze, Lee, S.: R.: P223 9.3.3<br />

Cruz, Lehmann, F.: L.: 9.4.4 P108<br />

Cugat, Lesoeur, R.: J.: 11.3.2 16.2.5, P103, P256,<br />

Cui, X.: 11.3.4, P257 16.4.8, P36<br />

Czücs, Lessi, G. C.: C.: P83 25.1.3<br />

Levingstone, T. J.: 11.3.8, P56<br />

Levy, d A. S.:<br />

Levy, Y.:<br />

P26<br />

P4<br />

D’lima, Liao, C.: D. D.: P63, 9.4.3, P96 11.3.4, 16.4.8,<br />

Liao, W.: P36, P96 P43, P146<br />

Dahlberg, Liekens, K.: L. E.: P58 22.2<br />

Daisuke, Li, H.: S.: P81 8.3.2<br />

Dallari, Li, T.: D.: P59, P169 P68<br />

Das, Lim, S.: 9.4.3 P180<br />

Davisson, Lin, S.: T.: P20, P270 P145<br />

De Lind, Bari, M.: C.: 11.1.8, 18.2 P23, P158<br />

De Lindahl, Coninck, A.: T.: P183 P133, P143<br />

De Lindemann, Girolamo, S.: L.: P149 25.4.4, P263<br />

De Linder-Ganz, Graaf, M.: E.: 9.4.7 P52, P76, P177, P188<br />

De Ling, Grauw, D.: J. C.: 11.3.7, 11.1.2 P112, P227<br />

De Linnenkohl, Vries - Van W.: Melle, M. P91 L.: 16.2.2, P259<br />

De Lisignoli, Windt, G.: T. S.: 9.1.1, 16.1.7 P133, P143<br />

Deberardino, Little, C. B.: T. M.: P85 22.1<br />

Decroos, Liu, C.: J. A.: P145 11.2.2, 16.2.8<br />

Dediu, Liu, H.: V.: P37 P128<br />

Deie, Liu, K.: M.: P101 P174<br />

Del Liukkonen, Piccolo, J.: N.: 19.1.3 P59, P68<br />

Della Lloyd, Valle, D. G.: C.: P241 25.3.5<br />

Desai, Lochner, P.: K.: P214 P131<br />

Desando, Lochnit, G.: G.: 25.3.4 16.1.9<br />

Desjardins, Lohmander, J.: S.: 16.3.3 19.3.1, 19.3.1<br />

Desnoyers, London, N. J.: 11.3.1, P217 11.4.4, 25.3.3<br />

Detiger, Loosli, Y.: S. E.: P70 P172<br />

Dey, Lopa, K.: S.: P50 25.4.4<br />

Dhert, Lopez-Alcorocho, W.: J.: 3.1.2, P107 9.3.4, 9.3.8,<br />

Lopez-Reyes, A.: 11.1.2, P84 11.3.5, 11.4.2,<br />

Lotz, M. K.: 16.4.6, 9.4.3, 11.3.4, P74, P79, 16.4.8,<br />

P151, 25.1.6, P156, P36, P198 P43,<br />

Dhollander, A. A.: P146 9.2.5, 11.3.3, 19.2.2,<br />

Lu, H.: P6, 11.3.9, P58, 15.2.1 P185<br />

Di Lu, Cesare, J.: P.: 9.2.7 9.2.7, P119<br />

Di Lucas, Martino, E.: A.: 15.1.2, P163 25.1.1, 25.2.9,<br />

Luciani, D.: P45, P5 P60, P65<br />

Di Luethi, Matteo, U.: B.: 9.4.5 15.1.2, 25.1.1, 25.2.9,<br />

Luginbühl, R.: P45, P70 P60<br />

Diaz Lui, J.: Romero, J.: 9.2.4, 25.4.7P83<br />

Dickinson, Łukasik, P.: S. C.: 16.4.2, P69, P190 P258<br />

Dijkstra, Lullini, G.: P.: P5 P42<br />

Dionigi, Luna-Barcenas, C.: G.: P37 P47, P61<br />

Djap, Lussier, M.: B.: 11.2.8 11.1.4<br />

Djian, P.: 11.3.2<br />

Domayer, m S.:<br />

Doornenbal, A.:<br />

Driscoll, Méthot, S.: M. D.:<br />

Duarte, Müller, S.: A.:<br />

Dubrana, Ma, C. B.: F.:<br />

Dubuc, Madaj, A.: J.:<br />

Dudzinski, Maden, M.: W.:<br />

Duffy, Madhuri, S. F.: V.:<br />

Dugard, Madonna, M. V.: N.:<br />

Durkan, Madry, H.: M. G.:<br />

Duthon, Mae, T.: V.:<br />

Dutt, Maeckelbergh, V.: L.:<br />

Maghdoori, B.:<br />

P116, P166<br />

P16<br />

25.3.3 16.4.9, P175<br />

25.2.3 P33<br />

P85 P100<br />

11.3.6 P250<br />

P249, 2.2.1 P250<br />

9.4.3 P38<br />

P40 P125<br />

15.3.1 11.4.3<br />

P207, 9.1.3 P221<br />

P38 P213<br />

P219<br />

Magnussen, R. A.: 9.1.3<br />

Mainard, D.: 16.1.6<br />

Mainil-Varlet, P.: P203<br />

Authors‘ Index 183 185<br />

Makris, e E. A.: 9.3.2, P35, P73<br />

Malda, J.: 3.1.2, 9.3.4, 11.1.2,<br />

Ebert, J.: P74, 25.3.5, P92 P94, P95,<br />

Mandelbaum, B.: P247 1.1, 1.4, P138<br />

Mandl, Ebrahimi, E.: S.: 16.2.2 P54<br />

Manferdini, Eckstein, F.: C.: 16.1.7 25.1.2<br />

Mangiapani, Eder, M.: D. S.: 25.1.7 11.2.1<br />

Manian, Egli, R.: A.: P70 P102<br />

Maniura, Eichinger, K.: M.: P48 P187<br />

Mann, Eisman, S.: J. A.: 9.3.5 P26<br />

Mansmann, El Mansouri, U.: F.: P132 11.2.5, P130<br />

Mao, Elewaut, J.: D.: 9.2.5, 9.3.1, 14.1 P6, P58<br />

Maor, Ellä, V.: G.: P97 P27<br />

Maquet, Elliott, D. V.: M.: 11.3.6 8.3.3<br />

Maréchal, Elliott, J. A.: M.: 25.2.7 P219, P226<br />

Marcacci, Elsner, J. J.: M.: 15.1.2, P52, P76, 25.1.1, P177, 25.2.9, P188<br />

Emans, P.: P37, 9.2.2, P44, 11.2.3, P45, P118, P60,<br />

P65 P120, P121, P126<br />

Marchand, Emre, T.: C.: 16.3.2 P224, P225<br />

Marcheggiani Endres, M.: Muccioli, 16.2.6, G. M.: P171 P65<br />

Mardani, English, R. M.: A.: P246 25.2.2<br />

Mardones, Erggelet, C.: R. M.: 24.1.3 25.2.3<br />

Marijnissen, Erggelet, J. D.: A.: 16.2.3 25.1.2<br />

Marlovits, Esfandiary, S.: E.: P246 11.4.5, 11.4.6, P80,<br />

Eskandarnezhad, S.: P54 P164<br />

Martínez, Esmaeili, A.: H. G.: P135 P246<br />

Martel-pelletier, Evans, C. H.: J.: 11.1.4, P154 11.2.5, 25.1.6,<br />

Evans, H.: P130 P31<br />

Martin, Evron, Z.: I.: P89 P273<br />

Martinez-Lopez, Evseenko, D.: V.: P84 25.4.1<br />

Martinez, Ewig, M.: I.: P98 P194<br />

Martinez, M. C.: P47<br />

Martinez, f V.: P47, P61, P88<br />

Masala, N.: 21.1.1<br />

Masson, Facchini, M.: A.: 9.3.9, 16.1.7, 16.2.5, 25.3.4 P103,<br />

Fahmi, H.: P256, 11.2.5, P257 25.1.6, P130<br />

Mastbergen, Fallon, M.: S.: 11.1.5, P94, P95, 15.3.2, P247 24.3.3,<br />

Fansa, A.: 25.1.2, P218, P242 P16, P25, P77,<br />

Fantasia, R.: P59 P153<br />

Masuda, Farr, J.: K.: 11.1.9 7.1, 25.3.8, P142<br />

Mateer, Feijen, J.: J. L.: 25.3.2 P42<br />

Mates, Felka, T.: A.: P13, 9.2.8 P124<br />

Matheny, Fellah, B.: L.: P200, 9.3.9, 16.2.5, P209 P103<br />

Mathieu, Ferkel, R.: C.: 16.3.2 8.1.2<br />

Matmati, Fernandes, M.: A. M.: P90 8.2.2, P113<br />

