+ All Categories
Home > Documents > the development of a xenograft-based scaffold for tendon

the development of a xenograft-based scaffold for tendon

Date post: 26-Mar-2023
Category:
Upload: khangminh22
View: 0 times
Download: 0 times
Share this document with a friend
225
THE DEVELOPMENT OF A XENOGRAFT-BASED SCAFFOLD FOR TENDON AND LIGAMENT RECONSTRUCTION BY THORSTEN MARKUS SEYLER A Dissertation Submitted to the Graduate Faculty of WAKE FOREST UNIVERSITY GRADUATE SCHOOL OF ARTS AND SCIENCES in Partial Fulfillment of the Requirements for the Degree of DOCTOR OF PHILOSOPHY MOLECULAR MEDICINE AND TRANSLATIONAL SCIENCE May 2014 Winston-Salem, North Carolina Approved By George J. Christ, Ph.D. (Committee Chair) Thomas L. Smith, Ph.D. (Doctoral Thesis Advisor) Gary G. Poehling, M.D. Mark E. Van Dyle, Ph.D. Michael F. Callahan, Ph.D.
Transcript

THE DEVELOPMENT OF A XENOGRAFT-BASED SCAFFOLD FOR TENDON AND LIGAMENT RECONSTRUCTION

BY

THORSTEN MARKUS SEYLER

A Dissertation Submitted to the Graduate Faculty of

WAKE FOREST UNIVERSITY GRADUATE SCHOOL OF ARTS AND SCIENCES

in Partial Fulfillment of the Requirements

for the Degree of

DOCTOR OF PHILOSOPHY

MOLECULAR MEDICINE AND TRANSLATIONAL SCIENCE

May 2014

Winston-Salem, North Carolina

Approved By

George J. Christ, Ph.D. (Committee Chair)

Thomas L. Smith, Ph.D. (Doctoral Thesis Advisor)

Gary G. Poehling, M.D.

Mark E. Van Dyle, Ph.D.

Michael F. Callahan, Ph.D.

ii

DEDICATION

There are a number of people without whom this thesis might not have been written, and

to whom I am greatly indebted. I dedicate this thesis to my parents Evelyn and Heinz

Werner. I hope that this achievement will complete their dream when they chose to give

me the best education they could and their unequivocal support throughout the years. I also

dedicate this thesis to my loving and supportive wife Tasha, who provided endless words

of encouragement and supported me each step of the way. She was always there cheering

me up and stood by me through the good times and bad times. Thank you for providing me

strength, courage and support that urge me to strive to achieve my goals in life.

iii

ACKNOWLEDGEMENTS

Foremost, I would like to express my sincere gratitude to my advisors Thomas L. Smith,

Ph.D., Mark E. Van Dyke, Ph.D., Gary G. Poehling, M.D. for their continuous support of

my research, for their patience, motivation, enthusiasm, and immense knowledge. Their

guidance helped me in all the time of research and writing of this thesis. I could not have

imagined better advisors and mentors for my Ph.D. study.

Besides my advisors, I would like to thank Patrick W. Whitlock, M.D. Ph.D. for being a

friend and incredible resource on both an academic and a personal level, for which I am

extremely grateful. This thesis would not have been possible without his help, support, and

insightful comments. I would like to acknowledge the rest of my thesis committee: Gary

G. Poehling, M.D., Michael F. Callahan, Ph.D., and George J. Christ, Ph.D. for their

encouragement, insightful comments, and hard questions.

My sincere thanks also goes to Beth P. Smith, Ph.D., for her personal support and great

patience during my time in the orthopaedic research laboratories.

Last but not the least, I would like to thank my fellow residents and lab mates in the

Orthopaedic Surgery Department at Wake Forest: Sandeep Mannava, M.D., Ph.D.;

Johannes F. Plate, M.D. PhD.; Daniel N. Bracey, M.D.; and Peter J. Apel, M.D., Ph.D. for

their support and friendship, the stimulating discussions, the sleepless nights we were

working together, and for all the fun we have had in the last few years. Thank you!

iv

TABLE OF CONTENTS

Contents

Dedication....................................................................................................................................ii

Acknowledgements...................................................................................................................iiiTable of Contents.................................................................................................................................ivList of Figures and Tables.................................................................................................................viList of Abbreviations.........................................................................................................................viiThesis Abstract......................................................................................................................................x

Chapter 1......................................................................................................................................1Introduction...........................................................................................................................................1

Rationale for Decellularization in Tendon and Ligament Tissue Engineering............................2Description of Decellularization Modalities...........................................................................................7References.......................................................................................................................................................11Figures and Tables........................................................................................................................................18

Chapter 2....................................................................................................................................19A Tissue-engineered Approach to Tendon and Ligament Reconstruction: Current Trends...................................................................................................................................................19

Abstract............................................................................................................................................................21Introduction.....................................................................................................................................................22Clinical Significance of Tissue Engineered Solutions......................................................................24Natural Alternative Graft Choices for Tendon and Ligament Reconstruction: Xenografts..29Summary and Conclusions.........................................................................................................................35References.......................................................................................................................................................37Figures and Tables........................................................................................................................................48

Chapter 3....................................................................................................................................51A Novel Process for Optimizing Musculoskeletal Allograft Tissue: Improving Safety, Ultra-Structural Properties and Cell Infiltration......................................................................51

Abstract............................................................................................................................................................53Introduction.....................................................................................................................................................55Materials & Methods...................................................................................................................................57Results..............................................................................................................................................................64Discussion.......................................................................................................................................................68References.......................................................................................................................................................72Figures and Tables........................................................................................................................................78

Chapter 4....................................................................................................................................87The Development of a Xenograft-derived Scaffold for Tendon and Ligament Reconstruction....................................................................................................................................87

Abstract............................................................................................................................................................89Introduction.....................................................................................................................................................91Material & Methods.....................................................................................................................................94Results...........................................................................................................................................................105Discussion....................................................................................................................................................111Conclusions..................................................................................................................................................115References....................................................................................................................................................116Figures and Tables.....................................................................................................................................124

v

Chapter 5.................................................................................................................................134The Effect of Cyclic Strain on the Tensile Properties of a Naturally-Derived, Decellularized Tendon Scaffold Seeded with Allogeneic Tenocytes and Associated mRNA Expression.........................................................................................................................................134

Abstract.........................................................................................................................................................136Introduction..................................................................................................................................................137Material & Methods..................................................................................................................................140Discussion....................................................................................................................................................148Conclusions..................................................................................................................................................154Competing interests...................................................................................................................................155Acknowledgements...................................................................................................................................156References....................................................................................................................................................157Figures and Tables.....................................................................................................................................162

Chapter 6.................................................................................................................................166Summary, Clinical Relevance, & Future Directions...............................................................166

Summary of Doctoral Thesis..................................................................................................................167Clinical Relevance to Orthopaedic Surgery.......................................................................................169Future Direction for Research................................................................................................................171References....................................................................................................................................................172

Curriculum Vitae..................................................................................................................182Post Graduate Education & Training...................................................................................................183Medical Licensures....................................................................................................................................184Professional Society Memberships.......................................................................................................185Honors & Awards......................................................................................................................................186Editorial Activities.....................................................................................................................................187Committees..................................................................................................................................................188Peer-reviewed Publications.....................................................................................................................189Book Chapters.............................................................................................................................................197Meeting Presentations...............................................................................................................................198Electronic Media........................................................................................................................................210Research Grants..........................................................................................................................................211

Permissions to reprint published articles in doctoral thesis........................................212

vi

LIST OF FIGURES AND TABLES

Chapter 1, Figures and Tables Chapter 1, Figure 1 Page 18 Chapter 2, Figures and Tables Chapter 2, Figure 1 Page 48 Chapter 2, Table 1 Page 49 Chapter 2, Table 2 Page 50 Chapter 3, Figures and Tables Chapter 3, Figure 1 Page 78 Chapter 3, Figure 2 Page 79 Chapter 3, Figure 3 Page 80 Chapter 3, Figure 4 Page 81 Chapter 3, Figure 5 Page 82 Chapter 3, Figure 6 Page 83 Chapter 3, Table 1 Page 84 Chapter 3, Table 2 Page 85 Chapter 3, Table 3 Page 86 Chapter 4, Figures and Tables Chapter 4, Figure 1 Page 124 Chapter 4, Figure 2 Page 125 Chapter 4, Figure 3 Page 126 Chapter 4, Figure 4 Page 127 Chapter 4, Figure 5 Page 128 Chapter 4, Figure 6 Page 129 Chapter 4, Figure 7 Page 130 Chapter 4, Figure 8 Page 131 Chapter 4, Table 1 Page 132 Chapter 4, Table 2 Page 133 Chapter 5, Figures and Tables Chapter 5, Figure 1 Page 163 Chapter 5, Figure 2 Page 164 Chapter 5, Table 1 Page 165

vii

LIST OF ABBREVIATIONS

AANA Arthroscopy Association of North America

AAOS American Academy of Orthopaedic Surgeons

AATB American Association of Tissue Banks

ACL Anterior cruciate ligament

Ad Adenovirus

ASTM American Society for Testing and Materials

α-gal Galα1-3Galβ1-4GlcNAc-R

BioRx Bioreactor

CHAPS 3-[(3-cholamidopropyl)dimethylammonio]-1-

propanesulfonate

DAPI 4', 6-diamidino-2-phenylindole

DMEM Dulbecco's modified Eagle's medium

DNA Deoxyribonucleic acid

DPBS Dulbecco's phosphate-buffered saline

E Elastic modulus, Young’s modulus

EDTA Ethylenediaminetetraacetic acid

EGTA Ethyleneglycoltetraacetic acid

ELISA Enzyme-linked immunosorbent assay

FBS Fetal bovine serum

FDA Food and Drug Administration

FDP Flexor digitorum profundus

GAG Glycosaminoglycan

viii

GAPDH Glyceraldehyde 3-phosphate dehydrogenase

H&E Hematoxylin and eosin

hFDATA Freeze-dried human Achilles tendon allograft

IL-6 Interleukin 6

kN Kilonewtons

MMP Matrix metalloproteinase

MPa Megapascal

Mrad Milliradian

MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-

carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-

tetrazolium)

NIH National Institute of Health

NR Neutral red

OREF Orthopaedic Research and Education Foundation

PAA Peracetic acid

PFU Plaque Forming Units

RNA Ribonucleic acid

RTPCR Reverse transcription polymerase chain reaction

SAL Sterility assurance level

SEM Scanning electron microscopy

SEM Standard error of the mean

SDS Sodium dodecyl sulfate

SV5 Simian virus 5

ix

TBP Tri-n-butyl phosphate

TNF-α Tumor necrosis factor-α

UTL Ultimate tensile load

UTS Ultimate tensile stress

VSV Vesicular Stomatitis virus

WHO World Health Organization

x

THESIS ABSTRACT

Tendon and ligament injuries place a significant burden on the United States economy.

Tissue-engineering is an emerging field that may potentially contribute to the development

of novel therapeutic strategies in areas of ligament and tendon reconstruction surgery.

Currently there is a limited supply of donor tendons available for surgical reconstruction.

The goal of tissue engineering is to develop scaffolds to replace damaged and injured

tissues. The major components of tissue engineering approaches include cells,

biomaterials, and an appropriate environment for promoting tissue remodeling. The work

in this thesis is based on the development of a novel decellularization and oxidation

protocol. The data presented characterize both allogenic and xenogenic scaffolds using

histological, mechanical and structural analyses. Scaffolds prepared using this protocol

demonstrate decreased immunogenic potential, are biocompatible in vitro and in vivo,

retain tensile properties compared to native source tissue, possess a modified ultrastructure,

have a decreased risk of disease transmission through viral load elimination, and remodel

and preserve functionality in vivo. Because these decellularized scaffolds provide the

necessary microstructure and extracellular cues for cell attachment, they were successfully

seeded and cultivated in a bioreactor. Cyclic preconditioning restored tensile properties

compared to fresh-frozen tendons. In summary, the application of this novel

decellularization and oxidation process is broadly applicable to create scaffolds from

several sources for tendon and ligament reconstruction. Bioreactor conditioning facilitates

tissue remodeling and improves the performance of scaffolds, making this construct a

potential design to reconstruct tendons and ligaments with sufficient load-bearing capacity

for rapid return of functionality.

1

CHAPTER 1

INTRODUCTION

Thorsten M. Seyler, M.D.

2

RATIONALE FOR DECELLULARIZATION IN TENDON AND LIGAMENT

TISSUE ENGINEERING

Tendon and ligament injuries place a significant burden on the United States

economy, accounting for nearly 32 million orthopaedic musculoskeletal injuries annually.1

Some of these ligamentous and tendinous injuries represent a treatment challenge and

reproducible satisfactory post-surgical results are difficult to achieve. Orthopaedic

surgeons have successfully reconstructed tendons and ligaments utilizing autografts and

allografts; however, re-rupture poses a significant complication that limits the surgeon’s

operative choices. Tissue-engineering is an emerging field that may contribute to the

development of novel therapeutic strategies in ligament and tendon reconstruction

surgery.2-4

The major objective of tissue engineering is to reconstruct tissues in order to replace

injured or damaged tissues without compromising function. An exciting concept is the

decellularization of allogenic or xenogenic tissues that can be used for reconstructive

surgery.5-7 The ultimate goal of tissue decellularization is to remove cellular and antigenic

material without affecting the composition of the extracellular matrix (ECM), preserve

biological activity, and maintain structural integrity. This produces an ideal

biological scaffold since it contains all the components of the tissue from which it

was derived except for the living cells. Preservation of the ECM has demonstrated site-

specific cell differentiation and remodeling by providing a three-dimensional

ultrastructure.8 The nature of the tissue source ultimately determines the physical

microstructure and promotes site-specific differentiation of progenitor cells.9

3

The insufficient vascularization of ligaments (i.e. anterior cruciate ligament)

prevents them from healing completely after an extreme tear or rupture, creating a need for

operative reconstruction. Using the anterior cruciate ligament (ACL) as an example, four

options have been utilized for the repair or replacement: (1) autografts, (2) allografts, (3)

xenografts, and (4) synthetic grafts. Autogenous tissue avoids the perceived risk of disease

transmission and demonstrates superior rates of graft incorporation and remodeling when

compared to allografts.10 Traditionally, autogenous bone-patellar tendon-bone grafts have

been considered the “gold standard” and combine several advantages such as greater

fixation strength, superior mechanical properties, and good long-term results when

compared to other grafting techniques.11, 12 The graft consists of the central one third of the

patellar tendon and includes a bone plug proximally from the patella and distally from the

tibia. Bone-patellar tendon-bone offers the strongest healing potential because it relies

mainly on bone-to-bone healing between the graft bone plug and the tunnel.13

Disadvantages associated with autogenous bone–patellar tendon–bone grafts including

quadriceps weakness, arthrofibrosis, patellofemoral pain, patellar tendinitis or rupture,

patellar fracture, and patella infera syndrome.14 Due to improvements in graft fixation

techniques, the preparation of multiple bundle grafts, and reduced donor site morbidity,

there has been an increase in popularity of the use of hamstring tendons as the autograft for

ACL reconstruction. The hamstring graft has evolved from using a single strand of either

semitendinosis or gracilis tendon to a quadruple loop combination of semitendinosis and

gracilis tendons. Although this graft choice has become more popular, hamstring tendon

autografts rely on soft tissue-to-bone healing, which occurs at a slower rate than bone-to-

4

bone healing.15 Disadvantages associated with hamstring grafts include potential harvest

pain and flexion torque loss.

The use of allogenic tissue is another common alternative for reconstructing the

ACL. Potential advantages of allografts include decreased donor site morbidity, immediate

availability of various graft sizes (off-the-shelf), less postoperative pain, better

postoperative function, and lower incidence of postoperative arthrofibrosis. The greatest

disadvantage of allografts is the potential risk of disease transmission. Contamination of

allografts can occur when the tissue is still confined to the donor’s corpus and during or

after tissue removal because of improper tissue processing procedures. In the past, the two

methods employed to avoid disease transmission were graft sterilization using ethylene

oxide treatment or gamma irradiation. Ethylene oxide is no longer used because of

associated synovitis and intra-articular graft destruction. Gamma irradiation is thought to

create free radicals and modify nucleic acids leading to virus and bacterial destruction. It

has also been associated with disruption of the structural integrity of the graft.16 Therefore,

both sterilization techniques are currently not used on allografts for ACL reconstruction.

Initial concerns of graft weakness using the ethylene oxide or high dose gamma irradiation

sterilization process have been addressed by employing multimodal process that involves

sterile harvesting of tissue from the donor, rigorous donor screening/testing, antibiotic

soaking, chemical sterilization processes, low dose irradiation (1.2-1.8 Mrad), and a final

preservation technique such as lyophilization or freezing.12, 17, 18 Despite these advances in

graft sterilization, one of the major drawbacks of using allografts is slow incorporation into

host bone and graft remodeling, which may lead to reduced graft strength and early

failure.15

5

In the 1980s, bovine xenografts cross-linked with glutaraldehyde were used with

moderate success. Because follow-up studies could not reproduce these promising early

findings, bovine xenografts failed to obtain Food and Drug Administration (FDA) approval

owing to high failure rates and complications associated with their use. The mechanism of

failure for the bovine-based devices included poor biocompatibility and graft rejection

attributed to excess glutaraldehyde, as well as improper biomechanical properties pre-

implantation, lack of host integration, and recurrent effusions.19, 20

The potential advantages of synthetic ligaments are multifold. They would

eliminate the problems of graft harvest, would be readily available for use, and would offer

no risk of disease transmission. However, a problem with synthetic grafts is identifying the

ideal combination of materials that would provide initial support for the knee while

allowing gradual ingrowth and graft remodeling. A spectrum of devices has been created

including Dacron, Kevlar, and carbon fiber grafts. The initial clinical success using non-

degradable synthetic scaffolds could not be maintained in long term studies because these

grafts frequently failed due to inappropriate mechanical properties, material fatigue

profiles, and material shedding.21-24 Adverse events associated with synthetic grafts are

based on extensive wear as well as foreign-body responses.19 Abraded synthetic particles

were detected in the joint space, lymph nodes, and other tissues leading to a chronic

inflammatory response.25

Despite the clinical success using autogenous and allogenous grafts, there is no

consensus among orthopaedic surgeons on the “perfect graft” choice.11, 14, 26 Surgeons

continue to seek alternative reconstruction techniques in an attempt to reduce

complications and improve long-term outcomes. Since naturally-derived scaffolds in tissue

6

engineering are of allogenic or xenogenic origin, the removal of cellular material and

antigens is critical. Most of the decellularization protocols are based on a combination of

physical, chemical, and enzymatic modalities. In order to develop an effective protocol for

decellularization, it is of utmost importance to account for many physical factors of the

tissue of interest including thickness, cellularity, density, and lipid content. Thicker tissues

may require more extensive decellularization modalities and longer exposure times. Highly

cellular tissues may require additional mechanical modalities and exposure to a

combination of detergents followed by rinsing. Fatty tissues may require the addition of

lipid solvents. It is clear that any decellularization modality intended to remove cellular

material can negatively impact ECM composition and the mechanical integrity of the

tissue. Therefore, it is important to find a balance between maximizing removal of cellular

material and antigens while minimizing adverse effects on structural integrity. The protocol

for decellularization and oxidation used in this thesis is a combination of physical,

chemical, and enzymatic modalities (Figure 1) that have been optimized for ligament and

tendon decellularization. As an introduction to decellularization processes, the following

sections will provide a brief overview of the most commonly used decellularization

modalities and the effects of these modalities upon biologic scaffold material.