Matsiko, Fernandes, A.: J.: 11.3.8 17.4.3<br />

Matsuda, Fickert, S.: H.: P62 P108, P110<br />

Matsukawa, Filanti, M.: T.: 16.1.8 P68<br />

Matsumoto, Filardo, G.: T.: 11.1.3 15.1.2, 25.1.1, 25.2.9,<br />

Matsuno, T.: P26, 11.2.7 P44, P45, P60,<br />

Matsuo, K.: 11.1.4 P65<br />

Matsuo, Finn, M.: T.: P207 P36<br />

Matsushita, Fisher, J.: T.: 11.1.3 P71, P78<br />

Matsuzaka, Fisher, M. B.: M.: 25.1.4 8.3.3<br />

Matsuzaki, Fitzcharles, T.: E.: 11.1.3 P163<br />

Mattacola, Fitzgerald, J.: C. G.: 25.2.2, 16.1.4 25.3.2,<br />

Flanigan, D.: 25.3.9, P230 P253<br />

Matthews, Flannery, C. G.: R.: P91 21.1.3, P206<br />

Matthies, Foglarová, N.: M.: P180 P41<br />

Mattiello-Sverzut, Foldager, C. B.: A. C.: 11.1.8, 25.1.3 P23, P158<br />

Mattielo, Fong, D.: S. M.: 25.1.3, 9.3.6 P24<br />

Matuska, Fontana, A.: 11.2.9 16.3.7, P140<br />

Mauck, Forbes, R.: J. F.: 8.3.3 P219<br />

Maumus, Forriol Campos, M.: F.: 16.1.7 11.3.1, 11.4.4, 25.3.3<br />

Mcadams, Fortier, L. A.: T. R.: 11.4.9, P138 15.1.1, 16.4.3,<br />

Mcallister, D.: 25.4.1 25.4.5, P160, P241<br />

Mccarrell, Fortuna, D.: T.: 25.3.4 15.1.1<br />

Mccarthy, Fouche, N.: H.: 25.4.2, P159 P92<br />

Mccauley, Francin, P.: J. C.: P3, 16.1.6 P4, P9<br />

Mccormack, Franklin, S. P.: R.: 11.3.1, P51 11.4.4, 25.3.3<br />

Mcgann, Freymann, L. U.: E.: 16.2.6, P219, P226 25.2.3, P171,<br />

Mcilwraith, W.: P197 3.2.1, 14.2, 15.2.2,<br />

Friederich, N. F.: 24.2.3 11.4.7<br />

Mclure, Friel, N. S. A.: W.: 25.1.8 P78<br />

Medina Frisbie, D. Mckeon, D.: J. M.: 25.3.9 15.2.2, 24.2.3<br />

Mehlhorn, Frondoza, C. A. G.: T.: 11.2.4, 16.1.2 P212<br />

Meisel, Fu, F. H.: H. J.: 9.1.4 P132<br />

Melas, Fujie, H.: I.: 16.1.3 P221<br />

Melchels, Fuller, H.: F. P.: 3.1.2 P29<br />

Meller, Furman, A.: B. D.: P27 25.1.7<br />

Mennan, C.: P262<br />

Merceron, C.: 16.2.5, P103, P256,<br />

P257<br />

Merli, M.: 25.1.1<br />

Meth, g I.: P159<br />

Meyerkort, D.: P95<br />

Meza-Zepeda, Görtz, S.: L.: 25.4.8 11.1.9, P4, P9<br />

Mhanna, Gabriel, C.: R.: P48 11.2.1<br />

Mifune, Gabusi, E.: Y.: P264 16.1.7<br />

Mikkelsen, Galley, N.: T. S.: 25.4.8, P206 P254<br />

Millan, Ganey, C.: T.: P48 P132<br />

Miller, Gansbacher, S. D.: B.: P34 P272<br />

Millett, Ganster, P. M.: J.: P199 P203<br />

Min, Gao, B.: J.: P49, P2, P8, P53, P11, P57, P12, P169 P18,<br />

Minami, A.: P152 P128, P222<br />

Minas, Garcia-Campillo, T.: H.: P22 2.3.3, 25.2.5, P82<br />

Miska, Garcia Carvajal, M.: Z. Y.: P47 P64<br />

Mitchell, Garcia, Z.: J.: P230 P61<br />

Mithoefer, Garciadiego-Cazeres, K.: D.: 1.3, 13.1.1, P47 16.3.8,<br />

Garrett, R.: 21.2.2 P2, P8, P11, P12,<br />

Miyamoto, S.: P207 P128, P222<br />

Mochizuki, Gauthier, O.: S.: P221 16.2.5, P103, P257<br />

Moens, Gawlitta, K.: D.: 9.4.6 16.4.6, P74<br />

Mohtadi, Geffroy, O.: N.: 11.3.1, 16.2.5 11.4.4, 25.3.3<br />

Mojtahed Gegout-Pottie, Jaberi, P.: F.: P233 16.1.6<br />

Mojtahed Gentili, C.: Jaberi, M.: P233 P197, P243<br />

Monemjou, Geoffroy, O.: R.: 25.1.6 P103<br />

Moorman, Georgi, N.: C. T.: P85 16.2.4<br />

Moran, Gersoff, N.: W.: P91 3.3.1<br />

Morawietz, Gerwien, P.: L.: 16.2.6 P108<br />

Moreau, Getgood, A.: P32 11.1.7, 19.2.3, 25.1.9,<br />

Moreira Teixeira, L.: P42 P19, P157, P186<br />

Moretti, Geurts, B.: M.: 25.4.4, P151, P156 P89<br />

Morgan, Ghanem, C.: N.: P173 25.2.6<br />

Moriguchi, Giannini, S.: Y.: 25.4.9, 25.3.4, P221 P5, P10, P44,<br />

Moroni, L.: P102 P86, P93, P141, P232,<br />

Morrison, S.: 21.1.1 P236<br />

Mrozinski, Giannoni, P.: A. C.: 11.2.4, P150 P212<br />

Muhonen, Gigout, A.: V.: P27 P149<br />

Mulier, Gilbert, M.: F.: P213 P106, P154<br />

Mullender, Gillogly, S. D.: M.: 9.4.7 25.2.4<br />

Mullineaux, Giovarruscio, D. R.: 25.2.2 P40<br />

Muneta, Giphart, J.: T.: P179 P163<br />

Muratoglu, Gitelis, S.: O. K.: 11.3.7, P1 P112, P227<br />

Murawski, Giza, E.: C. D.: P218, 8.1.1, P134 P220, P242<br />

Murphy, Gleeson, M.: J. P.: P102 11.3.8, P56<br />

Muto, Gobbi, T.: A.: P264 16.3.5, 25.2.1, P99,<br />

Myoui, A.: 25.4.9 P139, P269<br />

Gold, G.: 25.3.8, P142<br />

Goldberg, n V. M.:<br />

Gomez-Garcia, R.:<br />

16.4.4<br />

P22<br />

Němcová, Gomez, R.: M.:<br />

Nöth, Gomoll, U.: A. H.:<br />

Nürnberger, Gong, J. P.: S.:<br />

Naczk, Gonzalez, J.: G.:<br />

Goodrich, L.:<br />

Nagaraja, Gosselin, Y.: H.:<br />

Nagura, Grad, S.: I.:<br />

Nair, Graham, P.: W.:<br />

Nakagawa, Gramani-Say, Y.: K.:<br />

Nakaji, Gramizadeh, S.: B.:<br />

Nakamura, Grande, D.: N.:<br />

Nakamura, Greene, A.: S.:<br />

Nakamura, Greve, L.: T.:<br />

Nakashima, Grigolo, B.: M.:<br />

Nakata, Grishko, K.: V.:<br />

Nanda, Grodzinsky, S.: A. J.:<br />

Nandakumar, Grogan, S. P.: A.:<br />

Nansai, Groth, T.: R.:<br />

Naranda, Grygorowicz, J.: M.:<br />

Narcisi, Grzanna, R.: M. W.:<br />

Naruse, Guehring, K.: H.:<br />

Naveen, Guevara, S.: V.:<br />

Nebbaki, Gueven, S.: S.:<br />

Nedopil, Guicheux, A.: J.:<br />

Negrin, L. L.:<br />

Nehrer, Guilak, F.: S.:<br />

Nelson, Guillaume, B.: C.:<br />

Neri, Guillen-Garcia, S.: P.:<br />

Nesic, Guillen-Vicente, D.: I.:<br />

Netter, Guillen-Vicente, P.: M.:<br />

Neuhold, Guillot, P. A.: V.:<br />

Neumann, Gupta, R. R.: A.:<br />

Nevo, Guyton, Z.: G.:<br />

P41 P47<br />

P261 19.2.1, 25.2.5, P21<br />

P80 P62, P239<br />

P22 P181, P195, P238,<br />