7

DESCRIPTION OF DECELLULARIZATION MODALITIES

Physical Modalities

The most commonly used physical modalities in decellularization protocols include

freezing, force and direct pressure, sonication, and agitation. Freezing is an effective

method of cell lysis that is frequently used for decellularization of tendinous and

ligamentous tissue.27 However, the freezing process must be followed by a process that can

remove cellular material. Although freezing can lead to minor disruptions in the ECM, the

impact on mechanical properties is minimal.28

Mechanical force and hydrostatic pressure can be used to lyse and remove cells

in tissues with less densely organized ECM. Both methods are effective for

decellularization of tissues that are characterized by natural planes. However, this method

can lead to damage to the structural integrity of the tissue.

Sonication is a less frequently used modality to disrupt cell membranes and

decellularize tissues.29 It is often used as an adjunct to other modalities; little is known

about optimal magnitude, frequency, and duration of application.30

Mechanical agitation is frequently used in decellularization protocols. While

agitation itself can cause cell lysis, the main purpose of integrating mechanical agitation

into decellularization protocols is to facilitate exposure of the tissue to a chemical

decellularization agent in order to remove cellular debris.31, 32 The duration and intensity

of agitation varies based on the thickness and density of the tissue of interest.

Chemical Modalities

Multiple chemical agents can be used to decellularize tissues. Both hypertonic and

hypotonic solutions can be utilized to cause cell lysis. Treatment with hypertonic solutions

8

typically dissociates DNA from proteins33, whereas exposure to hypotonic solutions

typically causes cell lysis by an osmotic effect.34 Utilization of these agents does not impact

tissue microarchitecture but also does not remove cellular debris and, therefore, requires

combination with additional enzymatic or chemical treatments.

Acid and base treatments can disrupt cell membranes and intracellular organelles,

effectively removing nucleic acids.5 Frequently used agents are hydrochloric acid, sulfuric

acid, and ammonium hydroxide. While most of these agents are detrimental to the ECM

and need to be used with caution35, peracetic acid has proven to preserve ECM proteins

and mechanical strength.36, 37

Detergents are some of the most commonly used agents in decellularization

protocols.5, 7 They are typically divided into ionic, non-ionic, and twitterionic detergents.

Their mechanism of action is dissociation of proteins in the extracellular matrix and

removal of DNA. The effectiveness of a detergent depends on the tissue type and exposure

time.38 Non-ionic detergents such as triton X-100 are presumed to be milder and disrupt

lipid-lipid and lipid-protein bonds. They are effective in removing nuclear material and

cause a significant decrease in glycosaminoglycans (GAG) content.39 However, Triton X-

100 has also been shown to preserve collagen content while decreasing tensile strength of

individual collagen fibers with prolonged incubation. Ionic detergents such as sodium

dodecyl sulfate (SDS) or Triton X-200 are considered to be effective for removal of cellular

material. However, these agents tend to denature proteins by disrupting protein to protein

bonds; they have been associated with cytotoxicity.40 Zwitterionic agents such as 3-[(3-

cholamidopropyl)dimethylammonio]-1-propanesulfonate (CHAPS) have properties of

9

both non-ionic and anionic detergents, exhibiting a greater tendency to denature proteins

than non-ionic agents but resulting in less disruption to the ultrastructure than ionic agents.5

Organic solvents including Tri-n-butylphosphine (TBP) and alcohols inactivate

pyrogens, remove cells from dense tissues, and have minimal effects on the mechanical

properties of tissues.7 Alcohols can also be used to remove lipids from tissue and often are

used in initial steps of decellularization. TBP has shown to inactivate viruses which can

aid in reducing the risk for disease transmission in allogenic and xenogenic tissues.41

Chelating agents such as ethylenediaminetetraacetic acid (EDTA) and

ethyleneglycoltetraacetic acid (EGTA) are typically used with enzymatic modalities due to

their ineffectiveness when used alone.5-7 These agents bind metallic ions, disrupting cell

attachment to the ECM. EDTA is typically used in conjunction with trypsin.

Enzymatic Modalities

Exposure to enzymatic agents should be limited due to their adverse effects on the

ECM. The most commonly used enzymes in decellularization processes are trypsin,

nucleases, collagenase, lipase, dispase, and α-glactosidase.5 Nucleases cleave the

phosphodiester bonds between the nucleotide subunits of nucleic acids facilitating removal

of nucleotides.42 The use of collagenase is only advised when preservation of the

ultrastructure is not critically important. Trypsin is the most commonly used enzymatic

agent, cleaving peptide chains mainly at the carboxyl end of lysine or arginine, except when

either is followed by proline. Prolonged exposure to trypsin can cause disruption of the

ECM because of removal of laminin, fibronectin, and GAGs. While prolonged exposure to

trypsin is associated with a decrease in tensile strength, the overall content of collagen was

not reduced.43 Lipase is used in the decellularization of tissue with high lipid content;

10

however, its efficacy as sole lipid removing agent is in question.44 In the decellularization

of xenogenic tissues, α-glactosidase can be used to remove the α-Gal epitope and reduce

the immunogenic potential of the xenogenic tissue.45

11

REFERENCES

1. Butler DL, Juncosa-Melvin N, Boivin GP, Galloway MT, Shearn JT, Gooch C, et

al. Functional tissue engineering for tendon repair: A multidisciplinary strategy using

mesenchymal stem cells, bioscaffolds, and mechanical stimulation. Journal of Orthopaedic

Research. 2008;26(1):1-9.

2. Turner NJ, Badylak SF. Biologic scaffolds for musculotendinous tissue repair.

European cells & materials. 2013;25:130-43. Epub 2013/01/19.

3. Hogan MV, Bagayoko N, James R, Starnes T, Katz A, Chhabra AB. Tissue

engineering solutions for tendon repair. The Journal of the American Academy of

Orthopaedic Surgeons. 2011 Mar;19(3):134-42. Epub 2011/03/04.

4. Woo SL, Hildebrand K, Watanabe N, Fenwick JA, Papageorgiou CD, Wang JH.

Tissue engineering of ligament and tendon healing. Clinical orthopaedics and related

research. 1999 Oct(367 Suppl):S312-23. Epub 1999/11/05.

5. Crapo PM, Gilbert TW, Badylak SF. An overview of tissue and whole organ

decellularization processes. Biomaterials. 2011 Apr;32(12):3233-43. Epub 2011/02/08.

6. Gilbert TW. Strategies for tissue and organ decellularization. Journal of cellular

biochemistry. 2012 Jul;113(7):2217-22. Epub 2012/03/15.

7. Gilbert TW, Sellaro TL, Badylak SF. Decellularization of tissues and organs.

Biomaterials. 2006 Jul;27(19):3675-83. Epub 2006/03/08.

8. Nelson CM, Tien J. Microstructured extracellular matrices in tissue engineering

and development. Current opinion in biotechnology. 2006 Oct;17(5):518-23. Epub

2006/09/15.

12

9. Daly KA, Liu S, Agrawal V, Brown BN, Johnson SA, Medberry CJ, et al. Damage

associated molecular patterns within xenogeneic biologic scaffolds and their effects on host

remodeling. Biomaterials. 2012 Jan;33(1):91-101. Epub 2011/10/05.

10. Jackson DW, Grood ES, Goldstein JD, Rosen MA, Kurzweil PR, Cummings JF, et

al. A comparison of patellar tendon autograft and allograft used for anterior cruciate

ligament reconstruction in the goat model. Am J Sports Med. 1993 Mar-Apr;21(2):176-85.

11. Chang SK, Egami DK, Shaieb MD, Kan DM, Richardson AB. Anterior cruciate

ligament reconstruction: allograft versus autograft. Arthroscopy : the journal of

arthroscopic & related surgery : official publication of the Arthroscopy Association of

North America and the International Arthroscopy Association. 2003 May-Jun;19(5):453-

62. Epub 2003/05/02.

12. Poehling GG, Curl WW, Lee CA, Ginn TA, Rushing JT, Naughton MJ, et al.

Analysis of outcomes of anterior cruciate ligament repair with 5-year follow-up: allograft

versus autograft. Arthroscopy : the journal of arthroscopic & related surgery : official

publication of the Arthroscopy Association of North America and the International

Arthroscopy Association. 2005 Jul;21(7):774-85. Epub 2005/07/14.

13. Papageorgiou CD, Ma CB, Abramowitch SD, Clineff TD, Woo SL. A

multidisciplinary study of the healing of an intraarticular anterior cruciate ligament graft

in a goat model. Am J Sports Med. 2001 Sep-Oct;29(5):620-6.

14. Sherman OH, Banffy MB. Anterior cruciate ligament reconstruction: which graft

is best? Arthroscopy : the journal of arthroscopic & related surgery : official publication of

the Arthroscopy Association of North America and the International Arthroscopy

Association. 2004 Nov;20(9):974-80. Epub 2004/11/05.

13

15. Jackson DW, Corsetti J, Simon TM. Biologic incorporation of allograft anterior

cruciate ligament replacements. Clinical orthopaedics and related research. 1996

Mar(324):126-33. Epub 1996/03/01.

16. Vangsness CT, Jr., Garcia IA, Mills CR, Kainer MA, Roberts MR, Moore TM.

Allograft transplantation in the knee: tissue regulation, procurement, processing, and

sterilization. Am J Sports Med. 2003 May-Jun;31(3):474-81. Epub 2003/05/17.

17. Musculoskeletal Transplant Foundation MTF. MTF Allograft Tissue Instructions

for Use. 2006.

18. Jones DB, Huddleston PM, Zobitz ME, Stuart MJ. Mechanical properties of

patellar tendon allografts subjected to chemical sterilization. Arthroscopy : the journal of

arthroscopic & related surgery : official publication of the Arthroscopy Association of

North America and the International Arthroscopy Association. 2007 Apr;23(4):400-4.

Epub 2007/04/10.

19. Olson EJ, Kang JD, Fu FH, Georgescu HI, Mason GC, Evans CH. The biochemical

and histological effects of artificial ligament wear particles: in vitro and in vivo studies.

Am J Sports Med. 1988 Nov-Dec;16(6):558-70. Epub 1988/11/01.

20. Vunjak-Novakovic G, Altman G, Horan R, Kaplan DL. Tissue engineering of

ligaments. Annual review of biomedical engineering. 2004;6:131-56. Epub 2004/07/17.

21. Bolton CW, Bruchman WC. The GORE-TEX expanded polytetrafluoroethylene

prosthetic ligament. An in vitro and in vivo evaluation. Clinical orthopaedics and related

research. 1985 Jun(196):202-13. Epub 1985/06/01.

22. Lukianov AV, Richmond JC, Barrett GR, Gillquist J. A multicenter study on the

results of anterior cruciate ligament reconstruction using a Dacron ligament prosthesis in

14

"salvage" cases. Am J Sports Med. 1989 May-Jun;17(3):380-5; discussion 5-6. Epub

1989/05/01.

23. Schindhelm K, Rogers GJ, Milthorpe BK, Hall PJ, Howlett CR, Sekel R, et al.

Autograft and Leeds-Keio reconstructions of the ovine anterior cruciate ligament. Clinical

orthopaedics and related research. 1991 Jun(267):278-93. Epub 1991/06/01.

24. Weiss AB, Blazina ME, Goldstein AR, Alexander H. Ligament replacement with

an absorbable copolymer carbon fiber scaffold--early clinical experience. Clinical

orthopaedics and related research. 1985 Jun(196):77-85. Epub 1985/06/01.

25. Margevicius KJ, Claes LE, Durselen L, Hanselmann K. Identification and

distribution of synthetic ligament wear particles in sheep. J Biomed Mater Res. 1996

Jul;31(3):319-28. Epub 1996/07/01.

26. Herrington L, Wrapson C, Matthews M, Matthews H. Anterior cruciate ligament

reconstruction, hamstring versus bone-patella tendon-bone grafts: a systematic literature

review of outcome from surgery. The Knee. 2005 Jan;12(1):41-50. Epub 2005/01/25.

27. Jackson DW, Grood ES, Cohn BT, Arnoczky SP, Simon TM, Cummings JF. The

effects of in situ freezing on the anterior cruciate ligament. An experimental study in goats.

The Journal of bone and joint surgery American volume. 1991 Feb;73(2):201-13. Epub

1991/02/01.

28. Jackson DW, Grood ES, Wilcox P, Butler DL, Simon TM, Holden JP. The effects

of processing techniques on the mechanical properties of bone-anterior cruciate ligament-

bone allografts. An experimental study in goats. Am J Sports Med. 1988 Mar-

Apr;16(2):101-5. Epub 1988/03/01.

15

29. Azhim A, Yamagami K, Muramatsu K, Morimoto Y, Tanaka M. The use of

sonication treatment to completely decellularize blood arteries: a pilot study. Conference

proceedings : Annual International Conference of the IEEE Engineering in Medicine and

Biology Society IEEE Engineering in Medicine and Biology Society Conference.

2011;2011:2468-71. Epub 2012/01/19.

30. Hung SH, Su CH, Lee FP, Tseng H. Larynx decellularization: combining freeze-

drying and sonication as an effective method. Journal of voice : official journal of the Voice

Foundation. 2013 May;27(3):289-94. Epub 2013/03/16.

31. Gillies AR, Smith LR, Lieber RL, Varghese S. Method for decellularizing skeletal

muscle without detergents or proteolytic enzymes. Tissue engineering Part C, Methods.

2011 Apr;17(4):383-9. Epub 2010/10/27.

32. Woods T, Gratzer PF. Effectiveness of three extraction techniques in the

development of a decellularized bone-anterior cruciate ligament-bone graft. Biomaterials.

2005 Dec;26(35):7339-49. Epub 2005/07/19.

33. Cox B, Emili A. Tissue subcellular fractionation and protein extraction for use in

mass-spectrometry-based proteomics. Nature protocols. 2006;1(4):1872-8. Epub

2007/05/10.

34. Xu CC, Chan RW, Tirunagari N. A biodegradable, acellular xenogeneic scaffold

for regeneration of the vocal fold lamina propria. Tissue engineering. 2007 Mar;13(3):551-

66. Epub 2007/05/24.

35. Dong X, Wei X, Yi W, Gu C, Kang X, Liu Y, et al. RGD-modified acellular bovine

pericardium as a bioprosthetic scaffold for tissue engineering. J Mater Sci Mater Med. 2009

Nov;20(11):2327-36. Epub 2009/06/10.

16

36. Hodde J, Janis A, Hiles M. Effects of sterilization on an extracellular matrix

scaffold: part II. Bioactivity and matrix interaction. J Mater Sci Mater Med. 2007

Apr;18(4):545-50. Epub 2007/06/05.

37. Hodde JP, Record RD, Tullius RS, Badylak SF. Retention of endothelial cell

adherence to porcine-derived extracellular matrix after disinfection and sterilization.

Tissue engineering. 2002 Apr;8(2):225-34. Epub 2002/05/29.

38. Chen RN, Ho HO, Tsai YT, Sheu MT. Process development of an acellular dermal

matrix (ADM) for biomedical applications. Biomaterials. 2004 Jun;25(13):2679-86. Epub

2004/01/31.

39. Meyer SR, Chiu B, Churchill TA, Zhu L, Lakey JR, Ross DB. Comparison of aortic

valve allograft decellularization techniques in the rat. J Biomed Mater Res A. 2006

Nov;79(2):254-62. Epub 2006/07/04.

40. Seddon AM, Curnow P, Booth PJ. Membrane proteins, lipids and detergents: not

just a soap opera. Biochim Biophys Acta. 2004 Nov 3;1666(1-2):105-17. Epub 2004/11/03.

41. Horowitz B, Prince AM, Hamman J, Watklevicz C. Viral safety of

solvent/detergent-treated blood products. Blood coagulation & fibrinolysis : an

international journal in haemostasis and thrombosis. 1994 Dec;5 Suppl 3:S21-8; discussion

S9-S30. Epub 1994/12/01.

42. Rieder E, Kasimir MT, Silberhumer G, Seebacher G, Wolner E, Simon P, et al.

Decellularization protocols of porcine heart valves differ importantly in efficiency of cell

removal and susceptibility of the matrix to recellularization with human vascular cells. The

Journal of thoracic and cardiovascular surgery. 2004 Feb;127(2):399-405. Epub

2004/02/06.

17

43. Schenke-Layland K, Vasilevski O, Opitz F, Konig K, Riemann I, Halbhuber KJ, et

al. Impact of decellularization of xenogeneic tissue on extracellular matrix integrity for

tissue engineering of heart valves. Journal of structural biology. 2003 Sep;143(3):201-8.

Epub 2003/10/24.

44. Brown BN, Freund JM, Han L, Rubin JP, Reing JE, Jeffries EM, et al. Comparison

of three methods for the derivation of a biologic scaffold composed of adipose tissue

extracellular matrix. Tissue engineering Part C, Methods. 2011 Apr;17(4):411-21. Epub

2010/11/04.

45. Stone KR, Ayala G, Goldstein J, Hurst R, Walgenbach A, Galili U. Porcine

cartilage transplants in the cynomolgus monkey. III. Transplantation of alpha-

galactosidase-treated porcine cartilage. Transplantation. 1998 Jun 27;65(12):1577-83.

Epub 1998/07/17.

18

FIGURES AND TABLES

Figure 1. Detailed outline of decellularization protocol used for all experiments.

19

CHAPTER 2

A TISSUE-ENGINEERED APPROACH TO TENDON AND LIGAMENT

RECONSTRUCTION: CURRENT TRENDS

Thorsten M. Seyler, M.D.1, 2; Daniel N. Bracey, M.D.1, 2; Sandeep Mannava, M.D.,

Ph.D.1,3; Gary G. Poehling, M.D.1; Patrick W. Whitlock, M.D., Ph.D.1,4,5

1Department of Orthopaedic Surgery, Wake Forest University School of Medicine,

Winston Salem, North Carolina

2Molecular Medicine and Translational Science Graduate Program, Wake Forest

University Graduate School of Arts & Sciences, Winston Salem, North Carolina

3Neuroscience Graduate Program, Wake Forest University Graduate School of Arts &

Sciences, Winston Salem, North Carolina

4 Maurice E. Müller European Traveling Fellow in Hip Reconstruction, Zurich,

Switzerland

5Department of Orthopaedic Surgery, Children’s Hospital Los Angeles, Los Angeles,

California

20

The following manuscript was accepted for publication in Sports Injuries: Prevention,

Diagnosis, Treatment and Rehabilitation edited by Mahmut Nedim Doral and Jon Karlsson

in March, 2014. This chapter is reprinted with permission. Stylistic variations are due to

the requirements of the journal Thorsten M. Seyler made substantial contributions to the

conception of the work and performed the literature research. HE is responsible for drafting

the work and revising it critically for important intellectual content including final approval

of the version to be published. He is accountable for all aspects of the work in ensuring that

questions related to the accuracy or integrity of any part of the work are appropriately

investigated and resolved. He also served as corresponding author.