14.2, P249 15.2.2<br />

P230 16.3.9<br />

P264 16.2.3<br />

P38 P230<br />

P162 25.1.3<br />

P233 25.1.4<br />

25.4.9, 5.20, 9.3.7, P207, 16.2.1 P221<br />

P162 P76<br />

P162 16.4.3<br />

16.1.8 25.3.4<br />

P13, P207, P14, P221 P124<br />

9.2.8 P112<br />

P102 9.4.3, P43, P146<br />

P221 P48<br />

P249, P244 P250<br />

16.4.5, 11.2.4, P212 P150<br />

P7 P149<br />

P104, P88 P208<br />

11.2.5, P89 P130<br />

P261 9.3.9, 16.2.5, P103,<br />

16.3.6 P256, P257<br />

15.2.3, 25.1.7, 16.2.9, P52, P177 P116<br />

P85 16.1.6<br />

P107 P10<br />

P107 9.2.4, P83<br />

P107 16.1.6<br />

11.4.6 P258<br />

P147, P76 P271<br />

P215, P34 P273<br />

Neyret, P.: 9.1.3, 11.3.2, P100<br />

Neys, J.: 25.2.7<br />

Ng, T. H.: 9.3.5, P113


184<br />

H<br />

Høiby, T.: 25.4.8<br />

Habibovic, P.: P102<br />

Hackett, C.: 16.4.3<br />

Hadley, S.: 9.4.2<br />

Haile, A.: P34<br />

Hajdu, S.: 11.4.6<br />

Hakimiyan, A. A.: 16.1.5, P1<br />

Hall, M.: 9.4.2<br />

Hallinger, R.: P261<br />

Hamada, T.: 16.1.8<br />

Hambly, K.: 25.3.1, 25.3.5, P94,<br />

P95, P247, P248<br />

Hamboeck, M.: 11.4.6<br />

Hamilton, D.: P230<br />

Hamilton, W. P.: P175<br />

Haney, N. M.: 11.3.9<br />

Hansen, O. M.: P23<br />

Hansmann, D.: P131<br />

Hart, J.: P91<br />

Hart, R.: P15<br />

Hasegawa, H.: 25.4.9<br />

Hashemibeni, B.: P246<br />

Hashimoto, S.: 16.1.4<br />

Haudenschild, D.: 9.2.7, P119<br />

Hawkins, R. J.: 16.3.3<br />

Hayes, D. A.: P217<br />

Hazewinkel, H. A.: P16<br />

Heber-katz, E.: 16.1.4<br />

Hecht, J.: 9.2.7<br />

Heinecke, L. F.: 11.2.4, P212<br />

Helder, M. N.: 9.4.7, P172<br />

Helmert, B.: P108<br />

Henkelmann, R.: 16.1.2<br />

Henrotin, Y.: 11.3.6<br />

Henson, F.: 11.1.7, 25.1.9, P19,<br />

P157<br />

Herbort, M.: 17.4.2, P72<br />

Herlofsen, S. R.: 25.4.8, P90<br />

Hernandez, P.: 25.1.9<br />

Hershman, E.: P52, P76, P177, P188<br />

Herzog, M. M.: P229<br />

Higashiyama, R.: P7<br />

Hildner, F.: 11.2.1<br />

Hingsammer, A.: P167, P204<br />

Hinz, B.: 8.2.2<br />

Hiraiwa, H.: 16.1.8<br />

Hirschmüller, A.: 25.2.3<br />

Hirschmann, M. T.: 9.4.5, 11.4.7<br />

Ho, C.: P163<br />

Hoch, J. M.: 25.3.2, 25.3.9<br />

Hoemann, C. D.: 9.3.6, 16.3.2, 16.3.4,<br />

16.3.9, 24.2.2, P39,<br />

P189<br />

Hoenecke, H.: P43<br />

Hohaus, C.: P132<br />

Hollander, A. P.: 9.3.5, 16.1.1, 16.4.2,<br />

23.2, P258<br />

Holz, J.: 17.4.1<br />

Hong, E.: P119<br />

Hoogendoorn, R. J.: P172<br />

Hooper, G. J.: 11.4.8<br />

Horan, M.: P199<br />

Horie, M.: 16.4.9, P175<br />

Horton, M. T.: P3<br />

Hosiner, S.: P80<br />

Howard, J. S.: 25.2.2, 25.3.9, P253<br />

Hsieh, C.: P145<br />

Hsu, H.: 11.1.8<br />

Huang, C. C.: P109<br />

Huard, J.: P81<br />

Hubert, Z.: P175<br />

Huebner, J. L.: 25.1.7<br />

Hui, J. H.: P18<br />

Huiyin, N.: P105<br />

Hum, D.: 11.1.4<br />

Humphrey, E.: 25.4.2, P29<br />

Hunziker, E.: 11.1.1, 15.2.1, P231<br />

Hurtig, M. B.: 1.2, 11.2.2, 16.3.9,<br />

24.2.1, P234<br />

Hutmacher, D.: 3.1.1, 3.1.2, 25.4.6,<br />

P201<br />

Huysse, W. C.: P183<br />

Authors‘ Index<br />

Hwang, N. S.: P265<br />

I<br />

Iacono, F.: P65<br />

Iacono, V.: P237<br />

Ibarra, C.: 9.4.4, P22, P47, P61,<br />

P84<br />

Ibarra, J. C.: P88<br />

Ibarra, L.: P88<br />

Igarashi, T.: P152<br />

Iliopoulos, E.: 16.1.3<br />

Imabuchi, R.: P239<br />

Imade, K.: P221<br />

Imhauser, C. W.: P218<br />

Imhoff, A. B.: 16.2.9, P272<br />

Ingham, E.: P71<br />

Ingram, J.: P71<br />

Inoue, R.: 25.1.4<br />

Intema, F.: 15.3.2, 25.1.2<br />

Iosifidis, M. I.: 16.1.3<br />

Ishibashi, Y.: 25.1.4<br />

Ishiguro, N.: 16.1.8<br />

Ishizuka, S.: 16.1.8<br />

Israel, E. F.: P47<br />

Israeli, S.: P188<br />

Itoman, M.: P7<br />

Iu, J.: 11.2.2<br />

Iwamoto, M.: 2.1.2<br />

Iwasaki, N.: P152<br />

Izaguirre, A.: 9.4.4, P22, P47, P84,<br />

P88<br />

j<br />

Järvinen, E.: P27<br />

Jakob, R.: 3.3.2, P181<br />

Jakobsen, R.: 25.4.3, P254<br />

Janes, G.: P94<br />

Jansen, E. J.: P126<br />

Jansen, N. W.: 11.1.5<br />

Janssen, M. P.: P120<br />

Jay, G. D.: 21.1.1<br />

Jeon, J. E.: P201<br />

Jia, H.: 16.4.2<br />

Jiang, C.: P63, P96, P145<br />

Jiang, T.: P2, P8, P11, P12, P18,<br />

P222<br />

Jin, L.: P53, P57, P169<br />

Jin, S.: P43<br />

Johnson, W. E.: 25.4.2<br />

Johnstone, B.: 8.2.1<br />

Jomha, N. M.: 9.3.3, 9.4.1, P176,<br />

P180, P182, P219,<br />

P226<br />

Jones, L.: P34<br />

Jones, P.: P29, P31<br />

Jonitz, A.: P131<br />

Jorgensen, C.: 16.1.7<br />

Joukainen, A.: 19.1.3<br />

Junker, M.: P234<br />

Jurvelin, J. S.: 11.4.1, 19.1.3, P161<br />

k<br />

Küchler, A. M.: P90, P254<br />

Küntziger, T.: 16.4.7<br />

Kabiri, A.: P246<br />

Kafienah, W.: 9.3.5, 16.4.1, 16.4.2,<br />

P258<br />

Kalish, L.: P204<br />

Kamei, G.: P101<br />

Kamisan, N.: P104<br />

Kanamoto, T.: P207<br />

Kanaya, F.: P62<br />

Kandel, R.: 8.3.1, 11.2.2, 16.2.8<br />

Kannan, K.: P18<br />

Kaoui, N.: P55<br />

Kaplan, L. D.: P109<br />

Kapoor, M.: 11.1.4, 25.1.6<br />

Kaps, C.: 16.2.6, 25.2.3, P171,<br />

P197, P243<br />

Karas, V.: P241<br />

Karlsen, T.: 16.4.7, 25.4.3, P90<br />

Karnatzikos, G.: 16.3.5, P99, P139,<br />

P269<br />

Karperien, M.: 16.2.4, P42<br />

Karthikeyan, R.: P38<br />

Kasahara, Y.: P152<br />

Kaszkin-bettag, M.: P108<br />

Kellomäki, M.: P27<br />

Kenmoku, T.: P7<br />

Kennedy, J. G.: P218, P220, P242<br />

Kensicki, E.: P34<br />

Kerr, A.: P29, P31<br />

Kertzman, P.: P33<br />

Keshtgar, S.: P233<br />

Khan, H.: P214<br />

Khan, R. A.: P50<br />

Khanarian, N. T.: 11.3.9, 15.2.1<br />

Khanmohammadi, M.: P54<br />

Khanna, G.: P9<br />

Kik, M.: 11.1.2<br />

Kim, J.: P134<br />

Kim, M.: P49<br />

Kim, S.: P192, P193<br />

Kim, Y.: P49, P53, P57, P167,<br />

P169, P204, P268<br />

Kinds, M.: 15.3.2<br />

Kirk, S.: P1<br />

Kisiday, J. D.: 15.2.2<br />

Kita, K.: P207, P221<br />

Kitamura, N.: P62, P239<br />

Kiviranta, I.: 19.1.3, 25.3.6, P27<br />

Klapholz, Z.: 9.3.7<br />

Klein, T. J.: 8.2.3, 25.4.6, P201<br />

Kluk, H. L.: 11.3.7, P112<br />

Kobayashi, M.: P162<br />

Kobayashi, T.: P101<br />

Koczy, B.: P69<br />

Koenderink, G. H.: P172<br />

Koevoet, W.: 16.2.2, P259<br />

Koh, Y.: P268<br />

Kokkonen, H.: 19.1.3, P161<br />

Kokot, R.: P69, P190<br />

Kokubu, T.: P264<br />

Kolk, A.: 9.3.8<br />

Kon, E.: 10.4.2, 15.1.2, 25.1.1,<br />

25.2.9, P26, P44,<br />

P45, P60, P65<br />

Kongcharoensombat, W.: P101<br />

Kops, N.: 16.2.2, 16.4.5, P259<br />

Kordelle, J.: 16.1.9<br />

Koziol, K. K.: 9.3.5<br />

Kröger, H.: 19.1.3, P161<br />

Krüger, J.: 16.2.6<br />

Kragten, A. H.: P156<br />

Krasnokutsky, S.: 25.1.5<br />

Kraus, V. B.: 25.1.7<br />

Kreulen, C.: P134<br />

Kreuz, P.: 16.2.6, 17.4.4, 25.2.3<br />

Kronen, P.: P159<br />

Krstiansen, A.: P23<br />

Krueger, J.: P243<br />

Kubo, S.: 11.1.3<br />

Kuiper, J.: P125<br />

Kulmala, K.: 19.1.3<br />

Kumar, A.: 16.3.5, P99, P139,<br />

P269<br />

Kunz, M.: P106, P154<br />

Kuroda, R.: 11.1.3<br />

Kurokawa, T.: P62, P239<br />

Kuroki, H.: P162<br />

Kurosaka, M.: 11.1.3, P264<br />

Kwon, H.: P239<br />

Kwon, O.: P205, P210<br />

Kwon, S.: P205, P210<br />

Kyriakidis, A.: 16.1.3<br />

Lafantaisie-favreau, C.: P39<br />

l<br />

Lafeber, F.: 11.1.5, 15.3.2, 25.1.2,<br />

P16, P25, P77, P153<br />

Lai, S.: P174<br />

Lallemand, E.: 16.2.5, P103


Lambrecht, S.: 9.2.5, P6, P58<br />

Laouar, L.: 9.3.3, P176, P219<br />

Laprell, H.: 11.3.2<br />

Lara, J.: P135<br />

Lascau-coman, V.: 16.3.2, 16.3.4, 16.3.9<br />

Lattermann, C.: 25.2.2, 25.3.2,<br />

25.3.9, P230, P253<br />

Laursen, J.: P216<br />

Lavigne, P.: 3.3.3, P32<br />

Lavoie, J.: P32<br />

Law, G. K.: P219<br />

Le, D. Q.: P23<br />

Lecona, H.: P22<br />

Lee, E. H.: P18<br />

Lee, H. H.: 25.1.8, P127<br />

Lee, J.: P115, P115<br />

Lee, S.: P223<br />

Lehmann, L.: P108<br />

Lesoeur, J.: 16.2.5, P103, P256,<br />

P257<br />

Lessi, G. C.: 25.1.3<br />

Levingstone, T. J.: 11.3.8, P56<br />

Levy, A. S.: P26<br />

Levy, Y.: P4<br />

Liao, C.: P63, P96<br />

Liao, W.: P96<br />

Liekens, K.: P58<br />

Li, H.: P81<br />

Li, T.: P169<br />

Lim, S.: P180<br />

Lin, S.: P20, P145<br />

Lind, M.: 11.1.8, P23, P158<br />

Lindahl, A.: P133, P143<br />

Lindemann, S.: P149<br />

Linder-Ganz, E.: P52, P76, P177, P188<br />

Ling, D.: 11.3.7, P112, P227<br />

Linnenkohl, W.: P91<br />

Lisignoli, G.: 16.1.7<br />

Little, C. B.: 22.1<br />

Liu, C.: P145<br />

Liu, H.: P128<br />

Liu, K.: P174<br />

Liukkonen, J.: 19.1.3<br />

Lloyd, D. G.: 25.3.5<br />

Lochner, K.: P131<br />

Lochnit, G.: 16.1.9<br />

Lohmander, S.: 19.3.1, 19.3.1<br />

London, N. J.: P217<br />

Loosli, Y.: P70<br />

Lopa, S.: 25.4.4<br />

Lopez-Alcorocho, J.: P107<br />

Lopez-Reyes, A.: P84<br />

Lotz, M. K.: 9.4.3, 11.3.4, 16.4.8,<br />

25.1.6, P36, P43,<br />

P146<br />

Lu, H.: 11.3.9, 15.2.1<br />

Lu, J.: 9.2.7<br />

Lucas, E.: P163<br />

Luciani, D.: P5<br />

Luethi, U.: 9.4.5<br />

Luginbühl, R.: P70<br />

Lui, J.: 25.4.7<br />

Łukasik, P.: P69, P190<br />

Lullini, G.: P5<br />

Luna-Barcenas, G.: P47, P61<br />

Lussier, B.: 11.1.4<br />

m<br />

Méthot, S.: 25.3.3<br />

Müller, S.: 25.2.3<br />

Ma, C. B.: P85<br />

Madaj, A.: P250<br />

Maden, M.: 2.2.1<br />

Madhuri, V.: P38<br />

Madonna, V.: P40<br />

Madry, H.: 15.3.1<br />

Mae, T.: P207, P221<br />

Maeckelbergh, L.: P213<br />

Maghdoori, B.: P219<br />

Magnussen, R. A.: 9.1.3<br />

Mainard, D.: 16.1.6<br />

Mainil-Varlet, P.: P203<br />

Authors‘ Index 185<br />

Makris, E. A.: 9.3.2, P35, P73<br />

Malda, J.: 3.1.2, 9.3.4, 11.1.2,<br />

P74, P92<br />

Mandelbaum, B.: 1.1, 1.4, P138<br />

Mandl, E.: 16.2.2<br />

Manferdini, C.: 16.1.7<br />

Mangiapani, D. S.: 25.1.7<br />

Manian, A.: P102<br />

Maniura, K.: P48<br />

Mann, S.: 9.3.5<br />

Mansmann, U.: P132<br />

Mao, J.: 9.3.1, 14.1<br />

Maor, G.: P97<br />

Maquet, V.: 11.3.6<br />

Maréchal, M.: 25.2.7<br />

Marcacci, M.: 15.1.2, 25.1.1, 25.2.9,<br />

P37, P44, P45, P60,<br />

P65<br />

Marchand, C.: 16.3.2<br />

Marcheggiani Muccioli, G. M.: P65<br />

Mardani, M.: P246<br />

Mardones, R. M.: 24.1.3<br />

Marijnissen, A.: 25.1.2<br />

Marlovits, S.: 11.4.5, 11.4.6, P80,<br />

P164<br />

Martínez, H. G.: P135<br />

Martel-pelletier, J.: 11.1.4, 11.2.5, 25.1.6,<br />

P130<br />

Martin, I.: P89<br />

Martinez-Lopez, V.: P84<br />

Martinez, I.: P98<br />

Martinez, M. C.: P47<br />

Martinez, V.: P47, P61, P88<br />

Masala, N.: 21.1.1<br />

Masson, M.: 9.3.9, 16.2.5, P103,<br />

P256, P257<br />

Mastbergen, S.: 11.1.5, 15.3.2, 24.3.3,<br />

25.1.2, P16, P25, P77,<br />

P153<br />