21

ABSTRACT

Current graft choices for tendon and ligament reconstruction include autograft and

allografts, with more than 900,000 allografts used for reconstructions annually in the

United States. Given the limitations of autografts and allografts, both synthetic and

naturally-derived tissue-engineered scaffolds have been developed. Despite its early

success, synthetic grafts have been associated with a high incidence of chronic foreign

body inflammation, debris-induced synovitis, mechanical limitations, and graft failure.

Therefore, in recent years the focus has shifted to the use of naturally-derived scaffolds and

an evolving discipline called "functional tissue engineering" which uses a combination of

stem cells, biocompatible scaffolds, and mechanical stimulation to produce tissue

engineered constructs suitable to replace or repair load-bearing structures such as tendons

and ligaments.

22

INTRODUCTION

Tendon and ligament injuries place a large burden on the United States economy,

accounting for nearly 32 million orthopaedic musculoskeletal injuries annually 1. Tissue-

engineering, specifically, the production of an optimized scaffold for soft tissue

augmentation, reconstruction or regeneration may offer significant advantages over

traditional allografts and autografts. Traditional allograft tissue is limited in supply, poses

concerns surrounding disease transmission and inflammatory reactions to donor cell

material as well as possible decreases in the tensile properties of grafts that undergo

chemical sterilization or irradiation 2. Autograft tissue is also limited in supply and

associated with significant donor site morbidity (pain, muscle atrophy, tendonitis), as well

as increased procedural cost and delayed return to activity 3.

Because of the limitations associated with both autograft and allograft tissues, interest has

developed in engineering a replacement with biologic and mechanical properties similar to

that of the native tissue. This interest is reflected by the more than ten-fold increase in the

number of articles published over the last decade addressing tendon and ligament tissue

engineering (Figure 1). A tissue-engineered scaffold for use in tendon and ligament

reconstruction has the potential to reduce or prevent disease transmission from allografts

and diminish any potential immunologic foreign-body response associated with allograft

reconstruction. Considering the advantages of tissue engineered grafts, it is not surprising

that they are gaining popularity with surgeons. A recent survey of orthopaedic surgeons in

the United Kingdom showed that 86% of surgeons would consider using a tissue

engineered graft in anterior cruciate ligament reconstruction 4.

23

Tissue engineering strategies vary by complexity of the target tissue. One example is the

interest in developing an anterior cruciate ligament (ACL) substitute.5, 6 Although some of

the tissue engineered substitutes showed promising short-term results, most failed over

time.7-10 The challenges of engineering an ACL substitute are highlighted by the ligament’s

unique anatomy that is constituted by two bundles which exhibit different tensioning

patterns throughout the knee’s range of motion and also by matching the mechanical

properties of the ligament. Additional challenges include simulating the ligament’s in vivo

environment and the lack of adequate blood supply which limits healing potential. 11

Historically, most ACL substitutes have been constructed with synthetic materials that

oversimplify the ligament’s complex anatomy and structure. As a result, no ACL substitute

has replicated the superior results achieved with autograft reconstruction to date. 12, 13 In

contrast to the ACL, tissue engineering strategies to improve rotator cuff repair outcomes

rely on tissue augmentation rather than substitution.14 Most scaffolds developed to enhance

outcomes of rotator cuff surgery are derived from human or xenograft extracellular matrix

or dermis.14 Synthetic scaffolds for rotator cuff repair are rare when compared to scaffolds

used for ACL reconstruction. The biggest concern with natural derived scaffolds for rotator

cuff surgery is the in vivo host response. Reports from a prospective randomized trial using

xenogenic extracellular matrix scaffolds showed a 20 to 30% hypersensitivity reaction to

the scaffold material.15 The results using human dermal matrix for augmentation of large

rotator cuff tears showed promising results at two years with 85% of intact rotator cuff.16

The difference between the success rates of these scaffolds is likely due to the nature of the

scaffold material. Early xenogenic-derived scaffolds performed poorly because of

24

incomplete removal of cells and the presence of the xenoantigen galactose-α (1,3)-

galactose.14

The ideal scaffold for tendon and ligament reconstruction would be: (1) naturally derived

from either allogeneic or xenogeneic material amenable to host-cell mediated remodeling

in vivo; (2) devoid of cellular material to minimize inflammatory potential, disease

transmission, and host immune response; (3) cytocompatible; (4) of optimal micro-

architecture to promote efficient cell seeding, infiltration, and attachment of the recipient’s

own cells prior to or after implantation; and (5) distinguished by sufficient biomechanical

integrity to withstand rehabilitation until complete remodeling of the scaffold has occurred

17. Such a scaffold should serve as template for cell attachment, promote rapid remodeling,

possess increased strength, demonstrate improved healing, and permit early rehabilitation

and return to function after implantation. Tissue engineered scaffolds have the potential to

vastly improve the treatment of tendon and ligament injuries, especially for larger defects

associated with tumor, trauma, and congenital deficiencies where autograft or allograft

tissue may not be available in sufficient quantity for reconstruction.

CLINICAL SIGNIFICANCE OF TISSUE ENGINEERED SOLUTIONS

Different approaches and various materials have been employed throughout the years to

design tissue-engineered tendon and ligament scaffolds. Some scaffolds are designed to

strictly serve as augmentation devices that protect biological grafts from high loads in the

early postoperative period of graft weakness. 18 These devices are generally implanted

simultaneously with a biological graft (autograft or allograft) to share the mechanical load.

Other scaffolds are designed to replace the use of allograft and autograft tissues entirely,

25

such as in coracoclavicular ligament reconstruction for the treatment of grade V

acromioclavicular joint separations (semitendinosus) and reconstruction of chronic biceps

tendon ruptures (tendoachilles). The Musculoskeletal Transplant Foundation estimates that

over 900,000 allografts are used every year in the United States and that 1.5 million

musculoskeletal allografts were used in 2007 alone 19, 20 lending evidence to how large a

market this is. Allograft distribution is regulated by the American Association of Tissue

Banks (AATB) which initiated an inspection/accreditation program in 1986 that accredited

98 American tissue banks by the year 2006. With AATB oversight and regulations, disease

transmission from allograft tissue has fortunately become a rare event. However, pathogen

and disease transmission produces devastating consequences for a patient and every

measure should be taken to prevent this 21. Tissue engineered grafts derived from non-

human sources can eliminate the risk of disease transmission and minimize the risk of

contamination when novel, aggressive sterilization techniques are used.

Tissue autografts represent the historic “gold standard” in orthopaedic literature because

of their excellent clinical outcomes and because they avoid the risk of disease transmission

associated with allografts since the tissue is harvested from the patient’s own body.

However, despite these advantages, patient safety remains a concern with autografts when

considering the morbidity imparted to patients by autograft harvest. Harvesting the

autograft is an additional, invasive surgical procedure that presents risks which should be

avoided in vulnerable patient populations, such as polytrauma or oncologic patients.

Additionally, autograft harvest prolongs surgical and recovery times which ultimately

inflates the cost and burden on our health care system. Tissue engineered grafts, which are

26

commonly offered as “off-the-shelf” products at the time of surgery can avoid these

additional surgical risks and expenditures.

A. Clinical Significance: Rotator Cuff Reconstruction

Tissue-engineering may contribute to the development of novel therapeutic strategies in

rotator cuff reconstruction. Tissue engineered constructs are primarily designed to augment

or gap bridge large retracted tendon defects, in order to improve the healing and remodeling

of a torn rotator cuff.14 At present, there is no single, optimal surgical treatment that

successfully restores pre-injury shoulder strength, range of motion, and function 22. Despite

considerable surgical advances, these surgeries are still associated with a high rate (20-

70%) of recurrent tearing 23-25. Poor outcomes are commonly associated with risk factors

that include large tear size 26, high degree of muscle atrophy 27, poor tendon quality, and

inappropriate postoperative rehabilitation protocols 28. Soft tissue augmentation of the torn

rotator cuff has been attempted with various biologic materials including porcine small

intestine submucosa, dermal grafts, and/or tissue engineered synthetic grafts (Table I).

Although the results have been variable, these methods have the potential to improve the

healing and remodeling of the injured tendon 15, 29, 30. Rotator cuff injuries may require an

increased amount of tension to repair the tendon back to the humeral head secondary to

muscle atrophy and retraction. The use of tissue engineered constructs can alleviate the

excessive tension required for the repair of some rotator cuff tendons and this may improve

surgical outcomes. 31-33 In summary, large and chronic rotator cuff tears pose a significant

clinical problem that is inadequately addressed by current orthopaedic interventions.

Tissue engineered constructs offer a promising alternative to current graft options by

27

delivering a graft with favorable mechanical properties to the site of injury, which aids in

the initial implantation of tendons and ligaments immediately after surgery, as well as graft

integration and healing in the months to years following surgery. Use of a tissue-engineered

construct eliminates the constraints on graft availability and offers the advantage of

reduced morbidity, quicker rehabilitation, and more rapid graft bio-incorporation. Thus,

use of tissue-engineered constructs in rotator cuff reconstruction surgery might allow for a

substantial improvement over present surgical treatments. Implanting a tissue-engineered

graft with favorable mechanical properties will allow for an expedited return to work and

recreational activities for patients who have undergone rotator cuff reconstruction surgery

B. Clinical Significance: Anterior Cruciate Ligament

The anterior cruciate ligament is one the most commonly injured ligaments of the knee,

accounting for 175,000 reconstructions each year at a cost of $2 billion dollars in the United

States alone 34. Once torn, insufficient vascularization of the injured ACL limits healing

which may lead to joint instability, subsequent chondral and meniscal injury, osteoarthritis,

and long-term disability. To prevent these sequelae, patients with complete ACL ruptures

traditionally elect for operative reconstruction of the ligament. The current "gold standard"

for the treatment of ACL injuries is the placement of an intraarticular autograft or allograft.

Current graft choices for ACL reconstruction include bone–patellar tendon–bone, fascia

lata, and hamstring autografts, as well as bone–patellar tendon–bone and Achilles tendon

allografts. Autogenous bone-patellar tendon-bone grafts combine several advantages such

as greater fixation strength afforded by stable bone to bone healing, superior mechanical

properties, and good long-term results when compared to other grafting techniques 35.

28

Moreover, autogenous tissue avoids the potential risk of disease transmission and immune

rejection associated with allografts. Disadvantages of autogenous bone–patellar tendon–

bone grafts include quadriceps weakness, arthrofibrosis, patellofemoral pain, patellar

tendinitis or rupture, patellar fracture, and patella infera syndrome 36. Recently, there has

been a surge of interest in the use of autogenous hamstring tendon grafts due to

improvements in graft fixation techniques and the preparation of multiple bundle grafts.

Although this graft choice has become more popular, hamstring tendon grafts rely on soft

tissue-to-bone healing, which occurs at a slower rate than bone-to-bone healing 2 seen with

bone plugged tendon autografts. Other disadvantages of hamstring grafts include harvest

site morbidities, such as pain and flexion torque loss.

Allograft tissue is also commonly used in ACL reconstruction. Potential advantages of

allografts include elimination of donor site morbidity, immediate availability of various

graft sizes (off-the-shelf), less postoperative pain, better postoperative function, and lower

incidence of postoperative arthrofibrosis. However, one of the major drawbacks of using

allografts is slow incorporation into host bone and delayed graft remodeling, which may

reduce graft strength and cause early failure 2. Outcomes with allograft transplantation also

depend on the method of sterilization used to prevent disease transmission and

contamination. In the past, the two methods of graft sterilization used were ethylene oxide

treatment or gamma irradiation. Ethylene oxide is no longer used because of associated

synovitis and intra-articular graft destruction. Gamma irradiation is thought to work by

creating free radicals that modify nucleic acids leading to virus and bacterial destruction.

The downside of gamma irradiation, high dose gamma irradiation in particular, is that it

has also been associated with disruption of the structural integrity of the graft 37. Therefore,

29

both sterilization techniques are currently not used on allografts for ACL reconstruction.

With the advent of the freeze-dried and cryopreserved sterilization process of allografts,

initial concerns of graft weakness using the ethylene oxide sterilization process have been

addressed by employing multimodal processes that involve sterile harvesting of tissue from

the donor, rigorous donor screening/testing, antibiotic soaking, low dose irradiation (1.2-

1.8 Mrad), with final preservation techniques such as lyophilization or freezing 35 and good

graft strength and stiffness have been demonstrated using allogenous bone-patellar tendon-

bone grafts.3 More recently, a low-temperature, chemical sterilization process has been

used to effectively remove endogenous donor material such as blood and lipids and

eliminate bacteria, bacterial spores, fungi, and viruses without compromising mechanical

strength.38 Despite these advances in graft sterilization, one of the major drawbacks of

using allografts is slow incorporation into host bone and graft remodeling, which may lead

to reduced graft strength and early failure.2, 3

Despite the successes with both autogenous and allogenous grafts choices, there is no

consensus among orthopaedic surgeons on the “perfect graft” choice, and surgeons

continue to seek alternative reconstruction techniques in order to reduce complications and

improve long-term outcomes 36. Therefore, potential tissue-engineered solutions to

improve ACL reconstruction are of interest to surgeons and represent another significant

clinical application for orthopaedic tissue engineering.

NATURAL ALTERNATIVE GRAFT CHOICES FOR TENDON AND LIGAMENT

RECONSTRUCTION: XENOGRAFTS

30

Xenografts, which are taken from non-human species, are an alternative to traditional grafts

that have been described in the orthopaedic literature. In the 1980s, bovine xenografts

cross-linked with glutaraldehyde were used with moderate success. However, follow-up

studies could not reproduce the promising findings seen with short-term follow-up and

bovine xenografts ultimately failed to obtain Food and Drug Administration (FDA)

approval owing to high failure rates and complications associated with their use. The

mechanism of failure for the bovine-based devices included poor biocompatibility and graft

rejection attributed to excess glutaraldehyde, as well as improper biomechanical properties

pre-implantation, lack of host integration, and recurrent joint effusions 39. More recently, a

case series of 10 patients who underwent ACL reconstruction using processed porcine

bone-patellar tendon-bone xenografts was published that reported encouraging clinical

outcomes at 24 months and concluded that the porcine patellar tendon is an appropriate

graft for reconstructing a human ACL 40. The success of xenografts, however, is limited by

the presence of the alpha-galactosyl carbohydrate on xenografts which humans produce

natural antibodies against. The alpha-gal epitope is abundantly expressed on the cell

surfaces of xenografts derived from non-primate mammals and 1% of naturally circulating

IgG antibodies in all humans will bind the alpha-gal structure 41, 42. The interaction between

alpha-gal and its antibody has been implicated in the hyperacute rejection of transplanted

porcine organs and similarly would lead to immune rejection and ultimate failure of

transplanted musculoskeletal tissue grafts. Various treatment strategies, including alpha-

gal specific enzyme digestion have been unsuccessful at removing enough of the epitope

in order to prevent an immune response in both human and primate models 40, 43. To date,

31

alpha-gal remains an immunological barrier to the clinical application of xenograft

transplantation. Tissue Engineered Grafts: Scaffolds and Beyond

A successful tissue-engineered graft must possess mechanical properties similar to the

native tendon/ligament being replaced until both graft osteointegration and remodeling

have occurred. The “ligamentization” of a tissue-engineered graft is a complex process that

requires high porosity to encourage cell infiltration. After intra-articular implantation, as

with an ACL graft for example, the graft is populated with host cells derived from the

synovial fluid 44. Graft revascularization then occurs predominately from the infrapatellar

fat pad distally and from the posterior synovial tissues proximally 45. Finally, matrix

remodeling occurs, which increases tensile strength 2. Therefore, a successful tissue

engineered ACL graft must serve as a “scaffold” that can temporarily withstand the

mechanical demands placed on a ligament; while simultaneously promoting cell infiltration

to encourage site-specific remodeling that strengthens the graft and enhances

osteointegration in the femoral and tibial bone tunnels. Such tissue engineered scaffolds

can be derived from synthetic materials or natural biologic materials.

A. Synthetic Scaffolds

The use of synthetic ligaments for ACL reconstruction grafts gained popularity in the late

1980s under the false belief that they may possess superior structural properties to

withstand physiologic stresses and strains. 46 Advantages of synthetic grafts include the

elimination of autogenous tissue harvest, donor site morbidity, and risk of disease

transmission. However, synthetic grafts have been associated with a high incidence of

chronic foreign body inflammation, debris-induced synovitis, mechanical limitations, and

32

complete graft rupture 47, 48. The challenge with engineering successful synthetic grafts is

identifying the ideal combination of materials that would provide initial support for the

knee while allowing gradual ingrowth of host cells and graft remodeling. A spectrum of

devices has been created that includes Dacron, Kevlar, and carbon fiber grafts (Table II).

Others include polyethylene terephthalate (Leeds-Keio Ligament) which is a woven porous

tube, polypropylene (Ligament Augmentation Device) which is a diamond braided

cylindrical device and polytetrafluoroethylene (Gore-Tex) which is fibers wound into a

looped braid 49-51. These synthetic ligaments are designed to encourage the ingrowth of

new host tissue to further strengthen and integrate the ligament. Unfortunately, initial

success of these non-degradable synthetic materials could not be maintained in long-term

studies because they frequently failed due to inappropriate mechanical properties, material

fatigue profiles, and material shedding 52-55. Adverse events associated with synthetic grafts

are based on extensive wear as well as foreign-body responses 47. Further investigation

showed that abraded synthetic particles were accumulating in the joint space, lymph nodes,

and regional lymphatic tissues leading to a chronic inflammatory response. 56 Additionally,

these particles also have direct cytotoxic and antimitotic effects on neighboring cells. 57

Biomechanical investigation has shown that synthetic scaffolds do not effectively transmit

physiologic strain to seeded cells, resulting in a loss of the mechanical stimuli which is

essential for stem cell differentiation and ultimately sub-optimal ligament morphology 58-

61. Attention has largely shifted away from synthetic graft substitutes in light of the

consistent evidence supporting superior outcomes with gold standard autograft procedures.

62 A number of the issues associated with synthetic ligaments can be avoided with the use

33

of naturally-derived scaffolds and as a result, they have garnered considerable attention in

the tissue engineering literature.

B. Naturally-Derived Scaffolds

Recently, the focus in tissue engineering has shifted to the use of scaffolds derived from

natural tissues. Naturally-derived scaffolds can be synthesized from biologic elements,

such as reconstituted bovine collagen, or they can be created by decellularization of whole

tendons or ligaments to preserve only the extracellular matrix and structural elements.

Naturally-derived scaffolds are appealing because, compared to synthetic scaffolds; they

more closely mimic the structural, mechanical and biologic properties of a native tissue

and, therefore, are more likely to promote host cell infiltration and remodeling. Less

aggressive decellularization protocols may preserve the native growth factors present in

donor a material, which makes these naturally derived scaffolds more suitable for

regenerative engineering than synthetic materials. Biologic materials are compatible with

host physiology and their degradation will not create the harmful byproducts formed by

degradation of synthetics. To date, decellularized tissues have been successfully used in a

variety of tissue engineering and regenerative medicine applications 63-65. The authors of

this chapter have previously reported on the development of a tendon scaffold derived from

xenograft tendon (chicken flexor digitorum profundus tendon) 17, 66 as well as a scaffold

derived from allograft tendon (human freeze-dried Achilles tendon)67, 68 using a novel

decellularization/oxidation protocol. Both resulting scaffolds showed convincing evidence

of decellularization, were cytocompatible with human cells in vitro, had increased porosity

while maintaining the donor material’s tensile properties, and were able to promote host

34

cell infiltration in vivo using a mouse model. To improve scaffold strength, remodeling and

cell infiltration, cells can be directly seeded into the scaffold and conditioned with a

bioreactor.