Masuda, K.: 11.1.9<br />

Mateer, J. L.: 25.3.2<br />

Mates, A.: P13, P124<br />

Matheny, L.: P200, P209<br />

Mathieu, C.: 16.3.2<br />

Matmati, M.: 8.2.2, P113<br />

Matsiko, A.: 11.3.8<br />

Matsuda, H.: P62<br />

Matsukawa, T.: 16.1.8<br />

Matsumoto, T.: 11.1.3<br />

Matsuno, T.: 11.2.7<br />

Matsuo, K.: 11.1.4<br />

Matsuo, T.: P207<br />

Matsushita, T.: 11.1.3<br />

Matsuzaka, M.: 25.1.4<br />

Matsuzaki, T.: 11.1.3<br />

Mattacola, C. G.: 25.2.2, 25.3.2,<br />

25.3.9, P253<br />

Matthews, G.: P91<br />

Matthies, N.: P180<br />

Mattiello-Sverzut, A. C.: 25.1.3<br />

Mattielo, S. M.: 25.1.3, P24<br />

Matuska, A.: 11.2.9<br />

Mauck, R.: 8.3.3<br />

Maumus, M.: 16.1.7<br />

Mcadams, T. R.: P138<br />

Mcallister, D.: 25.4.1<br />

Mccarrell, T.: 15.1.1<br />

Mccarthy, H.: 25.4.2, P92<br />

Mccauley, J. C.: P3, P4, P9<br />

Mccormack, R.: 11.3.1, 11.4.4, 25.3.3<br />

Mcgann, L. E.: P219, P226<br />

Mcilwraith, W.: 3.2.1, 14.2, 15.2.2,<br />

24.2.3<br />

Mclure, S. W.: P78<br />

Medina Mckeon, J. M.: 25.3.9<br />

Mehlhorn, A. T.: 16.1.2<br />

Meisel, H. J.: P132<br />

Melas, I.: 16.1.3<br />

Melchels, F. P.: 3.1.2<br />

Meller, A.: P27<br />

Mennan, C.: P262<br />

Merceron, C.: 16.2.5, P103, P256,<br />

P257<br />

Merli, M.: 25.1.1<br />

Meth, I.: P159<br />

Meyerkort, D.: P95<br />

Meza-Zepeda, L.: 25.4.8<br />

Mhanna, R.: P48<br />

Mifune, Y.: P264<br />

Mikkelsen, T. S.: 25.4.8, P254<br />

Millan, C.: P48<br />

Miller, S. D.: P34<br />

Millett, P. J.: P199<br />

Min, B.: P49, P53, P57, P169<br />

Minami, A.: P152<br />

Minas, T.: 2.3.3, 25.2.5, P82<br />

Miska, M.: P64<br />

Mitchell, J.: P230<br />

Mithoefer, K.: 1.3, 13.1.1, 16.3.8,<br />

21.2.2<br />

Miyamoto, S.: P207<br />

Mochizuki, S.: P221<br />

Moens, K.: 9.4.6<br />

Mohtadi, N.: 11.3.1, 11.4.4, 25.3.3<br />

Mojtahed Jaberi, F.: P233<br />

Mojtahed Jaberi, M.: P233<br />

Monemjou, R.: 25.1.6<br />

Moorman, C. T.: P85<br />

Moran, N.: P91<br />

Morawietz, L.: 16.2.6<br />

Moreau, A.: P32<br />

Moreira Teixeira, L.: P42<br />

Moretti, M.: 25.4.4, P89<br />

Morgan, C.: P173<br />

Moriguchi, Y.: 25.4.9, P221<br />

Moroni, L.: P102<br />

Morrison, S.: 21.1.1<br />

Mrozinski, A. C.: 11.2.4, P212<br />

Muhonen, V.: P27<br />

Mulier, M.: P213<br />

Mullender, M.: 9.4.7<br />

Mullineaux, D. R.: 25.2.2<br />

Muneta, T.: P179<br />

Muratoglu, O. K.: 11.3.7, P112, P227<br />

Murawski, C. D.: P218, P220, P242<br />

Murphy, M.: P102<br />

Muto, T.: P264<br />

Myoui, A.: 25.4.9<br />

n<br />

Němcová, M.: P41<br />

Nöth, U.: P261<br />

Nürnberger, S.: P80<br />

Naczk, J.: P181, P195, P238,<br />

P249<br />

Nagaraja, H.: P230<br />

Nagura, I.: P264<br />

Nair, P.: P38<br />

Nakagawa, Y.: P162<br />

Nakaji, S.: 25.1.4<br />

Nakamura, N.: 25.4.9, P207, P221<br />

Nakamura, S.: P162<br />

Nakamura, T.: P162<br />

Nakashima, M.: 16.1.8<br />

Nakata, K.: P207, P221<br />

Nanda, S.: P112<br />

Nandakumar, A.: P102<br />

Nansai, R.: P221<br />

Naranda, J.: P244<br />

Narcisi, R.: 16.4.5, P150<br />

Naruse, K.: P7<br />

Naveen, S.: P104, P208<br />

Nebbaki, S.: 11.2.5, P130<br />

Nedopil, A.: P261<br />

Negrin, L. L.: 16.3.6<br />

Nehrer, S.: 15.2.3, 16.2.9, P116<br />

Nelson, B.: P85<br />

Neri, S.: P10<br />

Nesic, D.: 9.2.4, P83<br />

Netter, P.: 16.1.6<br />

Neuhold, A.: 11.4.6<br />

Neumann, A.: P147, P271<br />

Nevo, Z.: P215, P273<br />

Neyret, P.: 9.1.3, 11.3.2, P100<br />

Neys, J.: 25.2.7<br />

Ng, T. H.: 9.3.5, P113


186<br />

Nguyen, J. T.: P218<br />

Nguyen, M.: 24.3.1<br />

Nickel, J.: P106<br />

Nielsen, A. H.: P23<br />

Niemeyer, P.: 16.1.2, 16.2.3, 25.2.3,<br />

25.2.6<br />

Nierenberg, G.: P97<br />

Nishimoto, H.: P264<br />

Nishitani, K.: P162<br />

Niu, C.: P20<br />

Nixon, A. J.: 9.2.1, 11.2.6, 25.4.7,<br />

P91, P117<br />

Noël, D.: 16.1.7<br />

Nocco, E.: P188<br />

Nochi, H.: 11.2.7<br />

Novakofski, K.: 11.4.9, 16.4.3<br />

Nurmi, H.: 25.3.6<br />

Nyengaard, J. R.: P158<br />

Nygaard, J. V.: P23<br />

o<br />

O’brien, F. J.: 11.3.8, P56<br />

O’malley, M.: 25.1.8<br />

O’shaughnessey, K.: 11.2.9<br />

Ochi, M.: P101<br />

Ochs, B. G.: 9.2.8<br />

Oda, T.: 16.1.8<br />

Ofek, G.: P2, P8, P11, P12,<br />

P222<br />

Oh, H.: P53, P57, P169<br />

Ohkawa, S.: P101<br />

Ohmiya, Y.: P239<br />

Olee, T.: P146<br />

Olewinski, R.: 16.1.5, P1<br />

Olson, S. A.: 25.1.7<br />

Omachi, T.: 16.1.8<br />

Omelchenko, A.: P75<br />

Onodera, S.: P239<br />

Onodera, T.: P152<br />

Onuma, K.: P7<br />

Ophelders, D.: P118<br />

Oprenyeszk, F.: 11.3.6<br />

Ortega-sanchez, C.: P84<br />

Ortiz, A.: P22<br />

Ortolani, A.: P37<br />

Ortved, K.: 11.2.6, P117<br />

Owaidah, A.: 16.4.2<br />

Ownby, S. L.: 11.2.4, P212<br />

Ozeki, N.: P179<br />

p<br />

Pérez, F.: 9.4.4, P47, P88<br />

Paatela, T.: 25.3.6, P27<br />

Pachowksi, M.: 11.4.5<br />

Pacifici, M.: 2.1.2<br />

Pacione, C.: 16.1.5, P1<br />

Paessler, H.: 11.3.2<br />

Pagliazzi, G.: P86, P141, P236<br />

Pallante, A. L.: 11.1.9, 21.3.1<br />

Pandey, V.: 9.4.9<br />

Pang, J.: 11.1.6<br />

Pang, S.: 11.1.6<br />

Panseri, S.: P37<br />

Park, S.: 16.2.8, P49<br />

Park, Y.: P268<br />

Parker, J. C.: P125<br />

Parma, A.: P141, P232, P236<br />

Patchornik, S.: P215<br />

Patella, S.: 25.1.1, 25.2.9, P45,<br />