C. Cell Seeded Scaffolds and Bioreactors

Several studies have highlighted the deleterious effects of prolonged rest and the beneficial

effects of cyclic loading and activity on the regeneration of injured musculoskeletal tissues

69. Incorporating this knowledge to the field of tissue engineering led to a new evolving

discipline termed “functional tissue engineering” which represents a multidisciplinary

approach using stem cells, biocompatible scaffolds, and mechanical stimulation to produce

constructs that mimic the biomechanical properties of native tissue and have the potential

to revolutionize surgical reconstruction of injured tendons and ligaments70. The utilization

of a bioreactor to apply mechanical strain to a seeded construct has shown to stimulate cell

proliferation, and to increase production of collagen type I and III as well as extracellular

matrix proteins 58, 71-73. Perhaps more importantly, the mechanical stimulation organizes

collagen deposition to model the appropriate architecture needed for sufficient load bearing

capacity. 61, 74 Recently, the authors of this chapter showed that tensile properties of seeded

scaffolds decreased significantly after seeding in the absence of strain 75. This decrease in

tensile properties was temporally associated with a higher level of MMP-2 expression and

higher level of Collagen III expression. MMP-2 is a matrix metalloproteinase that plays a

role in the initial degradation of tensile properties observed in scaffolds and others have

found that after an initial degradation in tensile properties in “under-stimulated” tenocyte

seeded scaffolds, peak stresses can be restored by the addition of protease inhibitors or

35

mechanical loading 76. Collagen III is associated with scar formation during tendon healing

as well as the decreased tensile properties of scar tissue. Therefore, a higher level of

Collagen III expression may represent a shift towards the formation of scar tissue which is

inferior in tensile properties compared to native tissue. Application of cyclic strain to

seeded scaffolds produced constructs that were not significantly different from those of

fresh-frozen tendons emphasizing the importance of mechanical strain on cell-seeded

bioscaffolds to produce a functional tissue engineered construct. Although these studies

have improved our knowledge of ligament tissue engineering, bioreactor conditioned tissue

constructs are yet to be adopted into common clinical orthopaedic practice. Future in vivo

study in larger animal models will be necessary before this strategy can translate to human

use.

SUMMARY AND CONCLUSIONS

The reconstruction of tendon and ligament injuries remains a challenge for orthopaedic

surgeons. Because most of the synthetic grafts that have been developed in the past 30

years are prone to creep, fatigue, and mechanical failure, failure analysis of these grafts has

created the basis for future research. Unfortunately, to date no ideal substitute has been

found that can mimic natural human tissue. Every synthetic graft used has been found to

have drawbacks and many have subsequently required removal from the market, an ill-

fated trend that the Swedish surgeon Einar Eriksson already anticipated in 1976 when he

compared synthetic grafts with “shoestrings, which if used continually, eventually break”.

77 Therefore, it is off critical importance to understand the structure-function relationship

of the tissue that needs to be reconstructed in order to develop a tissue- engineered construct

36

that can adequately restore and maintain normal tissue function. As previously stated, we

believe that an ideal scaffold for tendon and ligament reconstruction would be naturally-

derived from either allogeneic or xenogeneic material.17, 67 Naturally-derived scaffolds

consist of extracellular matrix which serves as a template for cell infiltration,

differentiation, and remodeling. A number of naturally-derived scaffolds consisting of

extracellular matrix are already commercially available for reconstruction of

musculotendinous structures and have been tested clinically with promising results. Future

endeavors will identify additional tissue sources and optimize methods of procurement,

decellularization and preconditioning to successfully replace and reconstruct tendon and

ligament injuries.

37

REFERENCES

1. Butler DL, Awad HA. Perspectives on cell and collagen composites for tendon

repair. Clin Orthop Relat Res. 1999 Oct(367 Suppl):S324-32. Epub 1999/11/05.

2. Jackson DW, Corsetti J, Simon TM. Biologic incorporation of allograft anterior

cruciate ligament replacements. Clin Orthop Relat Res. 1996 Mar(324):126-33. Epub

1996/03/01.

3. Poehling GG, Curl WW, Lee CA, Ginn TA, Rushing JT, Naughton MJ, et al.

Analysis of outcomes of anterior cruciate ligament repair with 5-year follow-up: allograft

versus autograft. Arthroscopy. 2005 Jul;21(7):774-85. Epub 2005/07/14.

4. Rathbone S, Maffulli N, Cartmell SH. Most british surgeons would consider using

a tissue-engineered anterior cruciate ligament: a questionnaire study. Stem Cells Int.

2012;2012:303724. Epub 2012/05/09.

5. Leong NL, Petrigliano FA, McAllister DR. Current tissue engineering strategies in

anterior cruciate ligament reconstruction. J Biomed Mater Res A. 2013 Jun 4. Epub

2013/06/06.

6. Petrigliano FA, McAllister DR, Wu BM. Tissue engineering for anterior cruciate

ligament reconstruction: a review of current strategies. Arthroscopy. 2006 Apr;22(4):441-

51. Epub 2006/04/04.

7. Mody BS, Howard L, Harding ML, Parmar HV, Learmonth DJ. The ABC carbon

and polyester prosthetic ligament for ACL-deficient knees. Early results in 31 cases. J Bone

Joint Surg Br. 1993 Sep;75(5):818-21. Epub 1993/09/01.

38

8. Glousman R, Shields C, Jr., Kerlan R, Jobe F, Lombardo S, Yocum L, et al. Gore-

Tex prosthetic ligament in anterior cruciate deficient knees. Am J Sports Med. 1988 Jul-

Aug;16(4):321-6. Epub 1988/07/01.

9. Barrett GR, Line LL, Jr., Shelton WR, Manning JO, Phelps R. The Dacron ligament

prosthesis in anterior cruciate ligament reconstruction. A four-year review. Am J Sports

Med. 1993 May-Jun;21(3):367-73. Epub 1993/05/01.

10. Macnicol MF, Penny ID, Sheppard L. Early results of the Leeds-Keio anterior

cruciate ligament replacement. J Bone Joint Surg Br. 1991 May;73(3):377-80. Epub

1991/05/01.

11. Girgis FG, Marshall JL, Monajem A. The cruciate ligaments of the knee joint.

Anatomical, functional and experimental analysis. Clin Orthop Relat Res. 1975 Jan-

Feb(106):216-31. Epub 1975/01/01.

12. Legnani C, Ventura A, Terzaghi C, Borgo E, Albisetti W. Anterior cruciate

ligament reconstruction with synthetic grafts. A review of literature. Int Orthop. 2010

Apr;34(4):465-71. Epub 2010/02/17.

13. Mascarenhas R, MacDonald PB. Anterior cruciate ligament reconstruction: a look

at prosthetics--past, present and possible future. Mcgill J Med. 2008 Jan;11(1):29-37. Epub

2008/06/05.

14. Ricchetti ET, Aurora A, Iannotti JP, Derwin KA. Scaffold devices for rotator cuff

repair. J Shoulder Elbow Surg. 2012 Feb;21(2):251-65. Epub 2012/01/17.

15. Iannotti JP, Codsi MJ, Kwon YW, Derwin K, Ciccone J, Brems JJ. Porcine small

intestine submucosa augmentation of surgical repair of chronic two-tendon rotator cuff

39

tears. A randomized, controlled trial. J Bone Joint Surg Am. 2006 Jun;88(6):1238-44. Epub

2006/06/08.

16. Barber FA, Burns JP, Deutsch A, Labbe MR, Litchfield RB. A prospective,

randomized evaluation of acellular human dermal matrix augmentation for arthroscopic

rotator cuff repair. Arthroscopy. 2012 Jan;28(1):8-15. Epub 2011/10/08.

17. Whitlock PW, Smith TL, Poehling GG, Shilt JS, Van Dyke M. A naturally derived,

cytocompatible, and architecturally optimized scaffold for tendon and ligament

regeneration. Biomaterials. 2007 Oct;28(29):4321-9. Epub 2007/07/06.

18. Kennedy JC, Roth JH, Mendenhall HV, Sanford JB. Presidential address.

Intraarticular replacement in the anterior cruciate ligament-deficient knee. Am J Sports

Med. 1980 Jan-Feb;8(1):1-8. Epub 1980/01/01.

19. AAOS. Bone and soft-tissue allografts processing and safety. AAOS Now.

2011;May 2011.

20. Joyce MJ, Greenwald AS, Boden S, Brubaker S, Heim CS. Musculoskeletal

Allograft Tissue Safety. American Academy of Orthopaedic Surgeons Committee on

Biological Implants Tissue Work Group; March 2008; San Francisco CA2008.

21. Schubert T, Bigare E, Van Isacker T, Gigi J, Delloye C, Cornu O. Analysis of

predisposing factors for contamination of bone and tendon allografts. Cell Tissue Bank.

2012 Aug;13(3):421-9. Epub 2012/01/04.

22. Neri BR, Chan KW, Kwon YW. Management of massive and irreparable rotator

cuff tears. J Shoulder Elbow Surg. 2009 Sep-Oct;18(5):808-18. Epub 2009/06/03.

40

23. Bigliani LU, Cordasco FA, McIlveen SJ, Musso ES. Operative treatment of failed

repairs of the rotator cuff. J Bone Joint Surg Am. 1992 Dec;74(10):1505-15. Epub

1992/12/01.

24. Galatz LM, Ball CM, Teefey SA, Middleton WD, Yamaguchi K. The outcome and

repair integrity of completely arthroscopically repaired large and massive rotator cuff tears.

J Bone Joint Surg Am. 2004 Feb;86-A(2):219-24. Epub 2004/02/13.

25. Galatz LM, Griggs S, Cameron BD, Iannotti JP. Prospective longitudinal analysis

of postoperative shoulder function : a ten-year follow-up study of full-thickness rotator cuff

tears. J Bone Joint Surg Am. 2001 Jul;83-A(7):1052-6. Epub 2001/07/14.

26. Oh JH, Kim SH, Ji HM, Jo KH, Bin SW, Gong HS. Prognostic factors affecting

anatomic outcome of rotator cuff repair and correlation with functional outcome.

Arthroscopy. 2009 Jan;25(1):30-9. Epub 2008/12/30.

27. Goutallier D, Postel JM, Gleyze P, Leguilloux P, Van Driessche S. Influence of

cuff muscle fatty degeneration on anatomic and functional outcomes after simple suture of

full-thickness tears. J Shoulder Elbow Surg. 2003 Nov-Dec;12(6):550-4. Epub 2003/12/13.

28. Cole BJ, McCarty LP, 3rd, Kang RW, Alford W, Lewis PB, Hayden JK.

Arthroscopic rotator cuff repair: prospective functional outcome and repair integrity at

minimum 2-year follow-up. J Shoulder Elbow Surg. 2007 Sep-Oct;16(5):579-85. Epub

2007/07/17.

29. Dines JS, Grande DA, Dines DM. Tissue engineering and rotator cuff tendon

healing. J Shoulder Elbow Surg. 2007 Sep-Oct;16(5 Suppl):S204-7. Epub 2007/05/26.

41

30. Ide J, Kikukawa K, Hirose J, Iyama K, Sakamoto H, Mizuta H. Reconstruction of

large rotator-cuff tears with acellular dermal matrix grafts in rats. J Shoulder Elbow Surg.

2009 Mar-Apr;18(2):288-95. Epub 2008/12/09.

31. Gimbel JA, Van Kleunen JP, Lake SP, Williams GR, Soslowsky LJ. The role of

repair tension on tendon to bone healing in an animal model of chronic rotator cuff tears. J

Biomech. 2007;40(3):561-8. Epub 2006/04/08.

32. Aurora A, McCarron JA, van den Bogert AJ, Gatica JE, Iannotti JP, Derwin KA.

The biomechanical role of scaffolds in augmented rotator cuff tendon repairs. J Shoulder

Elbow Surg. 2012 Aug;21(8):1064-71. Epub 2011/09/03.

33. Mannava S, Plate JF, Whitlock PW, Callahan MF, Seyler TM, Koman LA, et al.

Evaluation of in vivo rotator cuff muscle function after acute and chronic detachment of

the supraspinatus tendon: an experimental study in an animal model. J Bone Joint Surg

Am. 2011 Sep 21;93(18):1702-11. Epub 2011/09/23.

34. Gottlob CA, Baker CL, Jr., Pellissier JM, Colvin L. Cost effectiveness of anterior

cruciate ligament reconstruction in young adults. Clin Orthop Relat Res. 1999

Oct(367):272-82. Epub 1999/11/05.

35. Chang SK, Egami DK, Shaieb MD, Kan DM, Richardson AB. Anterior cruciate

ligament reconstruction: allograft versus autograft. Arthroscopy. 2003 May-Jun;19(5):453-

62. Epub 2003/05/02.

36. Sherman OH, Banffy MB. Anterior cruciate ligament reconstruction: which graft

is best? Arthroscopy. 2004 Nov;20(9):974-80. Epub 2004/11/05.

42

37. Vangsness CT, Jr., Garcia IA, Mills CR, Kainer MA, Roberts MR, Moore TM.

Allograft transplantation in the knee: tissue regulation, procurement, processing, and

sterilization. Am J Sports Med. 2003 May-Jun;31(3):474-81. Epub 2003/05/17.

38. Jones DB, Huddleston PM, Zobitz ME, Stuart MJ. Mechanical properties of

patellar tendon allografts subjected to chemical sterilization. Arthroscopy. 2007

Apr;23(4):400-4. Epub 2007/04/10.

39. Vunjak-Novakovic G, Altman G, Horan R, Kaplan DL. Tissue engineering of

ligaments. Annu Rev Biomed Eng. 2004;6:131-56. Epub 2004/07/17.

40. Stone KR, Abdel-Motal UM, Walgenbach AW, Turek TJ, Galili U. Replacement

of human anterior cruciate ligaments with pig ligaments: a model for anti-non-gal antibody

response in long-term xenotransplantation. Transplantation. 2007 Jan 27;83(2):211-9.

Epub 2007/02/01.

41. Galili U. The alpha-gal epitope and the anti-Gal antibody in xenotransplantation

and in cancer immunotherapy. Immunol Cell Biol. 2005 Dec;83(6):674-86. Epub

2005/11/04.

42. Galili U. Significance of anti-Gal IgG in chronic xenograft rejection. Transplant

Proc. 1999 Feb-Mar;31(1-2):940-1. Epub 1999/03/20.

43. Stone KR, Walgenbach AW, Turek TJ, Somers DL, Wicomb W, Galili U. Anterior

cruciate ligament reconstruction with a porcine xenograft: a serologic, histologic, and

biomechanical study in primates. Arthroscopy. 2007 Apr;23(4):411-9. Epub 2007/04/10.

44. Min BH, Han MS, Woo JI, Park HJ, Park SR. The origin of cells that repopulate

patellar tendons used for reconstructing anterior cruciate ligaments in man. J Bone Joint

Surg Br. 2003 Jul;85(5):753-7. Epub 2003/08/02.

43

45. Nikolaou PK, Seaber AV, Glisson RR, Ribbeck BM, Bassett FH, 3rd. Anterior

cruciate ligament allograft transplantation. Long-term function, histology,

revascularization, and operative technique. Am J Sports Med. 1986 Sep-Oct;14(5):348-60.

Epub 1986/09/01.

46. Schindler OS. Surgery for anterior cruciate ligament deficiency: a historical

perspective. Knee Surg Sports Traumatol Arthrosc. 2012 Jan;20(1):5-47. Epub

2011/11/23.

47. Olson EJ, Kang JD, Fu FH, Georgescu HI, Mason GC, Evans CH. The biochemical

and histological effects of artificial ligament wear particles: in vitro and in vivo studies.

Am J Sports Med. 1988 Nov-Dec;16(6):558-70. Epub 1988/11/01.

48. Frank CB, Jackson DW. The science of reconstruction of the anterior cruciate

ligament. J Bone Joint Surg Am. 1997 Oct;79(10):1556-76. Epub 1997/10/31.

49. Laurencin CT, Ambrosio AM, Borden MD, Cooper JA, Jr. Tissue engineering:

orthopedic applications. Annu Rev Biomed Eng. 1999;1:19-46. Epub 2001/11/10.

50. Laurencin CT, Freeman JW. Ligament tissue engineering: an evolutionary

materials science approach. Biomaterials. 2005 Dec;26(36):7530-6. Epub 2005/07/28.

51. Laurencin CT, Khan Y, Kofron M, El-Amin S, Botchwey E, Yu X, et al. The ABJS

Nicolas Andry Award: Tissue engineering of bone and ligament: a 15-year perspective.

Clin Orthop Relat Res. 2006 Jun;447:221-36. Epub 2006/06/03.

52. Bolton CW, Bruchman WC. The GORE-TEX expanded polytetrafluoroethylene

prosthetic ligament. An in vitro and in vivo evaluation. Clin Orthop Relat Res. 1985

Jun(196):202-13. Epub 1985/06/01.

44

53. Lukianov AV, Richmond JC, Barrett GR, Gillquist J. A multicenter study on the

results of anterior cruciate ligament reconstruction using a Dacron ligament prosthesis in

"salvage" cases. Am J Sports Med. 1989 May-Jun;17(3):380-5; discussion 5-6. Epub

1989/05/01.

54. Schindhelm K, Rogers GJ, Milthorpe BK, Hall PJ, Howlett CR, Sekel R, et al.

Autograft and Leeds-Keio reconstructions of the ovine anterior cruciate ligament. Clin

Orthop Relat Res. 1991 Jun(267):278-93. Epub 1991/06/01.

55. Weiss AB, Blazina ME, Goldstein AR, Alexander H. Ligament replacement with

an absorbable copolymer carbon fiber scaffold--early clinical experience. Clin Orthop

Relat Res. 1985 Jun(196):77-85. Epub 1985/06/01.

56. Margevicius KJ, Claes LE, Durselen L, Hanselmann K. Identification and

distribution of synthetic ligament wear particles in sheep. J Biomed Mater Res. 1996

Jul;31(3):319-28. Epub 1996/07/01.

57. Garvin J, Qi J, Maloney M, Banes AJ. Novel system for engineering bioartificial

tendons and application of mechanical load. Tissue Eng. 2003 Oct;9(5):967-79. Epub

2003/11/25.

58. Altman GH, Horan RL, Martin I, Farhadi J, Stark PR, Volloch V, et al. Cell

differentiation by mechanical stress. FASEB J. 2002 Feb;16(2):270-2. Epub 2002/01/05.

59. Butler DL, Goldstein SA, Guilak F. Functional tissue engineering: the role of

biomechanics. J Biomech Eng. 2000 Dec;122(6):570-5. Epub 2001/02/24.

60. Shah N, Morsi Y, Manasseh R. From mechanical stimulation to biological

pathways in the regulation of stem cell fate. Cell Biochem Funct. 2014 Feb 27. Epub

2014/02/28.

45

61. Rodrigues MT, Reis RL, Gomes ME. Engineering tendon and ligament tissues:

present developments towards successful clinical products. J Tissue Eng Regen Med. 2013

Sep;7(9):673-86. Epub 2012/04/14.

62. Romanini E, D'Angelo F, De Masi S, Adriani E, Magaletti M, Lacorte E, et al. Graft

selection in arthroscopic anterior cruciate ligament reconstruction. J Orthop Traumatol.

2010 Dec;11(4):211-9. Epub 2010/12/25.

63. Tischer T, Aryee S, Wexel G, Steinhauser E, Adamczyk C, Eichhorn S, et al. Tissue

engineering of the anterior cruciate ligament-sodium dodecyl sulfate-acellularized and

revitalized tendons are inferior to native tendons. Tissue Eng Part A. 2010 Mar;16(3):1031-

40. Epub 2009/10/23.

64. Lin VS, Lee MC, O'Neal S, McKean J, Sung KL. Ligament tissue engineering using

synthetic biodegradable fiber scaffolds. Tissue Eng. 1999 Oct;5(5):443-52. Epub

1999/12/10.

65. Gentleman E, Livesay GA, Dee KC, Nauman EA. Development of ligament-like

structural organization and properties in cell-seeded collagen scaffolds in vitro. Ann

Biomed Eng. 2006 May;34(5):726-36. Epub 2006/02/08.

66. Whitlock PW, Van Dyke M, Poehling GG, Smith TL, Marker DR, Koman LA, et

al. Functional Tissue Engineering of Tendons and Ligaments. American Academy of

Orthopaedic Surgeons Annual Meeting; 2009; Las Vegas NV2009.

67. Whitlock PW, Seyler TM, Parks GD, Ornelles DA, Smith TL, Van Dyke ME, et al.

A novel process for optimizing musculoskeletal allograft tissue to improve safety,

ultrastructural properties, and cell infiltration. J Bone Joint Surg Am. 2012 Aug

15;94(16):1458-67. Epub 2012/07/13.

46

68. Whitlock PW, Smith TL, Shilt JS, Van Dyke ME, Poehling GG. A Novel Scaffold

for Tendon and Ligament Regeneration Derived from Human Allograft Tissue. Combined

Meeting of the International Orthopaedic Research Societies; October 2007; Honolulu

HI2007.

69. Buckwalter JA, Grodzinsky AJ. Loading of healing bone, fibrous tissue, and

muscle: implications for orthopaedic practice. J Am Acad Orthop Surg. 1999 Sep-

Oct;7(5):291-9. Epub 1999/10/03.

70. Butler DL, Juncosa-Melvin N, Boivin GP, Galloway MT, Shearn JT, Gooch C, et

al. Functional tissue engineering for tendon repair: A multidisciplinary strategy using

mesenchymal stem cells, bioscaffolds, and mechanical stimulation. J Orthop Res. 2008

Jan;26(1):1-9. Epub 2007/08/07.

71. Webb K, Hitchcock RW, Smeal RM, Li W, Gray SD, Tresco PA. Cyclic strain

increases fibroblast proliferation, matrix accumulation, and elastic modulus of fibroblast-

seeded polyurethane constructs. J Biomech. 2006;39(6):1136-44. Epub 2005/11/01.

72. Hohlrieder M, Teuschl AH, Cicha K, van Griensven M, Redl H, Stampfl J.

Bioreactor and scaffold design for the mechanical stimulation of anterior cruciate ligament

grafts. Biomed Mater Eng. 2013;23(3):225-37. Epub 2013/05/01.

73. Wang T, Gardiner BS, Lin Z, Rubenson J, Kirk TB, Wang A, et al. Bioreactor

design for tendon/ligament engineering. Tissue Eng Part B Rev. 2013 Apr;19(2):133-46.

Epub 2012/10/18.

74. Yilgor C, Yilgor Huri P, Huri G. Tissue engineering strategies in ligament

regeneration. Stem Cells Int. 2012;2012:374676. Epub 2012/01/14.

47

75. Whitlock PW, Seyler TM, Northam C, Smith TL, Van Dyke ME, Poehling GG, et

al. The Effect of Cyclic Strain on the Tensile Properties of a Naturally-Derived,

Decellularized Tendon Scaffold Seeded with Allogeneic Tenocytes and Associated mRNA

Expression. J Surg Orthop Adv. 2012;Accepted for publication.

76. Cousineau-Pelletier P, Langelier E. Relative contributions of mechanical

degradation, enzymatic degradation, and repair of the extracellular matrix on the response

of tendons when subjected to under- and over- mechanical stimulations in vitro. J Orthop

Res. 2010 Feb;28(2):204-10. Epub 2009/09/03.

77. Eriksson E. Sports injuries of the knee ligaments: their diagnosis, treatment,

rehabilitation, and prevention. Med Sci Sports. 1976 Fall;8(3):133-44. Epub 1976/01/01.

48

FIGURES AND TABLES

Figure 1. Number of Pubmed listed publications on the topic of tissue engineering of ligaments. Search conducted on 04/24/2013 using the keywords “tissue engineering” and “ligament”. Thirteen publications found for the year 2013 so far which represents last data point in graph. Increased activity noted since 1999/2000.

49

Table I. Augmentation Devices for Rotator Cuff Repair

Name Material Source Comments Distributed by

Restore Small Intestinal Submucosa Porcine DePuy Orthopaedics CuffPatch Small Intestinal Submucosa Porcine cross-linked Biomet/Organogenesis GraftJacket Dermis Human Wright Medical Technology

ArthroFlex Dermis Human Arthrex Conexa Dermis Porcine α-Gal

reduced Tornier

TissueMend Dermis Bovine fetal Stryker/TEI Biosciences Zimmer Collagen Repair (Permacol) Patch

Dermis Porcine cross-linked Zimmer/Tissue Science Laboratories

Bio-Blanket Dermis Bovine cross-linked Kensay Nash Allopatch HD Fascia lata Human Musculoskeletal Tissue Foundation

(MTF)

Gore Dualmesh Polytetrafluoroethylene (PTFE) Synthetic Gore Medical Bard PTFE Felt Pledgets

Polytetrafluoroethylene (PTFE) Synthetic CR Bard

SportMesh Poly(urethaneurea) Synthetic Biomet/Arthrotek X-Repair Poly-L-lactide Synthetic Synthasome Biomerix RCR Patch Polycarbonate

poly(urethaneurea) Synthetic Biomerix

OrthADAPT PR Bioimplant

Pericardium with woven polymer

Equine/Synthetic cross-linked Pegasus Biologics

50

Table II. Prosthetics and Augmentation Devices for ACL Reconstruction

Name Material Disadvantage Proplast polytetrafluoroethylene (PTFE) with

embedded carbon or aluminum oxide fibers high rupture rate due to inadquate mechanical properties, inflammatory reaction

Polyflex ultra-high-polymerized polyethylene high rupture rate due to inadquate mechanical properties, inflammatory reaction

Dacron polyethylene terephthalate (PET) Poor long-term stability Ligament Augmentation Device (LAD) braided polypropylene Weak implant-graft interface, tendency to

stretch out and cause instability, inflammatory response, and synovitis and effusions

Gore-Tex braided bundles of polytetrafluoroethylene (PTFE)

Immunological response to wear particles, progressive long-term loosening

ABC prosthetic ACL carbon and polyester fibres oriented in a partial braid by a zig-zag pattern

high failure rate due to abrasion of the ligament at the tibial tunnel

Leeds-Keio ligament polyethylene terephthalate mesh Long-term failures, poor instability Integraft carbon fibers Poor shear stress behaviour, high failure

rates, inflammatory response

Ligament Advanced Reinforcement System (LARS)

polyethylene terephthalate (PET) Laxity, rupture risk, poor long-term stability

Trevira-hochfest ligament polyethylene terephthalate (PET) inadequate resistance to abrasion and torsion forces, long-term failure

51

CHAPTER 3

A NOVEL PROCESS FOR OPTIMIZING MUSCULOSKELETAL ALLOGRAFT

TISSUE: IMPROVING SAFETY, ULTRA-STRUCTURAL PROPERTIES AND

CELL INFILTRATION.

Patrick W. Whitlock, M.D., Ph.D.1,2;Thorsten M. Seyler, M.D.1,2; Griffith D. Parks,

Ph.D.3; David A. Ornelles, Ph.D.3; Thomas L. Smith, Ph.D.1,2; Mark E. Van Dyke, Ph.D.

1,2; Gary G. Poehling, M.D.1,2

1Department of Orthopaedic Surgery, Wake Forest University Health Sciences, Medical

Center Boulevard, Winston-Salem, North Carolina, 27157

2Institute for Regenerative Medicine, Wake Forest University Health Sciences, Medical

Center Boulevard, Winston-Salem, North Carolina, 27157

3Department of Microbiology and Immunology, Wake Forest University Health Sciences,

Medical Center Boulevard, Winston-Salem, North Carolina, 27157

52

The following manuscript was published in the Journal of Bone and Joint Surgery

American Volume on August 15th 2012. This chapter is reprinted with permission. Stylistic

variations are due to the requirements of the journal. Thorsten M. Seyler made substantial

contributions to the conception of the work and all experiments. Participated in the

acquisition, analysis, or interpretation of data for all experiments. Drafting the work and

revising it critically for important intellectual content including final approval of the

version to be published. Drs. Whitlock and Seyler are accountable for all aspects of the

work in ensuring that questions related to the accuracy or integrity of any part of the work

are appropriately investigated and resolved.

53

ABSTRACT

Background: This study evaluated the properties of a freeze-dried human Achilles tendon

allograft (hFDATA) - derived scaffold for use in ACL reconstruction. Our hypothesis was

that hFDATAs could be processed using a method to remove cellular and infectious

material to produce a cyto-compatible, architecturally- modified scaffold possessing tensile

properties suitable for ACL reconstruction.

Methods: Fifty-two hFDATAs were provided by a tissue bank. Twenty-one were used as

controls to assess cellularity, DNA content, micro-architecture, porosity, cyto-

compatibility, and tensile properties in vitro (n=13) and in vivo (n=8). Thirty-one were

processed to produce scaffolds that were similarly assessed for those properties in vitro

(n=23) and in vivo (n=8). The elimination of “spiked” non-enveloped and enveloped virus

was also determined after each processing step in vitro.

Results: Cellularity and DNA content were decreased significantly in hFDATA-derived

scaffolds. The scaffolds’ porosity increased significantly, and they were cyto-compatible

in vitro. Processing resulted in significantly increased percent elongation (138.47% of that

observed for hFDATAs) of scaffolds. No other significant differences in tensile properties

were observed in vitro or in vivo. The number of infiltrating host cells and the depth to

which those cells infiltrated was significantly greater in the scaffolds. No enveloped viruses

and only two of 108 non-enveloped viral particles were detected in the scaffolds after

processing equating to a sterility assurance level (SAL) of 0.2 x 10-7.

Conclusions: hFDATAs were processed using a method that removed cellular and

infectious material to produce a decellularized, cyto-compatible, architecturally-modified

scaffold with tensile properties that differed minimally from human allograft tissue both in

54

vitro and in vivo. The process also resulted in an increase in scaffold porosity that led to an

increase in cell infiltration in vivo.

Level of evidence: Basic science study

Clinical Relevance: hFDATA-derived scaffolds have the potential to eliminate disease

transmission and inflammation in recipients and to promote early and improved cell

infiltration while retaining the initial tensile properties necessary to withstand

rehabilitation after implantation.

55

INTRODUCTION

The anterior cruciate ligament (ACL) is a commonly injured knee ligament;

400,000 patients undergo reconstruction annually.1-3 Graft choices for ACL reconstruction

include autograft and allografts, with 60,000 allograft reconstructions annually.2,3

Allografts and autografts demonstrate remodeling following ACL reconstruction; a process

termed “ligamentization”, ideally results in a tissue that is histologically and biochemically

similar to native ACL.4-8

Synthetic and naturally-derived ACL repair scaffolds are associated with a high

incidence of chronic foreign body inflammation, debris-induced synovitis, mechanical

limitations, and graft failure.9-11 Tissue-engineered strategies for ACL reconstruction rely

predominantly upon biodegradable polymeric scaffolds. However, current biodegradable

scaffolds do not initially possess the mechanical properties necessary for reconstruction

and are associated with potential release of toxic byproducts during degradation.12-14 The

focus has shifted to the use of naturally derived scaffolds and alternative materials for ACL

reconstruction.15,16 Naturally-derived scaffolds combine important advantages including:

decreased operative time and patient morbidity, established fixation methods,

biocompatibility, preservation of extracellular protein matrix, and biodegradation allowing

for maintenance of tensile properties. Disadvantages include potential transmission from

xenograft and allograft source material, dense micro-architecture delaying or inhibiting

remodeling of implanted grafts, immunogenic cellular epitopes such as alpha-galactosyl

(xenografts), and residual donor antigenic material within the scaffold (e.g. DNA), which

may promote sterile inflammation and scaffold failure.12-14

56

Decellularized tissues have been used in many orthopaedic applications.17-19 However,

prior attempts to produce decellularized tissue grafts investigated the histological

elimination of donor cells and sterilization of donor tissue and have not addressed the dense

micro-architecture of the donor tissue or the complete elimination of donor cellular

material. Limited success or failure of those grafts was partly associated with undetectable

residual cellular material in the dense micro-architecture of the donor tissue.17-19 Recently,

successful methods for producing a decellularized, architecturally-modified scaffold

derived from tendon with minimal residual cellular material in the resulting scaffold have

been reported.20

The successful ACL reconstruction with allografts is well-documented.3,21,22 This

study evaluated the biologic and biomechanical properties of an architecturally-modified,

freeze-dried human Achilles tendon allograft (hFDATA) - derived scaffold for use in ACL

reconstruction. We hypothesized that hFDATAs could be processed to remove cellular and

infectious material, producing decellularized, cyto-compatible, architecturally-modified

scaffolds that promoted rapid cell infiltration in vivo, and possessed tensile properties in

vitro and in vivo similar to a tissue currently used for ACL reconstruction, hFDATA.

57

MATERIALS & METHODS

Preparation of hFDATA-derived Scaffolds

Fifty-two human freeze-dried Achilles tendon allografts (hFDATAs) (MTF™)

were prepared as previously described, Appendix A. 20 hFDATAs were washed in aqueous,

hypotonic solutions followed by trypsin digestion and a combined oxidative (peracetic

acid)-detergent (Triton X-100™) step. The products, referred to as scaffolds, were rinsed,

and freeze-dried, and stored at -80°C until further use (Figure 1).

Histologic Analysis of hFDATAs and Scaffolds

Mid-substance portions of the hFDATAs (n=10) and scaffolds (n=10) were fixed,

processed and embedded for histology. Sections were stained using hematoxylin and eosin

(H&E, Sigma) or 4,6-diamidino-2-phenylindole (DAPI, Vector) to identify cellular and

nuclear components, respectively. Representative light (H&E) and fluorescence (DAPI)

micrographs were taken at 200X magnification.

DNA Content of hFDATAs and Scaffolds

Samples of hFDATAs (n=10) and scaffolds (n=10) were weighed and placed into

sterile 1.5 ml micro-centrifuge tubes. Total DNA was isolated from both samples using

DNeasy™ (Qiagen). Total DNA content at λ=280 nm was calculated using a

spectrophotometer (Thermo Spectronic), and normalized to the samples’ initial dry weight.

Scanning Electron Microscopy (SEM) of hFDATAs and Scaffolds

Specimens of hFDATAs (n=10) and scaffolds (n=10) were processed and cross-

sectional SEMs were obtained at 15.0 kV, 50 Pa, 150X magnification (Hitachi S-2600

Scanning Electron Microscope, Hitachi High Technologies America).

58

Mercury Intrusion Porosimetry of hFDATAs and Scaffolds

Samples of hFDATAs (n=3) and scaffolds (n=3) were weighed and placed into a

penetrometer, which was evacuated, and filled with mercury (initial pressure of ~30 psi)

(Micromeritics Instrument Corporation). The penetrometer was transferred into a high

pressure chamber (50,000 psi) while the intruded volume was recorded. The intruded

mercury volume per gram sample was measured by the porosimeter and was assumed to

be equal to pore volume (Vpore). Porosity (ε) was calculated using the formula: % porosity

= (1- envelope density/skeletal density)*100.

Residual Peracetic Acid Analysis

Commercially available peracetic acid (PAA) test strips (EMD Chemicals Inc.)

measured PAA concentration in the initial processing solution and after each wash step at

24h, 48h, and 72h in parts per million (ppm), (n=10).

In Vitro Cyto-compatibility of Scaffolds: Direct Contact Method

Scaffolds (n=10) were removed from storage and placed into 70% ethanol for 12

hours at 4°C. Scaffolds were removed and washed briefly three times in 50 ml of cell

culture media containing antibiotic-antimycotic (Sigma) before the MTS® (Promega) and

neutral red assays. Sterile latex fragments, the negative control (n=10), were treated

similarly. Cell proliferation/metabolic activity (MTS®) and cell viability (neutral red

release/uptake assay) of NIH 3T3 cells were determined as previously described. 20,23,24

Assessment of Virus-inactivating Efficacy During Production of Scaffolds

hFDATAs were recovered from disease-free donors under aseptic conditions (n=4).

The tendon central portion was cut into 1cm x 2cm sections and immersed in 20mL of

DMEM (Gibco) supplemented with 2% FBS (Valley Labs). Three tissue sections from

59

each donor were spiked with 108 Plaque Forming Units (PFU) of Vesicular Stomatitis virus

(VSV, enveloped), Simian virus 5 (SV5, enveloped), or adenovirus (Ad, non-enveloped).

Spiked fragments were incubated on a shaker (160rpm) at 37°C for 12 hours. After

incubation, remaining virus titers were determined at four different steps in the scaffold

production process by standard viral plaque assays on CV-1 (SV5 and VSV) or 293

(Adenovirus) cell monolayers to determine the virus-inactivating efficacy of each

processing step.

Tensile Testing of hFDATAs and Scaffolds

hFDATAs (n=10) and scaffolds (n=10) were equilibrated in sterile Dulbecco’s

Phosphate Buffered Saline (DPBS, Gibco) at 37°C for 30 minutes. Specimens were cut

using a custom “dogbone-type” punch (Freeman™) approximating ASTM™ standards.

The punch measurements included an outer length of 40 mm, inner length of 20 mm x outer

width of 8 mm, inner width of 4 mm x measured thickness. The outer width (8 mm) tapered

at the sample neck to the inner width (4 mm) to form the desired specimen shape or “dog-

bone”. Each tested specimen was individually measured with digital calipers; the length,

width, thickness and cross-sectional area of each were recorded. The specimens were

mounted in custom, serrated pneumatic clamps on a uniaxial load frame (Instron™ 5544)

utilizing a 2kN static load cell (Instron™) with an accuracy of 0.5% of the tested specimen

to 1/500th of load cell capacity. Tendons were pre-loaded to 0.1N, pre-conditioned 10

times to 0.1mm extension, and loaded to failure at a rate of 10 mm/minute (strain rate of

1.11%/second). Tendons were kept moist during testing with DPBS mist. Ultimate tensile

load (UTL, N) was the maximal load recorded during each test. Ultimate tensile stress

(UTS, MPa) at break was the maximal load recorded during each test normalized to

60

specimen cross-sectional area. The percent strain at UTS was calculated using the

displacement data and initial specimen length measured with digital calipers after

placement of samples in the test grips. Elastic modulus (E, MPa) and stiffness (N/mm)

were calculated from the linear portion of the stress–strain curve and from load-elongation

data, respectively.25-30

In Vivo Implantation of hFDATAs and Scaffolds

The Institutional Animal Care and Use Committee approved the animal protocol.