P60, P65<br />

Patil, A. J.: 9.3.5<br />

Pattappa, G.: 16.2.3<br />

Pattyn, C.: 9.2.5<br />

Paul, C.: 9.4.7<br />

Pauli, C.: 9.4.3, P43<br />

Payne, K. A.: 25.1.8<br />

Pearsall, A.: P13, P14, P124<br />

Pelet, S.: 11.3.1, 11.4.4, 25.3.3<br />

Pelletier, J.: 11.1.4, 11.2.5, 25.1.6,<br />

P130<br />

Pennock, A.: P199<br />

Authors‘ Index<br />

Perdisa, F.: 25.1.1, 25.2.9, P45,<br />

P60<br />

Perez, G.: 25.1.6<br />

Perez, J.: 9.2.3<br />

Perez, M.: P61<br />

Peterbauer-Scherb, A.: 11.2.1<br />

Petersen, W.: P72<br />

Petre, B.: P163<br />

Petrera, M.: 11.2.2, 16.2.8<br />

Petrigliano, F.: 25.4.1<br />

Peyrafitte, J.: 16.1.7<br />

Pfeiffer, F.: P51<br />

Philippon, M.: 16.3.1, 24.1.1, P229<br />

Piacentini, A.: 16.1.7<br />

Picard, C.: P32<br />

Picard, G.: P189<br />

Pilz, I. H.: 16.1.2<br />

Pineda, C.: P22<br />

Piontek, T.: P181, P195, P238,<br />

P249, P250<br />

Piovan, G.: P237<br />

Pishva, E.: P233<br />

Plaas, C.: P194<br />

Poddar, M.: P81<br />

Polacek, M.: P98<br />

Polak, J.: P258<br />

Pollock, R.: P87<br />

Pomerantseva, I.: 9.2.6, P265<br />

Poole, A. R.: 19.3.2<br />

Porichis, S.: 25.2.6<br />

Portron, S.: 16.2.5, P103, P256,<br />

P257<br />

Posey, K.: 9.2.7<br />

Potel, J.: P100<br />

Potter, H. G.: P160<br />

Poubelle, P. E.: P39<br />

Pourazar, A.: P246<br />

Power, J.: 11.1.7<br />

Pownder, S. L.: P160<br />

Pravda, M.: P41<br />

Presle, N.: 16.1.6<br />

Pro, S. L.: P138<br />

Prockop, D.: 16.4.9<br />

Prusinska, A.: P249<br />

Puetzer, J.: P184<br />

Pulido, P. A.: P3, P4<br />

Puvanan, K.: P105<br />

Pyda, A.: P250<br />

q<br />

Quaglia, A.: P263<br />

Quiñones-Uriostegui, I.: P22<br />

Quinn, T. M.: 8.2.2, 11.2.8, P113,<br />

P122, P123<br />

Quintin, A.: 9.2.4, P83<br />

Quirbach, S.: P187<br />

r<br />

Rackwitz, L.: P261<br />

Rahatekar, S. S.: 9.3.5<br />

Rai, M.: 16.1.4<br />

Rakhra, K.: P202<br />

Ramanujam, N.: P234<br />

Ramponi, L.: P93, P232, P236<br />

Randolph, M. A.: 9.2.6, P265<br />

Rani, N.: P59, P68<br />

Rappoport, L.: 16.1.5, P1<br />

Rasch, H.: 11.4.7<br />

Raschke, M.: P72<br />

Razzano, P.: 9.3.7<br />

Recht, M. P.: 9.4.2, 25.1.5<br />

Rederstorff, E.: 9.3.9, P256<br />

Redl, H.: 11.2.1<br />

Reed, K.: P13, P14<br />

Reeve, R. E.: P175<br />

Regatte, R.: 25.1.5<br />

Reiff, R.: 25.1.3<br />

Reischling, P.: 25.3.8, P142<br />

Reke, N.: P250<br />

Responte, D. J.: P35<br />

Restrepo, A.: 11.3.1, 11.4.4, 25.3.3,<br />

P30<br />

Richardson, J. B.: 24.1.2, P29, P31, P92,<br />

P125, P217, P262<br />

Riek, J.: P170<br />

Rios, D.: P199<br />

Rivard, G.: 16.3.2<br />

Rivera-Bermudez, M.: P206<br />

Rivera, A. L.: 16.4.4<br />

Robert, H.: P100<br />

Roberts, S.: 25.3.3, 25.4.2, P29,<br />

P31, P92, P125, P173,<br />

P262<br />

Robin, B. N.: P175<br />

Robinson, D.: P26, P215, P273<br />

Robinson, E.: P125<br />

Rodeo, S.: 15.1.3<br />

Rodkey, W. G.: P200, P209<br />

Rodrigo, J. J.: 16.3.3<br />

Rodriguez Iñigo, E.: P107<br />

Rolauffs, B.: 9.2.8<br />

Roosendaal, G.: 11.1.5, P25, P153<br />

Rosenberger, R.: P187<br />

Rosenzweig, D. H.: 8.2.2, 11.2.8, P122,<br />

P123<br />

Ross, K.: P160<br />

Rossi, J.: 11.2.6<br />

Rothdiener, M.: 9.2.8<br />

Royen Van, B.: 9.4.7<br />

Rozen, N.: P188<br />

Rudert, M.: P106, P154, P261<br />

Ruffilli, A.: P5, P10, P86, P93,<br />

P141, P232, P236<br />

Ruike, T.: 11.2.7<br />

Runco, G.: P33<br />

Rushton, N.: 11.1.7, 25.1.9, P19,<br />

P157<br />

Russo, A.: P37, P237<br />

Rusu, D.: P39<br />

Ruvalcaba, E.: P61<br />

s<br />

Słomczykowski, M.: P181<br />

Südkamp, N. P.: 16.1.2, 25.2.6<br />

Süzer, F.: 25.2.8<br />

Sabatino, M.: P89<br />

Sabbioni, G.: P68<br />

Sachot, S.: 16.2.7<br />

Safi, A.: P15<br />

Sague, J. L.: P70<br />

Sah, R. L.: 11.1.9, 21.1.2<br />

Saito, T.: P179<br />

Sakai, T.: 16.1.8<br />

Sakamoto, F. A.: P168<br />

Sakata, R.: P264<br />

Sakaue, M.: 25.4.9<br />

Salih Mohamed, K.: P102<br />

Saliken, D.: 9.4.1<br />

Salo, J.: 19.1.3, P161<br />

Saltzman, C.: 24.3.1<br />

Salzmann, G. M.: 16.2.3, 25.2.6<br />

Sampson, H.: 16.4.9, P175<br />

Samuels, J.: 25.1.5<br />

Samuelson, E.: 25.3.7<br />

Samulski, J.: 14.2<br />

Sanchez, C.: 11.3.6<br />

Sanchez, L. J.: 8.1.3<br />

Sanchez, R. M.: P47<br />

Sandell, L. J.: 16.1.4, 19.3.3<br />

Sandri, M.: P37<br />

Sanen, K.: P118, P120<br />

Sansone, V.: 25.4.4<br />

Santamaria-Olmedo, M.: P84<br />

Santoro, R.: P89<br />

Santos, E.: P107<br />

Saris, D. B.: 9.1.1, 9.3.4, 9.3.8,<br />

11.3.5, 11.4.2, P77,<br />

P79, P133, P143,<br />

P151, P156, P198<br />

Sasaki, E.: 25.1.4<br />

Sasaki, H.: 11.1.3<br />

Satake, T.: P162<br />

Satchell, P. W.: P160


Schadow, S.: 16.1.9<br />

Schattenberg, T.: P108<br />

Schepens, A.: P56<br />

Schiavinato, A.: P203<br />

Schils, J. P.: P168<br />

Schimenti, J. C.: 25.4.5<br />

Schmal, H.: 16.1.2<br />

Schmidt, T. A.: 21.1.1<br />

Schmitt, B.: 19.1.1<br />

Schnabel, L. V.: 15.1.1, 25.4.5, P160<br />

Schneider, E.: P168<br />

Schoenhuber, H.: P263<br />

Schon, B. S.: 25.4.6, P46<br />

Schon, L.: P34<br />

Schrijver, E.: 16.4.6<br />

Schrobback, K.: 25.4.6, P201<br />

Schuchman, E. H.: 16.2.7<br />

Schuster, T.: P261<br />

Schutgens, R. E.: P25<br />

Schuurman, A. H.: P55<br />

Schweitzer, M.: P202<br />

Sciarretta, F.: P66, P267<br />