Sixteen skeletally mature New Zealand White rabbits (28-32 weeks old, 3.5-4 kg weight)

were used for studying tensile properties and histology of ACL reconstruction in an

established animal model.31 Rabbits were assigned to 1: ACL reconstruction using the

hFDATAs (n=8); or 2: ACL reconstruction using the scaffolds (n=8). Anesthesia was

induced by 35 mg/kg ketamine, supplemented by 5% of isoflurane. Anesthesia was

maintained by 1.5-2% of isoflurane and oxygen. Intramuscular enrofloxacin (10 mg/kg)

was used for antibiotic prophylaxis. Surgery was performed using aseptic technique. A

midline skin incision, followed by a lateral parapatellar arthrotomy exposed the right knee.

The patella was dislocated medially with the knee extended, and native ACL was resected

by sharp dissection. The tibial bone tunnel was created in a retrograde fashion using a

Micro VECTOR™ Drill Guide System (Smith & Nephew, Andover, Massachusetts, USA)

with a 3.2 mm cannulated bullet and a 3.2 mm drill necked down to 3.0 mm distally with

the transtibial angle set at 55°. The femoral tunnel was created from the insertion site of

the native ACL in the femoral notch to the lateral femoral condyle at the 10-o'clock position

using a free-hand surgical technique. The final graft was prepared by trimming the

midportion of either an hFDATA or scaffold using a #15 blade. The midportion was

61

prepared to a diameter of 3 mm and a length of 50 mm. Both ends of the graft were secured

with a Vicryl stitch. The proximal Vicryl stitch was passed through the lumen of a large

straight needle and the graft was passed through the tibial and femoral bone tunnels, leaving

the proximal stitch within the femoral tunnel. The proximal Vicryl stitch was used to pull

the graft into final position and to ensure a snug fit within the bone tunnels. Adequate

exposure for placement of the femoral holding stitch, was obtained by dissection to expose

the lateral femoral condyle through the existing midline. After passing the graft, the knee

joint was moved through 5-10 cycles of full motion before both ends of the graft were

secured under slight tension in 30-35° of flexion to the periosteum using 3-0 Prolene suture

(Ethicon). The deep layers of the wound were closed in a using 4-0 Vicryl, and the skin

was closed using a 4-0 nylon (Ethicon) subcuticular suture and tissue glue. Postoperatively,

rabbits moved freely in individual cages and were assessed daily for pain. Rabbits were

given 0.05 mg/kg of buprenorphine intramuscularly every six to eight hours for three to

five days after surgery to control pain. At twelve weeks, the animals were sacrificed by

intracardiac injection of pentobarbital Na.

Tensile Testing of Implanted hFDATAs and Scaffolds

Ten rabbit knees implanted with either hFDATA (n=5) or scaffolds (n=5) were used

for tensile analysis.31,32 The right hind limbs were disarticulated at the hip and stored at –

20°C until testing. Before testing, the limbs were thawed, and all extraneous soft tissues

removed. The femur-ACL graft-tibia complexes were potted using Bondo epoxy (Mar-

Hyde Corp.). These were fixed in clamps with the femur and tibia oriented at angles of 45°

and 30°, respectively. This preparation allowed tensile loading along the long axis of the

graft in the material testing system (Instron model 5544, 2kN static load cell). A preload of

62

0.5 N was applied for 60 seconds to determine the resting gauge length of the graft and the

cross-sectional area of the midsubstance using digital calipers, followed by cyclic

preconditioning of the constructs from 0 to 0.3 mm elongation (10 mm/min) to achieve a

steady state. Load-to-failure testing was performed by increasing the tensile load

continuously at a rate of 10 mm/min.31,32 Ultimate tensile load (UTL, N), ultimate tensile

stress (UTS, MPa) at break, % strain at UTS, elastic modulus (E, MPa) and stiffness

(N/mm) were calculated as previously described.25-30

Histology of Implanted Scaffolds

Six rabbit knees, implanted with either the hFDATAs (n=3) or scaffolds (n=3) were

processed for histological analysis. The harvested femur-ACL graft-tibia complexes were

fixed in 10% neutral buffered formalin, decalcified in formic acid/Immunocal (Decal

Chemical), dehydrated with ethanol and embedded in paraffin. Consecutive 5 µm-thick

sections cut perpendicular to the tunnel axis and the intra-articular portion of the graft were

stained with H&E and Masson’s trichrome stain. Histological analysis (50X and 200X

magnification) of the bone–graft interface was performed using three sections from each

specimen to assess fibrocartilage formation, new bone formation, graft bonding to adjacent

tissue, and cell penetration depth and number of cells within the graft. Two reviewers

blinded to the group assignments scored each characteristic according to a previously

published scoring system.33

Statistical Methods

For non-parametric data, the Mann-Whitney U Test was used for analysis. For in

vitro experiments (MTS, neutral red, DNA content, residual PAA) data were analyzed in

triplicate. All numerical data were averaged, a standard error of the mean (SEM) was

63

calculated, and a comparison was made between appropriate groups using a two-tailed,

Student’s t-test with unequal variances. Differences were considered significant at p<0.05.

Source of Funding

This study was supported by a research grant from AANA to PWW and GGP and

grants from OREF and ANNA to TMS, PWW and GGP. hFDATAs were supplied by

MTF™.

64

RESULTS

Histologic Analysis of hFDATAs and Scaffolds

Abundant nuclear material (DNA/RNA) was evident in longitudinal sections of

hFDATAs after H&E (Figure 2A) and DAPI staining (Figure 2B). Minimal inter-fascicular

and intra-fascicular space was present in the H&E-stained sections of hFDATAs prior to

processing (Figure 2A). After processing, no nuclear material was evident via H&E

staining of the scaffolds (Figure 2C). DAPI staining revealed no DNA or RNA within the

scaffolds (Figure 2D). After processing, a subjective increase in intra-fascicular and inter-

fascicular space was observed in the scaffolds via H&E staining (Figure 2C)

(magnification, 200 X).

DNA Content of hFDATAs and Scaffolds

Scaffold DNA content was significantly decreased by 67.33% (0.11 + 0.07 μg

DNA/mg tissue dry weight, n=10) after processing when compared to the untreated

hFDATAs (0.31 + 0.03 µg DNA/mg tissue dry weight, n=10), p=0.00003.

Scanning Electron Microscopy (SEM) of hFDATAs and Scaffolds

SEM confirmed the dense micro-architecture observed in sections of the

hFDATAs, (Figure 3A), and the increase in pore size and porosity of the scaffolds, (Figure

3B) (magnification, 150 X).

Mercury Intrusion Porosimetry of hFDATAs and Scaffolds

Mean total porosity measured by intrusion porosimetry was 49.63% ± 0.57 for the

hFDATAs (n=3) and 87.50% ± 8.69 for the scaffolds (n=3), p = 0.008. While the scaffolds

had median pore size of 17.54 ± 3.73 μm, median pore size of the hFDATAs was 13.23 ±

3.20 μm, p=0.204. The scaffold mean pore size was 14.02 ± 3.06 μm, and the hFDATA

65

specimens was 9.99 ± 2.47 μm, p=0.151. Although there was a trend towards increased

pore size in the scaffolds, this difference was not significant.

Residual Peracetic Acid Analysis

The residual peracetic acid (PAA) concentration in the initial solution and the 24h

wash was greater than 50 ppm, the limit of detection of the test strips. PAA concentration

was 7.92 ± 1.89 ppm after the 48h wash and 0.42 ± 1.20 ppm after the 72h wash (n=10).

Only one scaffold (1/12, 8.5%) had residual PAA at the detectable limit (5.0 ppm) after the

fourth and final wash.

In Vitro Cyto-compatibility of Scaffolds

Direct Contact Method Metabolic activity determined using the MTS assay

(absorbance at λ=490 ηm) for NIH 3T3 cells exposed to the scaffolds was 93.71% (1.01

+/- 0.04, n=10) of that observed for cells exposed to media only (1.08 +/- 0.07, n=20,

positive control) and was not statistically significant, p=0.36, (Figure 4A). Cell viability

determined using the neutral red release/uptake assay (absorbance at λ=540 ηm) for NIH

3T3 cells exposed to the scaffolds was 102.13% (0.19 +/- 0.02, n=10) of that observed for

NIH 3T3 cells exposed to media alone (0.17 +/- 0.02, n=10), a difference which was not

statistically significant, p=0.60, (Figure 4B). The cells exposed to the scaffolds and the

positive control (cells exposed to culture media only) differed significantly from the values

obtained for the latex negative control (n=10) in both assays, p<0.0001. The absorbance

observed for the negative control was also <10% of the absorbance observed for positive

controls in each assay.

66

Assessment of Virus-inactivating Efficacy During Production of Scaffolds

The virus-inactivating efficacy of virus-spiked hFDATA’s (1.0x108 infectious

units) during scaffold production demonstrated complete inactivation of the enveloped

viruses VSV and SV5 after the first step, (Figure 5). For Adenovirus, infectious virus was

detected after the first step (~1X103 PFU), but no PFU could be detected after the second

step, (Figure 5). The limit of detection for the plaque assay is 2 PFU for Adenovirus and 5

PFU for SV5 and VSV. Data points indicate the total detectable virus at the time of initial

tissue “spike” and immediately following each step of the scaffold production process.

Tensile Testing of hFDATAs and Scaffolds

All specimens failed mid-substance and none were excluded from analysis. No

gross slippage or intermittent loss of load during elongation was observed during testing,

(Table 2). Ultimate tensile load at break (UTL) of the scaffolds was 100.30% (184.69 +

32.62 N, n=10) of that observed for the hFDATAs (184.14 + 24.36 N, n=10), p=0.99.

Stiffness was reduced in scaffolds to 72.63% (65.87 +/- 38.66 N/mm, n=10) of the value

observed for hFDATAs (90.69 +/- 11.13 N/mm, n=10), p=0.15. The ultimate tensile stress

at break (UTS) was 69.24% (20.40 +/- 3.69 MPa, n=10) of the hFDATAs (29.47 +/- 5.13

Mpa, n=10), p=0.17. The calculated elastic modulus (E) of the scaffolds was 59.30%

(166.34 + 36.09 MPa) of the hFDATAs (280.51 + 52.45 MPa), p=0.09. The percent

elongation at UTS (mm/mm*100) observed for the scaffolds was 138.47% (33.11 + 2.79%)

of that observed for the hFDATAs (23.91 + 1.57), a difference which was significant,

p=0.01.

67

Tensile Testing of Implanted hFDATAs and Scaffolds

None of the hFDATAs or scaffolds failed in vivo. Scaffold failure occurred in the

mid-substance (1/5), at the femoral (2/5), the tibial end (2/5) of the tendon. hFDATA failure

occurred at the femoral (3/5), and at the tibial end (2/5), (Table 3). Tensile testing of the

scaffolds determined the UTL was 116.16% (37.74 + 12.13 N, n=5) of that observed for

the hFDATAs (32.49 + 16.08 N, n=5), p=0.08. Scaffold stiffness was 105.24% (16.49 +

6.56 N/mm, n=5) of the value observed for hFDATAs (15.66 + 8.02 N/mm, n=5), p=0.94.

Scaffold UTS was 96.75% (9.90 + 3.12 MPa, n=5) of that observed for hFDATAs (10.23

+ 7.67 Mpa, n=5), p=0.96. Scaffold calculated elastic modulus (E) was 107.94% (26.08 +

8.56 MPa) of that observed for the hFDATAs (24.16 + 17.36 MPa), p=0.92. The percent

elongation at UTS (mm/mm*100) observed for the scaffolds was 120.03% (122.75 +

19.74%) of that observed for the hFDATAs (102.27 + 39.49%), a difference which was

not significant in contrast to the values observed in vitro, p=0.66.

Histology of Implanted of Scaffolds

No significant differences in the formation of new bone or fibrocartilage, or graft

bonding to adjacent tissue were observed between the scaffolds (n=3) and hFDATAs (n=3)

after implantation in vivo (Figure 6, Table 1). A significant increase in host cells infiltrating

the scaffold was observed (Figure 6C, 6D) when compared to hFDATAs (Figure 6A, 6B),

p=0.002. A significant increase in the host cell penetration depth within scaffolds (Figure

6C, 6D) was observed compared to hFDATAs (Figure 6A, 6B) after implantation, p=0.002.

68

DISCUSSION

The maintenance of necessary tensile properties, elimination of potential sources

of disease transmission and optimization of ultra-structural architecture to accelerate graft

remodeling are critical elements of musculoskeletal scaffold design. This study confirmed

that human allograft tissue can be processed to remove infectious viral material to produce

a decellularized, architecturally-modified, cyto-compatible scaffold that promotes cell

infiltration in vivo, yet retains tensile properties similar to hFDATA in vitro or in vivo.

Use of allograft tissue has the potential for disease transmission from contaminated tissue.34

Guelich et al reported a positive bacterial culture rate of 9.7% (24 of 247 allografts) in their

retrospective review of 35 ACL reconstruction patients who underwent ACL allograft

reconstruction.35 Similarly, Diaz-de-Rada et al. found positive bacterial cultures in

13.25% (24 of 181 allografts) of their allograft tissues.36 While the high rate of positive

bacterial cultures is concerning, it may not translate into equivalent rates of clinical

infections. Few cases of septic arthritis where cultures grew the same organism as the pre-

implantation cultures are reported.37,38 The most concerning scenario involves the

distribution of human allograft tissue deemed safe as a result of laboratory error or

incomplete testing. Such tissue potentially could transmit fatal diseases (Hepatitis B/C or

HIV) to allograft recipients with devastating consequences.39 Although the literature

confirms that this risk is low, the confirmed cases not only represent a major

medical/surgical challenge and pose serious medical-legal and ethical issues. The oxidative

elimination of infectious viral material in this study resulted in a level of viral sterility that

surpassed that of current industry standards.3 Further studies are planned to validate the

effect of scaffold production on bacterial and fungal sterility.

69

Scheffler et al. compared the remodeling process and restoration of mechanical

function of a free soft tissue autograft and an identical allograft in an in vivo sheep model.40

The authors found a significant delay in remodeling and decreased tensile properties for

the allograft group compared to the autograft group. They speculated that delayed

remodeling resulted from differences in the extracellular and collagenous matrix and

immunogenic responses to the allograft tissue. Others have noted similar findings in

allografts. 3,4,17-19 In our experiments, a significant decrease in residual cellular material was

observed in processed scaffolds, minimizing the immunogenic and inflammatory response

to scaffold implantation. The observed increase in total porosity in conjunction with the

removal of cellular material from the original allograft, resulting from the combined

oxidative-decellularization solution using PAA and Triton X-100™, was associated with

early and increased host cell infiltration into the implanted scaffold. Infiltration of cells

may promote earlier repopulation, remodeling, and scaffold integration compared to

traditional allograft tissue. At harvest, the scaffolds appeared to be in the early maturation

phase of healing with increased cell infiltration and possibly a shorter necrotic phase than

the hFDATA as would be expected after decellularization and oxidation.8 Clinically, this

finding may translate into accelerated rehabilitation and earlier return to activity or sport.

However, further studies are necessary to prove this.

Peracetic acid (PAA) has been successfully used to sterilize human patellar tendon

allografts without impacting their mechanical properties.41 Scheffler and coworkers

analyzed the remodeling of PAA-sterilized ovine ACL allografts and found that PAA

slowed the remodeling process and impacted mechanical properties at 6 and 12 weeks

compared to nonsterilized allografts and autografts.42 In contrast, we did not detect residual

70

PAA, nor did the use of PAA in our decellularization protocol significantly decrease the

scaffold’s tensile properties either in vitro or in vivo, render the scaffolds cytotoxic, or

provoke an inflammatory response in vitro. The significant increase in the percent-

elongation at UTS observed in the scaffolds in vitro is expected. The oxidative and

enzymatic disruption of the hFDATA necessary to increase porosity and remove infectious

particles and donor cellular material during scaffold production.20 It is likely a consequence

of the disruption of covalent and non-covalent interactions within the scaffold leading to

decreased resistance to deformation.

Limitations of the current study include the use of a single allograft tendon type,

hFDATA. However, other types of tissues have responded favorably to the same

decellularization protocol.43 Furthermore, freeze-dried allografts are an excellent source

material because they possess equivalent tensile properties prior to and after implantation

in vivo, they incorporate similarly to fresh-frozen allografts, are less immunogenic than

fresh or fresh-frozen grafts, and are not associated with disease transmission in recipients,

even when such grafts came from an infected donor. However, fresh-frozen and fresh grafts

from those same donors did result in disease transmissions in recipients.44 Another

limitation is the time period that implanted scaffolds were studied. Further longitudinal in

vivo studies are necessary to determine the behavior of implanted scaffolds, with an

additional comparison to autograft tendon, at time points greater than 12 weeks in order to

investigate the long-term clinical results of allograft-derived scaffolds. Similarly, tissue

processing protocols may require optimization to influence scaffold behavior for time

points over 12 weeks. Although this process may increase the number of usable donor

allografts, there is still a limited supply of donors.

71

The tensile properties determined for the hFDATAs and the scaffolds in the in vitro

and in vivo portions of the study were performed using established testing parameters for

both analyses that allow direct comparison of the scaffolds and the hFDATAs in vitro or

in vivo with equivalent error.25-32 However, overall dimensions and fixation of each

analysis, particularly the complexity of the in vivo portion, make a direct comparison of in

vitro to in vivo tensile and material properties inappropriate. As such, no comparison should

be made between the in vitro and in vivo groups with respect to these specific properties.

Similarly, the small specimens used in this study for tensile testing represent only a small

portion of a hFDATA lacking an intact paratenon. Therefore, tensile and material

properties cannot be directly compared to prior results for hFDATA and other common

grafts used for ACL reconstruction. The tensile properties of grafts from human donors

were variable and unpredictable as previously described (mean ultimate tensile strength

(UTS) of the human Achilles tendon is 1189 N (range = 360-1,965)), making statistical

analysis challenging for even large (>100) sample numbers.45 Therefore, a source of

musculoskeletal tissue with more homogenous tensile properties and greater availability,

such as xenograft tissue, may prove valuable.

In summary, this study supported the hypothesis that human allograft tissue can be

processed to remove infectious material to produce a decellularized, cyto-compatible,

architecturally-modified scaffold with tensile properties similar to human allograft tissue

in vitro and in vivo. The process also succeeded in removing inflammatory and infectious

material from hFDATA’s and resulted in an increased porosity of the hFDATA-derived

scaffolds that subsequently led to increased cell infiltration in vivo.

72

REFERENCES

1. Spindler KP, Wright, RW. Anterior cruciate ligament tear. N Engl J Med

2008;359:2135-42.

2. The American Orthopaedic Society for Sports Medicine Conference on Allografts in

Orthopaedic Sports Medicine. Keystone (CO), July 14–17, 2005.

3. Cohen, SB, Sekiya, JK. Allograft Safety in Anterior Cruciate Ligament Reconstruction.

Clin Sports Med 2007;26:597–605.

4. Jackson DW, Grood ES, Goldstein JD, et al. A comparison of patellar tendon autograft

and allograft used for anterior cruciate ligament reconstruction in the goat model. Am J

Sports Med 1993;21(2):176–85.

5. Kleiner JB, Amiel D, Roux RD, et al. Origin of replacement cells for the anterior cruciate

ligament autograft. J Orthop Res 1986;4(4):466–74.

6. Arnoczky SP. Biology of ACL reconstructions: what happens to the graft? Instr Course

Lect 1996;45:229–33.

7. Arnoczky SP, Tarvin GB, Marshall JL. Anterior cruciate ligament replacement using

patellar tendon. An evaluation of graft revascularization in the dog. J Bone Joint Surg Am

1982;64(2):217–24.

8. Gulotta, LV, Rodeo, SA. Biology of Autograft and Allograft Healing in Anterior

Cruciate Ligament Reconstruction. Clin Sports Med 2007;26:509–524.

9. Durselen L, Claes L, Ignatius A, Rubenacker, S. Comparative animal study of three

ligament prostheses for the replacement of the anterior cruciate and medial collateral

ligament. Biomaterials 1996;17:977–82.

73

10. Muren O, Dahlstedt L, Brosjo E, Dahlborn M, Dalen N. Gross osteolytic tibia tunnel

widening with the use of Gore-Tex anterior cruciate ligament prosthesis: a radiological,

arthrometric and clinical evaluation of 17 patients 13–15 years after surgery. Acta Orthop

2005;76:270–423.

11. Roolker W, Patt TW, van Dijk CN, Vegter M, Marti RK. The Gore-Tex prosthetic

ligament as a salvage procedure in deficient knees. Knee Surg Sports Traumatol Arthrosc

2000;8:20–527.

12. Petrigliano FA, McAllister DR, Wu BM. Tissue Engineering for Anterior Cruciate

Ligament Reconstruction: A Review of Current Strategies. Arthroscopy 2006;22(4):441-

451.

13. Ge Z, Yang F, Goh JCH, Ramakrishna S, Lee EH. Biomaterials and scaffolds for

ligament tissue engineering. J Biomed Mater Res 2006;77A: 639–652.

14. Derwin KA, Baker AR, Spragg RK, Leigh DR, Iannotti JP. Commercial extracellular

matrix scaffolds for rotator cuff tendon repair: Biomechanical, biochemical and cellular

properties. J. Bone Joint Surg 2006;88(12):2665-2672.

15. Tischer T, Vogt S, Aryee S, Steinhauser E, Adamczyk C, Martinek V, Milz S, Imhoff

AB. Tissue engineering of anterior cruciate ligament: a new method using acellular tendon

allografts and autogenous fibroblasts. Arch Orthop Trauma Surg 2007;127:735-741.

16. Tischer T, Aryee S, Wexel G, Steinhauser E, Adamczyk C, Eichhorn S, Milz S,

Martinek V, Gänsbacher B, Imhoff AB, Vogt S. Tissue engineering of the anterior cruciate

ligament-sodium dodecyl sulfate-acellularized and revitalized tendons are inferior to native

tendons. Tissue Eng Part A. 2010;16(3):1031-40.

74

17. Iannotti JP, Codsi MJ, Kwon YW, Derwin K, Ciccone J, Brems JJ. Porcine small

intestine submucosa augmentation of surgical repair of chronic two-tendon rotator cuff

tears. A randomized, controlled trial. J Bone Joint Surg Am. 2006;88(6):1238-44.

18. Walton JR, Bowman NK, Khatib Y, Linklater J, Murrell GA. Restore orthobiologic

implant: not recommended for augmentation of rotator cuff repairs. J Bone Joint Surg Am.

2007;89(4):786-91.

19. Snyder SJ, Arnoczky SP, Bond JL, Dopirak R. Histologic evaluation of a biopsy

specimen obtained 3 months after rotator cuff augmentation with GraftJacket Matrix.

Arthroscopy. 2009;25(3):329-33.

20. Whitlock PW, Smith TL, Poehling GG, Shilt JS, Van Dyke ME. A naturally derived,

cytocompatible, and architecturally optimized scaffold for tendon and ligament

regeneration. Biomaterials 2007;28:4321-4329.

21. Cole DW, Ginn TA, Chen GJ, Smith BP, Curl WW, Martin DF, Poehling GG. Cost

comparison of anterior cruciate ligament reconstruction: autograft versus allograft.

Arthroscopy 2005;21:786–90

22. Poehling GG, Curl WW, Lee CA, Ginn TA, Rushing JT, Naughton MJ, Holden MB,

Martin DF, Smith BP. Analysis of outcomes of anterior cruciate ligament repair with 5-

year follow-up: allograft versus autograft. Arthroscopy 2005;21:774–85.

23. Pariente JL, Kim BS, Atala A. In vitro biocompatibility assessment of naturally derived

and synthetic biomaterials using normal human urothelial cells. J Biomed Mater Res

2001;55:33–9.

24. (a) ISO10993-5: Biological evaluation of medical devices. Test for cytotoxicity: in

vitro methods. Geneva: Switzerland; (b) Annual book of American Society for Testing and

75

Materials standards: F748: Practice for selecting generic biological test methods for

materials and devices. F813; Standard practice for direct contact cell culture evaluation of

materials for medical devices. Philadelphia, PA: American Society for Testing and

Materials; 1991.

25. Mow VC, Flatow EL, Ateshain GA. Chapter 5 Biomechanics and Woo SLY, Kai-Nan

A, Frank CB, Livesay GA, Ma CB, Zeminski J, Wayne JS, Myers BS. Chapter 24

Anatomy, Biology, and Biomechanics of Tendon and Ligament. In: Orthopaedic basic

science. Biology of the musculoskeletal system. Chicago: American Academy

of Orthopaedic Surgeons; 2000. p. 133–80 and p. 581–616.

26. Woo SLY, Abramowitch SD, Kilger R, Liang R. Biomechanics of knee ligaments:

injury, healing, and repair. J Biomech 2006;39:1–20. [21] Fung YC. Biomechanics:

mechanical properties of living tissues. New York: Springer; 1993.

27. Lee TQ, Woo SLY. A new method for determining cross-sectional shape and area of

soft tissues. J Biomech Eng 1988;110(2):110–4.

28. Peterson RH, Woo SLY. A new methodology to determine the mechanical properties

of ligaments at high strain rates. J Biomech Eng 1986;108(4):365–7.

29. Peterson RH, Gomez MA, Woo SLY. The effects of strain rate on the biomechanical

properties of the medial collateral ligament: a study of immature and mature rabbits. In:

Proceedings of the Transactions of the Orthopaedic Research Society. Chicago:

Orthopaedic Research Society; 1987.

30. Woo SLY, Ohland KJ, McMahon PJ. Biology, healing and repair of ligaments. In:

Finerman GAH, Noyes FR, editors. Biology and biomechanics of the traumatized synovial

76

joint: the knee as a model. Chicago: American Academy of Orthopaedic Surgeons; 1992.

p. 241–73.

31. Grana WA, Egle DM, Mahnken R, Goodhart CW. An analysis of autograft fixation

after anterior cruciate ligament reconstruction in a rabbit model. Am J Sports Med. 1994

May-Jun;22(3):344-51.

32. Danto MI, Woo SL. The mechanical properties of skeletally mature rabbit anterior

cruciate ligament and patellar tendon over a range of strain rates. J Orthop Res. 1993

Jan;11(1):58-67.

33. Yeh WL, Lin SS, Yuan LJ, Lee KF, Lee MY, Ueng SW. Effects of hyperbaric oxygen

treatment on tendon graft and tendon-bone integration in bone tunnel: biochemical and

histological analysis in rabbits. J Orthop Res. 2007 May;25(5):636-45.

34. Vangsness CT, Wagner PP, Moore TM, Roberts MR. Overview of safety issues

concerning the preparation and processing of soft-tissue allografts. Arthroscopy. 2006

Dec:22(12):1351-1358.

35. Guelich DR, Lowe WR, Wilson B. The routine culture of allograft tissue in anterior

cruciate ligament reconstruction. Am J Sports Med. 2007;35:1495-1499.

36. Diaz-de-Rada P, Barriga A, Barroso J, et al. Positive culture in allograft ACL-

reconstruction: what to do? Knee Surg Sports Traumatol Arthrosc. 2003;11:219-222.

37. Crawford C, Kainer M, Jernigan D, et al. Investigation of postoperative allograft-

associated infections in patients who underwent musculoskeletal allograft implantation.

Clin Infect Dis. 2005;41:195-200.

38. Lee EH, Ferguson D, Jernigan D, et al. Invasive group-a streptococcal infection in an

allograft recipient. A case report. J Bone Joint Surg Am. 2007;89:2044-2047

77

39. Tugwell BD, Patel PR, Williams IT, Hedberg IT, Hedberg K, Chai F, Nainan OV,

Thomas AR, Woll JE, Bell BP, Cieslak PR. Transmission of hepatitis C virus to several

organ and tissue recipients from an antibody-negative donor. Ann Intern Med. 143(9):648-

654.

40. Scheffler SU, Schmidt T, Gangéy I, Dustmann M, Unterhauser F, Weiler A. Fresh-

frozen free-tendon allografts versus autografts in anterior cruciate ligament reconstruction:

delayed remodeling and inferior mechanical function during long-term healing in sheep.

Arthroscopy. 2008 Apr;24(4):448-58.

41. Lomas RJ, Jennings LM, Fisher J, Kearney JN. Effects of a peracetic acid disinfection

protocol on the biocompatibility and biomechanical properties of human patellar tendon

allografts. Cell Tissue Bank. 2004;5(3):149-60.

42. Scheffler SU, Gonnermann J, Kamp J, Przybilla D, Pruss A. Remodeling of ACL

allografts is inhibited by peracetic acid sterilization. Clin Orthop Relat Res. 2008

Aug;466(8):1810-8.

43. Stabile KJ, Odom D, Smith TL, Northam C, Whitlock PW, Smith BP, Van Dyke ME,

Ferguson CM. An acellular, allograft-derived meniscus scaffold in an ovine model.

Arthroscopy. 2010 Jul;26(7):936-48.

44. Mahirogullari M, Ferguson CM, Whitlock PW, Stabile KJ, Poehling GG. Freeze-dried

allografts for anterior cruciate ligament reconstruction. Clin Sports Med. 2007;26:625-637.

45. Louis-Ugbo J, Leeson B, Hutto WC. Tensile properties of fresh human calcaneal

(Achilles) tendons. Clinical Anatomy. 2004;17(1):30-35.

78

FIGURES AND TABLES

Figure 1. Experimental flowchart detailing the total number of human Freeze-Dried

Achilles Tendon Allografts (hFDATAs) used and their subsequent processing and

analysis.

79

Figure 2. Histologic Analysis of hFDATA’s and hFDATA-derived Scaffolds. A) H&E-stained longitudinal section of hFDATA; B) DAPI-stained longitudinal section of hFDATA; C) H&E-stained section of scaffold; D) DAPI-stained section of scaffold. Panels A,C at 50X magnification. Panels B,D at 200X magnification.

80

Figure 3. Scanning Electron Microscopy of hFDATA’s and hFDATA-derived Scaffolds. A) Cross-section of hFDATA; B) Cross-section of scaffold. 150X magnification.

81

Figure 4. In Vitro Cyto-compatibility of hFDATA-derived Scaffolds: Direct Contact Method. A) Metabolic activity of cells exposed to scaffold and positive control (media alone). B) Cell viability of cells exposed to scaffold and positive control (media alone). Data expressed as mean + SEM.

82

Figure 5. Assessment of Virus-inactivating Efficacy During Production of FDATA-derived Scaffolds. Data expressed as mean + SEM.

83

Figure 6. Histology of Implanted of hFDATA-derived Scaffolds at 12 Weeks. A) hFDATA 50x. B) hFDATA 200x. C) Scaffold 50x. D) Scaffold 200x. Cross-section centered on the graft tunnel with the surrounding graft-tunnel interface at the periphery of the frame, distal femur, Masson’s Trichrome stain.

84

Table 1. Scoring Results for Histological Analysis of Implanted Scaffolds and hFDATA’s after 12 weeks. Data expressed as mean, (range).

Table 1. Scoring Results for Histological Analysis of Implanted Scaffolds and hFDATA’s after 12 weeks.

Scaffold hFDATAs p-value

Fibrocartilage formation (0-3) 2.7 (2-3) 2 (1-3) 0.145 New bone formation (0-3) 2.5 (2-3) 1.7 (1-3) 0.18 Tendon graft bonding to adjacent tissue (0-3) 3 (3-3) 2.5 (2-3) 0.18 Cell penetration depth (0-3) 2.3 (1-3) 0 (0-0) 0.002 Cells within graft (0-3) 2.7 (2-3) 1 (1-1) 0.002

Maximum Score (15) 13.2 (10-15) 7.2 (5-10) 0.001

85

Table 2. Tensile Testing of hFDATA’s and hFDATA-derived Scaffolds: The ultimate tensile load at break (UTL, N), stiffness (N/mm), ultimate tensile stress at break (UTS, MPa), elastic modulus (E, MPa) % elongation at UTS (mm/mm*100) observed for the scaffolds (n=10) were 100.30% (p=0.99), 72.63% (p=0.15), 69.24% (p=0.17), 59.30% (p=0.09) and 138.47% (P=0.01) of the values observed for the hFDTAs (Mean + SEM, Student’s two-tailed t-test, equal variances).

Table 2. Tensile Properties In vitro Property hFDATA Scaffold % hFDATA p-value

UTL + SEM [N] 184.14 + 24.36 184.69 + 32.62 100.3 0.99

Stiffness + SEM [N/mm] 90.69 + 11.13 65.87 + 38.66 72.63 0.15

UTS + SEM [Mpa] 29.47 + 5.13 20.40 + 3.69 69.24 0.17

Elastic Modulus + SEM [Mpa] 280.51 + 52.45 166.34 + 36.09 59.3 0.09 % Elongation at UTS + SEM [mm/mm*100] 23.91 + 1.57 33.11 + 2.79 138.47 0.01

86

Table 3. Tensile testing of Implanted hFDATA’s and hFDATA-derived Scaffolds at 12 Weeks. The ultimate tensile load at break (UTL, N), stiffness (N/mm), ultimate tensile stress at break (UTS, MPa), elastic modulus (E, MPa) % elongation at UTS (mm/mm*100) observed for the scaffolds (n=10) were 116.16% (p=0.08), 105.24% (p=0.94), 96.75% (p=0.96), 107.94% (p=0.92) and 120.03% (P=0.66) of the values observed for the hFDTAs (Data expressed as mean + SEM, % of hFDATA. Student’s two-tailed t-test, equal variances).

Table 3. Tensile Properties In vivo Property hFDATA Scaffold % hFDATA p-value

UTL + SEM [N] 32.49 + 16.08 37.74 + 12.13 116.16 0.08 Stiffness + SEM [N/mm] 15.66 + 8.02 16.49 + 6.56 105.24 0.94

UTS + SEM [Mpa] 10.23 + 7.67 9.90 + 3.12 96.75 0.96

Elastic Modulus + SEM [Mpa] 24.16 + 17.36 26.08 + 8.56 107.94 0.92 % Elongation at UTS + SEM [mm/mm*100] 102.27 + 39.49 122.75 + 19.74 120.03 0.66

87

CHAPTER 4

THE DEVELOPMENT OF A XENOGRAFT-DERIVED SCAFFOLD FOR

TENDON AND LIGAMENT RECONSTRUCTION

Thorsten M. Seyler, M.D.1,2; Daniel N. Bracey, M.D.1,2; Johannes F. Plate, M.D.1,3; Mark

O. Lively, Ph.D.2,4; Sandeep Mannava, M.D., Ph.D.1,3; Thomas L. Smith, Ph.D.1,2,5;

Justin M. Saul, Ph.D.6; Mark E. Van Dyke, Ph.D.1,5; Patrick W. Whitlock, M.D., Ph.D.1,7

1Department of Orthopaedic Surgery, Wake Forest School of Medicine, Winston Salem,

North Carolina

2Molecular Medicine and Translational Science Graduate Program, Wake Forest

University Graduate School of Arts & Sciences, Winston Salem, North Carolina

3Neuroscience Graduate Program, Wake Forest University Graduate School of Arts &

Sciences, Winston Salem, North Carolina

4Department of Biochemistry, Wake Forest School of Medicine, Winston Salem, North

Carolina

5Virginia Tech-Wake Forest University, School of Biomedical Engineering and

Sciences, Blacksburg, Virginia

6Department of Chemical, Paper and Biomedical Engineering, Miami University,

Oxford, Ohio

7Department of Orthopaedic Surgery, Children’s Hospital Los Angeles, Los Angeles,

California

88

The following manuscript was submitted for publication to the Arthroscopy: The Journal

of Arthroscopic & Related Surgery. This chapter is currently under review. Stylistic

variations are due to the requirements of the journal. Thorsten M. Seyler made substantial

contributions to the conception of the work and all experiments. Participated in the

acquisition, analysis, or interpretation of data for all experiments. Drafting the work and

revising it critically for important intellectual content including final approval of the

version to be published. He is accountable for all aspects of the work in ensuring that

questions related to the accuracy or integrity of any part of the work are appropriately

investigated and resolved. He also served as corresponding author.

89

ABSTRACT

Purpose: Xenografts are highly attractive graft choices as they are abundantly available.

However, to make xenotransplantation a clinical reality, it is necessary to remove xenograft

antigens associated with xenograft rejection. This work reports on the development of a

decellularized xenograft-based scaffold suitable for tendon and ligament reconstruction.

Methods: Porcine patellar tendons were processed using a novel decellularization and

oxidation protocol that combines physical, chemical, and enzymatic modalities.

Results: A decrease in cellularity based on histology and a significant decrease in DNA

content were observed in the scaffolds compared with the native tendon (p<0.001).

Porosity and pore size was increased significantly (p<0.001); SEM images confirmed the

increase in scaffold pore size and porosity. In vitro cytocompatibility of scaffolds by direct

contact methods demonstrated that viability for cells exposed to the scaffolds and for the

negative controls was not statistically significant. The α-gal xenoantigen level was

significantly lower in the decellularized scaffold group compared to fresh-frozen, non-

decellularized tissue (p<0.001). The relative degree of antigen removal and immunological

response was determined through measuring the pro-inflammatory cytokines, tumor

necrosis factor (TNF)-α and Interleukin-6 (IL-6). TNF-α and IL-6 levels were significantly

(p<0.001) reduced compared with untreated controls when human acute monocytic

leukemia cells were exposed to scaffold, native tendon, and after LPS stimulation. These

results were confirmed by an attenuated response to scaffolds in vivo after implantation in

a non-human primate model.

90

Conclusions: These results provide evidence that a novel decellularization and oxidation

process can effectively remove cells, lipids, and other sources of antigenic material such

as the α-gal xenoantigen while preserving the three-dimensional structure.

Clinical relevance: The use of this novel decellularization and oxidation process favorably

modifies tendon microarchitecture and aid in the removal xenoantigens. Process can be

combined with other modalities to further facilitate xenoantigen removal.

Key words: ligament, tendon, xenograft, tissue engineering, scaffold, alpha-gal

91

INTRODUCTION

Allograft tissue is commonly used in musculoskeletal surgical procedures, and its

popularity has grown rapidly, exceeding the use of autograft tissue in many countries.