Scimeca, M.: 11.2.6, P117<br />

Secretan, C. C.: 9.3.3<br />

Sekiya, I.: 16.4.9, P179<br />

Seo, H.: 9.2.3, P223<br />

Sepúlveda, J.: P135<br />

Sernik, J.: 9.3.3<br />

Serrão, P. R.: 25.1.3<br />

Serra, R.: 9.2.3<br />

Servien, E.: 9.1.3, P100<br />

Severino, N.: P33<br />

Seyhan, B.: P224, P225<br />

Sgaglione, N. A.: 9.3.7<br />

Shabshin, N.: P188<br />

Shah, N. V.: 9.3.7<br />

Shani, J.: P26<br />

Shannon, F.: P102<br />

Shelyakova, T.: P37<br />

Sherman, O.: 9.4.2<br />

Shetty, A. A.: P192, P193<br />

Shibuya, H.: P101<br />

Shih, C.: P174<br />

Shilpa, P.: P105<br />

Shimomura, K.: 25.4.9, P207, P221<br />

Shino, K.: P221<br />

Shintani, N.: P231<br />

Shirai, T.: P162<br />

Shive, M. S.: 11.3.1, 11.4.4, 21.2.3,<br />

25.3.3, P30<br />

Shnirelman, A.: P75<br />

Shterling, A.: P52, P177<br />

Siclari, A.: P197, P243<br />

Sidler, M.: P159<br />

Siebold, R.: 11.3.2<br />

Siegert, A.: 11.4.8<br />

Sieker, J.: P154<br />

Sierra-mulet, A.: 9.4.1, P180, P182<br />

Sierra, A.: P176<br />

Sierra, L.: 9.4.4<br />

Siffri, P. C.: 16.3.3<br />

Signorile, L.: P150, P150<br />

Simões, A.: P33<br />

Simental, M.: P135<br />

Simonaro, C. M.: 16.2.7<br />

Singh, N.: 9.3.5<br />

Sinquin, C.: P256<br />

Siston, R.: P230<br />

Skarpas, G.: P111<br />

Skinner, J.: P87<br />

Small, K.: 25.2.5<br />

Smit, T. H.: 9.4.7, P172<br />

Smyth, N.: P242<br />

Sobol, E.: P75<br />

Sodha, S.: 25.2.5<br />

Sokolowski, M.: P166<br />

Solar-Cafaggi, S.: P122<br />

Solis-Arrieta, L.: P22, P47, P61<br />

Sommerfeldt, M.: P176<br />

Son, Y.: P115<br />

Song, L.: P270<br />

Soto, A.: P135<br />

Sourice, S.: 9.3.9, 16.2.5, P256,<br />

P257<br />

Authors‘ Index 187<br />

Souza, M. C.: P24<br />

Sovani, S.: P146<br />

Spalding, T.: 2.3.2, 16.2.9, P186<br />

Sparks, H.: P91<br />

Spector, M.: 11.1.8, P158<br />

Speirs, A. D.: P202<br />

Spencer, S.: P186<br />

Squillace, D. M.: P2, P8, P11, P12,<br />

P222<br />

St-Arnaud, R.: 25.1.6<br />

Stagni, C.: P59, P68<br />

Stanish, W. D.: 11.3.1, 11.4.4, 16.3.9,<br />

25.3.3<br />

Steadman, J.: P200, P209<br />

Steinert, A. F.: P106, P154<br />

Steinmeyer, J.: 16.1.9<br />

Steinwachs, M.: 13.1.2, 21.2.1, 25.2.6<br />

Stelzeneder, D.: P164, P167, P192,<br />

P193<br />

Stenberg, J.: P133<br />

Stendal, J. H.: 25.4.3<br />

Stoddart, M.: P147, P271<br />

Stoeckle, U.: 9.2.8<br />

Strauss, E.: 11.3.9, 15.2.1<br />

Strazzari, A.: P37<br />

Strijkers, G.: 9.4.7<br />

Stroebel, S.: 25.4.6<br />

Stukenborg-Colsman, C.: P194<br />

Sugita, N.: 25.4.9<br />

Suhaeb, A.: P208<br />

Sukegawa, A.: P152<br />

Sukegawa, K.: P7<br />

Sullivan, M.: P134<br />

Sun, J.: 16.3.2, 16.3.4, 16.3.9,<br />

P39, P189<br />

Sundback, C. A.: 9.2.6, P265<br />

Sundman, E.: P241<br />

Suri, M.: 16.3.3<br />

Surowiec, R.: P163<br />

Surtel, D. A.: 9.2.2, 9.4.8, 11.2.3,<br />

P118, P120, P121,<br />

P126<br />

Susa, T.: P221<br />

Syed, H.: P21<br />

Szomolanyi, P.: P30<br />

Szulc, A.: P195, P238<br />

Szumowski, U.: 11.4.3<br />

t<br />

Töyräs, J.: 19.1.3, P161<br />

Tabet, S. K.: 25.3.8<br />

Taheri, M.: P32<br />

Takahashi, I.: 25.1.4<br />

Takamatsu, A.: 16.1.8<br />

Takaso, M.: P7<br />

Takayama, K.: 11.1.3<br />

Takazawa, K.: P101<br />

Tamay De Dios, L.: P84<br />

Tamez-pena, J.: 11.4.4, P30<br />

Tampere, T.: 9.4.6<br />

Tampieri, A.: P37<br />

Tanideh, N.: P233<br />

Taniguchi, N.: 25.1.6<br />

Tavakoli, A.: P233<br />

Taylor, D.: P85<br />

Taylor, S.: P71<br />

Temple-Wong, M. M.: 21.1.2<br />

Teramura, T.: 25.4.9<br />

Tharakan, B.: P175<br />

Theodoropoulos, J.: 11.2.2, 16.2.8<br />

Thermann, H.: 25.2.8, P194<br />

Thier, S.: P110<br />

Thomas, D.: 16.4.9<br />

Thompson, E.: P56<br />

Thompson, P.: P186<br />

Tichy, B.: P80<br />

Timoncini, A.: P93<br />

Tischer, T.: P131<br />

Toh, S.: 25.1.4<br />

Totterman, S. M.: 11.4.4, P30<br />

Tran-Khanh, N.: 25.3.3<br />

Trattnig, S.: 11.4.4, 11.4.5, 19.1.1,<br />

P30, P164, P166<br />

Trimborn, M.: 16.2.6<br />

Trivedi, J.: P173<br />

Trutiak, N.: P234<br />

Tsai-Wu, J.: P145<br />

Tsai, T.: P174<br />

Tse, Y.: 11.1.6<br />

Tseng, A.: 9.2.6<br />

Tsuchida, A. I.: 9.3.8, 11.3.5, 11.4.2,<br />

P151, P156<br />

Tsuda, E.: 25.1.4<br />

Turner, L. J.: 25.4.4<br />

Turner, S.: P173<br />

u<br />

Uchida, K.: P7<br />

Uhl, M.: 25.2.6<br />

Umeda, T.: 25.1.4<br />

Urabe, K.: P7<br />

Usas, A.: P81<br />

Uynuk-ool, T.: 9.2.8<br />

Uzun, M.: P224, P225<br />

v<br />

Vacanti, J.: 9.2.6, P265<br />

Valderrabano, V.: P64<br />

Valverde-Franco, G.: 11.1.4<br />

Van Amerongen, E. A.: P55<br />

Van Blitterswijk, C.: 16.2.4, P42<br />

Van Buul, G.: 16.4.5<br />

Van De Putte, T.: 25.2.7<br />

Van Den Akker, G.: 9.4.8<br />

Van Der Kraan, P.: P259<br />

Van Der Lee, J.: P133<br />

Van Der Veen, A. J.: P172<br />

Van Griensven, M.: 11.2.1<br />

Van Meegeren, M. E.: 11.1.5, P25, P153<br />

Van Osch, G. J.: 3.2.2, 16.2.2, 16.4.5,<br />

P150, P198, P259<br />

Van Rhijn, L. W.: 9.2.2, 9.4.8, 11.2.3,<br />

P118, P120, P121,<br />

P126<br />

Van Rijen, M. H.: 16.4.6<br />

Van Roermund, P. M.: 24.3.2, 25.1.2<br />

Van Roon, J. A.: P153<br />

Van Royen, B. J.: P172<br />

Van Veghel, K.: 11.1.5<br />

Van Weeren, P.: 9.3.4, 11.1.2<br />

Vanhauwermeiren, H.: 25.2.7<br />

Vannini, F.: 25.2.9, P5, P10, P44,<br />

P86, P93, P141, P232,<br />

P236<br />

Vasanawala, S.: 25.3.8<br />

Vasara, A. I.: 25.3.6, P27<br />

Vasheghani Farahani, F.: 25.1.6<br />

Vasilceac, F. A.: 25.1.3, P24<br />

Vasishta, V. G.: P211<br />

Vaz De Lima, M.: P33<br />

Vecsei, V.: 16.3.6<br />

Veen Van Der, A.: 9.4.7<br />

Vekszler, G.: 11.4.6<br />

Velasquillo, C.: P22, P47, P61, P84,<br />

P88<br />

Velebný, V.: P41<br />

Venugopal, V.: 16.3.8<br />

Verbruggen, G.: 9.2.5, 11.3.3, 19.2.2,<br />

P6, P58<br />

Verdonk, P. C.: 9.2.5, 9.4.6, 11.3.2,<br />

11.3.3, 19.2.2, P6,<br />

P58, P178, P183,<br />

P185, P188<br />

Verdonk, R.: 9.2.5, 9.4.6, 11.3.2,<br />

11.3.3, 19.2.2, P6,<br />

P58, P178, P183,<br />

P185, P188, P217<br />

Verhaar, J.: 16.2.2, 16.4.5, P259<br />

Vernon, L.: P109<br />

Victor, J.: 19.2.2<br />

Vignes-Colombeix, C.: P256<br />

Vijayan, S.: P87<br />

Villafuertes, E.: 16.4.5


188<br />

Villalobos Córdova, F.: 9.4.4, P88<br />

Villalobos Jr, F. E.: P47<br />

Vinatier, C.: 9.3.9, 16.2.5, P103,<br />

P256, P257<br />

Vincken, K. L.: 11.4.2<br />

Viren, T.: 19.1.3<br />

Vogrin, M.: P244<br />

Vogt, S.: P272<br />

Volpi, P.: P263<br />

Von Keudell, A.: 25.2.5, P21, P82<br />

Von Rechenberg, B.: P159<br />

Von Windheim, J.: P234<br />

Voncken, J.: 9.4.8, 11.2.3, P120<br />

Vonk, L.: 11.3.5, P79<br />

Voss, L.: 9.2.2, P120<br />

w<br />

Waarsing, J.: 16.4.5<br />

Wagner, P.: P105<br />

Waldman, S.: 11.1.6<br />

Walker, K. E.: 11.2.4<br />

Walker, P.: 9.4.2, 25.1.5, P214<br />

Waller, C. S.: P217<br />

Walsh, D.: 2.2.2<br />

Walter, N. M.: P38<br />

Wang, Q.: 9.2.8<br />

Ware, M. C.: 16.3.3<br />

Watt, S. M.: 16.4.2<br />

Watts, A. E.: 25.4.7<br />

Wawrzynek, W.: P69, P190<br />

Weckbach, S.: P108<br />

Weinans, H.: 16.4.5<br />

Weinstein, T.: P273<br />

Weiss, P.: 9.3.9, 16.2.5, P103,<br />

P256, P257<br />

Weissenberger, M.: P154<br />

Welsch, G. H.: 11.4.5, 11.4.6, 19.1.1,<br />

19.1.2, P164<br />

Welter, J. F.: 16.4.4<br />

Welting, T. J.: 9.2.2, 9.4.8, 11.2.3,<br />

P118, P120, P121,<br />

P126<br />

Wendt, D.: P89<br />

Wessinger, P. H.: 16.3.3<br />

Wexel, G.: P272<br />

Widhalm, H. K.: 11.4.6<br />

Widhalm, K.: 11.4.6<br />

Widuchowski, J.: P69, P190<br />

Widuchowski, W.: P69, P190<br />

Wiegant, K.: 25.1.2, P16, P77<br />

Wiewiorski, M.: P64<br />

Wijnands, K.: P126<br />

Wilensky, D.: P109<br />

Willemot, L.: 9.4.6, P178<br />

Williams, I, R. J.: P85<br />

Williams, R.: 11.4.9, P160, P217<br />

Williams, S.: P71, P78<br />

Wilson, G.: P13, P14, P124<br />

Wilton, T.: P217<br />

Wimmer, M. A.: 16.1.5, P1<br />

Winalski, C. S.: 16.2.9, P168<br />

Windhager, R.: P116, P166<br />

Winterborn, A.: 11.1.6<br />

Woiciechowsky, C.: P171<br />

Wolfová, L.: P41<br />

Wondrasch, B.: 11.4.5<br />

Wood, D. J.: 25.3.5, P94, P95,<br />

P247<br />

Woodell-May, J.: 11.2.9<br />

Woodfield, T.: 11.4.8, 25.4.6, P46<br />

Wright, K.: P29, P31, P262<br />

Wuebbenhorst, D.: P272<br />

Wyland, D.: 16.3.3<br />

x<br />

Xiafei, R.: P18<br />

Xiao, X.: 25.1.8, P127<br />

Authors‘ Index<br />

y<br />

Yamamoto, Y.: 25.1.4<br />

Yamashita, S.: 16.1.8<br />

Yang, S.: P49<br />

Yang, Y.: P48<br />

Yang, Z.: P18<br />

Yao, J. Q.: P18<br />

Yaroshinsky, A.: 11.4.4<br />

Yasuda, K.: P62, P239<br />

Yik, J.: 9.2.7, P119<br />

Yoshikawa, H.: 25.4.9, P207, P221<br />

Yu, H.: P226<br />

Yuan, L.: P20<br />

z<br />

Zaffagnini, S.: P45, P65<br />

Zak, L.: P80<br />

Zaman, T.: P104, P105, P208<br />

Zantop, T.: P72<br />

Zaslav, K. R.: P26<br />

Zati, A.: 25.3.4<br />

Zayed, N.: 11.2.5, P130<br />

Zbyn, S.: 19.1.1<br />

Zeitler, E. M.: 25.1.7<br />

Zelle, S.: P72<br />

Zenclussen, M. L.: P171<br />

Zenobi-Wong, M.: P48<br />

Zhang, R.: P270<br />

Zhao, X.: P265<br />

Zhao, Z.: P2, P8, P11, P12,<br />

P128, P222<br />

Zheng, M. H.: P95, P247<br />

Zhou, X.: P219, P226<br />

Zorzi, C.: P40, P177, P188,<br />

P237<br />

Zuehlke, D.: P194<br />

Zur, G.: P52


Authors‘ Index 189


190<br />

Notes


Please join join us us for for the<br />

Sponsored Symposium<br />

“The Evolution of Autologous Chondrocyte<br />

“The Evolution of Autologous Chondrocyte<br />

Implantation and <strong>Cartilage</strong> <strong>Repair</strong>”<br />

Implantation and <strong>Cartilage</strong> <strong>Repair</strong>”<br />

Description: In this cartilage repair symposium, participants<br />

Description: In this cartilage repair symposium, participants<br />

will learn about the genesis of autologous chondrocyte<br />

will learn about the genesis of autologous chondrocyte<br />

implantation, the importance of the chondrocyte and how<br />

implantation, the importance of the chondrocyte and how<br />

the standards of clinical evidence, global regulation and<br />

the standards of clinical evidence, global regulation and<br />

manufacturing have evolved in this dynamic and exciting field<br />

manufacturing have evolved in this dynamic and exciting field<br />

Date: Monday, May 14th<br />

Date: Monday, May 14th<br />

Time: 13:00 - 14:00<br />

Time: 13:00 - 14:00<br />

Location: Grand Salon<br />

Location: Grand Salon<br />

For more information please visit us at booth #6.<br />

For more information please visit us at booth #6.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!