Despite improvements in safety standards for allograft tissue donation, procurement,

testing, processing, preservation, storage and distribution, the risk of disease and

malignancy transmission via transplantation of allograft tissues cannot be completely

excluded and such an event may cause a potentially life-threateningcomplication.(1-3)

Project NOTIFY was launched in 2010 by the World Health Organization (WHO) and

signifies an effort to improve recognition, reporting, tracking and investigation of adverse

outcomes associated with the use of allograft tissues.(1) The preliminary report on the work

of Project NOTIFY documented cases of transmission of human immunodeficiency virus

(HIV, n = 12), hepatitis C virus (HCV, n = 10), human T-lymphotropic virus (HTLV, n =

1), unspecified hepatitis (n = 1), tuberculosis and other bacteria (n = 49). Concomitantly,

Varettas reported the results of swab and tissue samples received for bioburden testing of

cadaveric allograft tissue samples at the time of tissue retrieval and found bacterial or

fungal growth from 29.1 % of samples.(4)

One approach to eliminate the risk of disease transmission and address allograft

shortage is xenotransplantation, the transplantationof non-human animal tissues into a

human recipient.(5, 6) Perhaps the most intriguing advantage of xenotransplantation is the

infinite amount of xenograft tissue available. This availability has been the basis for the

rapidly developing xenograft market with porcine- and bovine-derived grafts being the

most relevant xenograft tissues for orthopaedic procedures due to similarities in size and

biomechanical properties.(7) However, the immune differences in cross-species

92

transplantation can induce a rejection of animal tissues by humans. Many studies have

shown that xenogeneic tissues express superficial epitopes such as the α-gal epitope

(Galα1-3Galβ1-4GlcNAc-R), responsible for triggering this tissue rejection. The α-gal

epitope interactswith thehuman natural anti-Gal antibody and causes a complement-

mediated as well as a direct T cell-mediated immune response, making α-gal a major

obstacle to the clinical use of xenotransplanted scaffolds.(8, 9) Careful breeding of animals

free of specific pathogens may make xenotransplantation safer in some respects than

human allotransplant donors; however, the potential transmission of pathogens (viral

infection, prion-mediated infection, and bacterial infection) from the xenograft donor to

the human recipient exists.(5, 6)

Issues of acceptable and sufficient graft material are a particular challenge for

traumatic injuries to the musculoskeletal system and complex reconstructions in

musculoskeletal oncology.(10, 11) A readily available animal source for

xenotranplantation and the addition of cells to these xenograft tissues may represent a more

effective approach to the treatment these injuries than traditional methods. Thus,

approaches to produce decellularized xenografts have potential benefits both now and in

the future.

The purpose of the present study was to evaluate the biologic, immunological, and

biomechanical properties of a scaffold derived by architectural modification of fresh-frozen

porcine patella tendon using a decellularization protocol that combines physical, chemical

and enzymatic modalities. We hypothesized that porcine xenograft tendon could be

processed to produce a decellularized, cytocompatible, xenoantigen-depleted, and

93

architecturally-modified scaffold that would be suitable for tendon and ligament

reconstruction.

94

MATERIAL & METHODS

1.1. Preparation of Xenograft-derived Scaffolds

Porcine xenograft-derived scaffolds were prepared using a novel decellularization

protocol that was previously developed in our laboratory.(12, 13) All fresh-frozen native

porcine tendons were obtained through a single vendor (Animal Technologies, Tyler, TX)

Four native porcine patella tendons were placed into a clean, autoclaved, 1000 ml

PYREX® wide mouth round media storage bottle (Corning, Tewksbury, MA). One liter

of DNase-free/RNase-free, distilled water was added to each bottle. The bottle was placed

onto a rotating shaker (Barnstead MaxQ400, Dubuque, IA) at 160 r.p.m. at 37°C, for 12

hours. After 12 hours, the water was discarded, and the cycle was repeated. At the

conclusion of the second cycle, the water was discarded and 500ml of 0.05% trypsin

(Sigma Aldrich, St Louis, MO), 4.0 mM sodium bicarbonate (Sigma Aldrich, St Louis,

MO), and 0.5 mM tetrasodium EDTA (Sigma Aldrich, St Louis, MO), prepared in Hank's

balanced salt solution (Sigma Aldrich, St Louis, MO) was added. The sample was placed

onto the rotating shaker at 160 r.p.m. at 37°C for 1 hour. At the end of the cycle, the trypsin

solution was discarded and 1 liter Dulbecco's Modified Eagle Medium (Gibco® DMEM,

Life Technologies, Grand Island, NY) containing 10% fetal bovine serum (FBS) (Valley

Labs, Winchester, VA) and antibiotic and antimycotic solution containing 10,000 units/ml

penicillin G, 10 mg/ml streptomycin sulfate and 25 μg/ml amphotericin B (Sigma Aldrich,

St Louis, MO) was added in order to halt trypsin digestion of the samples. The sample was

placed back onto the rotary shaker at 160 r.p.m., 37°C, for 24 hours. After 24 hours, the

DMEM–FBS solution was discarded. One liter of the DNase-free/RNase-free distilled

water was added; the sample was placed onto the rotary shaker at 160 r.p.m. at 37°C for

12 hours. After 12 hours, the water was discarded and the cycle was repeated. At the

95

conclusion of the second cycle, the water wash was discarded and 1 liter of 1.5% peracetic

acid (PAA; oxidant; Sigma Aldrich, St Louis, MO) solution with 2.0% Triton X-100

(detergent; Sigma Aldrich, St Louis, MO) in distilled, deionized water (diH2O) was added;

the sample was placed onto the rotary shaker at 160 r.p.m. at 37°C for 2 hours. After 2

hours, the solution was discarded, and three 1 liter washes with diH2O were performed,

each for 12 hours at 37°C and 160 r.p.m. on the rotary shaker. At the end of the third wash,

the samples were removed and placed individually into clean, sterile 50 ml conical tubes

and frozen for 24 hours at −80°C. The product, referred to as scaffold, was freeze-dried

(Labconco, Freeze Dry System, Kansas City, MO) for 24 hours before being returned to

the freezer and stored at −80°C until further use.

1.2. Histological Analysis

Mid-substance portions of the fresh-frozen porcine patella tendons and scaffolds

were placed in 10% phosphate-buffered formalin at room temperature for 4 hours. The

tendons then were processed for histology, embedded in paraffin, and sectioned on a

microtome to obtain 5.0 μm thick, longitudinal sections. Sections were mounted on slides

and stained using hematoxylin and eosin (H&E; Sigma Aldrich, St Louis, MO), three color

trichrome (Masson’s trichrome stain, Sigma Aldrich, St Louis, MO), and 4,6-diamidino-2-

phenylindole (DAPI; Vector, Burlingame, CA) to identify cellular and nuclear

components, respectively. Representative light (H&E, Masson’s trichrome) and

fluorescence (DAPI) micrographs were taken (Zeiss Axioplan 2, Carl Zeiss, Jena,

Germany).

1.3. Scanning Electron Microscopy (SEM) Analysis

Scanning electron microscopy was performed on native porcine patella tendons and

decellularized/oxidized scaffolds. Specimens were removed from storage at −80°C,

96

equilibrated in DNase-free/RNase-free water at 37°C for 30 minutes, patted dry and fixed

in 2.5% SEM-grade glutaraldehyde diluted in phosphate buffered saline for 2 hours. After

fixation, specimens were rinsed in running diH2O for 30 minutes, followed by dehydration

using graded ethanol solutions (50%, 70%, 80%, 90%, 95%, and 100% for 20 minutes

each). Specimens were then transferred in 100% ethanol solution to a CO2 critical-point

dryer. After drying, specimens were mounted onto an SEM stand and gold sputter coated.

Cross-sectional electron-micrographs were obtained at 25.0 kV, 50 Pa, 50× magnification

using a Phillips 515 SEM with backscatter, cathodoluminescence, and EDAX detectors

(Phillips, Eindhoven, Netherlands).

1.4. Mercury Intrusion Porosimetry

Sections of both native porcine patella tendon (n=11) and scaffold (n=12) were

weighed and placed in a porosimeter (AutoPore IV 9500, Micromeritics, Norcross, GA),

which was evacuated and filled with mercury (evacuation pressure 50 μmHg, evacuation

time 5 mins, mercury filling pressure 0.51 psia. The porosimeter was transferred into a high

pressure chamber [33,000 psi] and the intruded volume was recorded. The volume of

intruded mercury per gram of sample was assumed to be equal to the pore volume. The

percent porosity was calculated as (1- envelope density/skeletal density) x 100.

1.5. Detection and Identification of Proteins with Mass spectrometry

The identities of proteins within the tendon scaffolds were analyzed by using mass

spectrometry (MS). Freezer-milled scaffold samples (n=3) were reduced with dithiothreitol

and alkylated with iodoacetamide before overnight digestion with trypsin at pH 8 (Trypsin

Gold, Promega, Madison, WI). Aliquots were submitted for MS (Proteomics Laboratory -

David H. Murdock Research Institute, Kannapolis, NC; www.dhmri.org). Mass

spectrometry analysis was performed by using an electrospray ionization (ESI) ion trap

97

LTQ Orbitrap XL hybrid FTMS (Fourier Transform Mass Spectrometer; Thermo Fisher

Scientific, Waltham, MA) coupled to a nanoACQUITY UltraPerformance LC (UPLC)

system (Waters, Milford, MA). The Mascot version 2.2.07 (Matrix Science, Boston, MA)

search engine was used to search the protein databases (Swiss Prot 2012_06 and National

Center for Biotechnology Information (NCBI) NCBInr 20120627 with the following

settings: MS/MS ion search, taxonomy filter = Sus scrofa (pig) all entries,

enzyme = missed cleavages = 1, fixed modifications = carbamidomethyl (C), variable

modifications = oxidation of Met and Pro (to detect hydroxyl proline in collagens), peptide

mass tolerance = 1.2 Da, fragment mass tolerance = 0.6 Da, mass values = monoisotopic,

protein mass = unrestricted, instrument type = ESI-FTICR, decoy database also

searched = 1, significance threshold = 0.05, maximum number of hits = 0, use MudPIT

protein scoring = 1, ions score cut-off = 0, include same-set proteins = 1, include sub-set

proteins = 0, include unassigned = 0, and require bold red = 1. MS hits with ≥1000 protein

scores were reported.

1.6. Tensile Testing

Fresh-frozen native porcine patellar tendons (n=18) and scaffolds (n=18) were

removed from storage at −80°C and allowed to equilibrate in sterile DPBS at 37°C for

30 min. Tendons and scaffolds then were cut into “dogbone” specimens using a custom

made punch (Style L5134, Freeman, Fremont, OH) and placed into an uniaxial load frame

(Instron 5544, Needham, MA) for tensile testing. Specimens were pre-loaded to 0.1 N, and

then pre-conditioned 10 times to 0.1 mm extension prior to being loaded to failure at a rate

of 10 mm/min (strain rate of 1.11%/s). Ultimate tensile stress (UTS, MPa) at break was

calculated as the maximal load recorded during each test. The percent strain at UTS was

calculated using the displacement data recorded by the instrument and the initial length of

98

the specimen. Elastic modulus (E, MPa) and stiffness (N/mm) were calculated from the

linear portion of stress–strain curves generated from the load and displacement data

obtained from the load frame.

1.7. Analysis of Residual Peracetic Acid

Colorimetric PAA test strips (Sensitivity/range 0-85 parts per million, LaMotte

Company, Chestertown, MD) were used for the selective determination of PAA traces in

the initial processing solution and after successive rinsing processes at 2, 12, 24, and 36

hours (n = 6). Samples were analyzed in triplicates. Residual PAA levels was expressed in

parts per million.

1.8. DNA Content

Mid-substance sections of both native porcine patella tendon (n=8) and scaffold

(n=8) were pulverized using a Freezer/Mill® (SPEX Sample Prep, Metuchen, NJ).

Twenty-five mg of tissue was placed in sterile 1.5-mL microcentrifuge tubes. Total DNA

was isolated from each sample using a DNeasy Blood & Tissue kit (Qiagen, Valencia,CA)

and analyzed in triplicates. The total DNA content was calculated from the absorption at

280 nm using a spectrophotometer (Spectronic; Thermo Scientific) and normalized to the

initial dry weight of the sample.

1.9. Measurement of α-gal Epitope Levels

To determine the immunogenic potential of the xenograft scaffolds, α-gal epitope

expression on porcine patellar tendon scaffolds and native porcine patellar tendons was

assessed by using a competition ELISA.(14, 15) This assay measures the level of α-gal

expression in tissues by using the monoclonal anti-Gal M86 antibody (Axxora, Enzo Life

Sciences Company, Farmingdale, NY) and assesses the activity of free M86 remaining in

the supernatant. Porcine scaffolds and native porcine tendons were freezer-milled and

99

homogenized in Dulbecco’s phosphate buffered saline (DPBS, Gibco® DMEM, Life

Technologies, Grand Island, NY) and brought to a concentration of 200 mg/mL. This

starting concentration was subjected to two-fold serial dilutions and an equal volume of

monoclonal anti-Gal M86 antibody diluted 1:50 was added to each tube yielding a final

dilution of 1:100 of anti-Gal M86 antibody. The tubes containing the homogenized

tendons (n=10) or scaffolds (n=10) and monoclonal antibody were incubated overnight at

4°C with constant agitation. During incubation, the M86 antibody bound to the α-gal

epitopes on the homogenized tissues and the extent of binding was proportional to the

number of epitopes present in the tissues. Subsequently, tissues and cell-bound antibody

were removed by centrifugation in a microfuge at 14,000 rpm. The activity of free

monoclonal anti-Gal M86 remaining in the supernatant was determined in ELISA wells

coated with α-gal epitopes linked to bovine serum albumin (Dextra, Redding, UK) as the

solid phase antigen using horseradish peroxidase (HRP)-conjugated goat anti-mouse IgM

antibody (Dako, Copenhagen, Denmark) as the secondary antibody. As a standard to

calculate α-Gal epitopes numbers, mouse myeloma SP2/0 cells (ATCC, Manassas, VA)

were used based on because previous studies that have shown these cells express

approximately 1 x 106 α-Gal epitopes per cell.(14)

2.10 In Vitro Cytocompatibility of Scaffolds Assessed by Direct Contact

In vitro cell viability/cytotoxicity of scaffolds was monitored using two

colorimetric methods (CellTiter 96® AQueous One Solution Cell Proliferation Assay

[MTS], Promega, Madison, WI & Neutral Red-based Toxicology Assay TOX4, Sigma

Aldrich, St Louis, MO). Prior to use, scaffolds (n=8) were removed from −80°C storage,

cut to a size of 0.2 cm x 0.2 cm, and pre-wet with 70% ethanol for 2 hours, and then washed

three times in 50 mL of Dulbecco's Modified Eagle Medium (Gibco® DMEM, Life

100

Technologies, Grand Island, NY) containing an antibiotic and antimycotic solution

(10,000 units/ml penicillin G, 10 mg/ml streptomycin sulfate and 25 μg/ml amphotericin

B; Sigma Aldrich, St Louis, MO). Latex fragments (n=10), which are considered cytotoxic,

were used as a positive control.(16) Negative controls consisting of cells in media only (n

= 10) and blank controls containing media only (n=10) were assayed in a similar fashion.

Scaffold pieces were placed in the center of sub-confluent NIH 3T3 cells (ATCC,

Manassas, VA) in 96-well plates (Nunc MaxiSorp, Thermo Fisher Scientific, Waltham,

MA according to established standards.(17) The cell-material contact was maintained for

48 hours at 37°C and 5% CO2. At the end of the incubation, the scaffold pieces were

removed and the two separate assays were performed.

For the MTS assay, 20µl of CellTiter 96® AQueous One Solution Reagent was

pipetted into each well of the 96-well plate and the plate was incubated at 37°C and 5%

CO2 for 90 minutes. The amount of soluble formazan produced by cellular reduction of

MTS was measured at 490nm on a microplate reader (SpectraMax, Molecular Devices,

Sunnyvale, CA). The absorbance obtained was directly proportional to the metabolic

activity of the cell populations and inversely proportional to the cytotoxicity of the

material. For the Neutral Red-based assay, cell culture media was removed and the cell

layers were rinsed with 200 μl of cold Dulbecco’s phosphate buffered saline (DPBS,

Gibco® DMEM, Life Technologies, Grand Island, NY). 100 μl of Neutral Red solution was

added into each well and incubated at 37°C and 5% CO2 for 2 hours. At the end of the

incubation period, the medium was carefully removed and the cells quickly rinsed with

Neutral Red assay fixative. The fixative solution was removed and the incorporated dye

was then solubilized in 100 μl of Neutral Red assay solubilization solution. The plates were

101

agitated on a small bidirectional rotator (Barnstead 4630Q, Dubuque, IA) for 10 min.

Absorbance was measured at 540 nm using a 96-well plate microplate reader. Background

absorbance was measured at 690 nm and subtracted from 540 nm measurements. Again,

the measured absorbance obtained was directly proportional to the viability of the cell

populations and inversely proportional to the cytotoxicity of the material.

2.11. Induction of Tumor necrosis factor-α (TNF- α) and Interleukin 6 (IL-6)

To further define the cytotoxic and inflammatory potential of the scaffolds, the pro-

inflammatory cytokine release of TNF-α and IL-6 was measured in vitro. Both native

porcine tendons (n=6) and porcine scaffold (n=6) were co-cultured with the human acute

monocytic leukemia cell line (THP-1; ATCC, Manassas, VA) using a previously published

protocol.(18) THP-1 cells were cultured in suspension in Roswell Park Memorial Institute

medium (RPMI 1640, Gibco® DMEM, Life Technologies, Grand Island, NY) containing

10% fetal bovine serum (Valley Labs, Winchester, VA) and 1% antibiotic and antimycotic

solution (Sigma Aldrich, St Louis, MO). A suspension of 40,000 cells containing 60 ng/ml

of phorbol 12-myristate 13-acetate (Sigma Aldrich, St Louis, MO) was added to 6-well

plates (Corning Life Sciences, Kennebunk, ME) and incubated at 37°C and 5% CO2

overnight. At the end of the incubation period, the media was carefully removed using a

pipette, the cells were rinsed in Dulbecco’s phosphate buffered saline (DPBS; Gibco®

DMEM, Life Technologies, Grand Island, NY) ,and then cultured in media containing 30

mg/ml either freezer-milled porcine tendon scaffold, freezer-milled native porcine tendon,

or lipopolysaccharides (LPS; Sigma Aldrich, St Louis, MO). Lipopolysaccharides are a

known inducer of a pro-inflammatory response and was used at 10 ng/ml as a positive

control. Cells in media only (no LPS) were used as a negative control. After overnight

incubation at 37°C and 5% CO2, the supernatant was collected using a pipette and any

102

cellular debris was removed by centrifugation at 10,000 g for 10 min. The cleared

supernatant was assayed for levels of pro-inflammatory cytokines TNF-α and IL-6 using

two commercially available ELISA kits (Quantikine human TNF-α & Quantikine human

IL-6, R&D Systems, Minneapolis, MN).

2.12. Non-human primate in vivo experiments

To assess the efficacy of the decellularization method outlined above in depleting

α-gal epitope, native porcine tendons and scaffolds were implanted in Old World primates

and their immune reaction to the scaffold was assessed. All animal procedures were

approved by the Institutional Animal Care and Use Committee and conducted according

to the National Institutes of Health (NIH) Guide for the Care and Use of Laboratory

Animals. Like humans, Old World primates lack the α-gal epitope and continuously

produce natural anti-Gal antibodies that constitute about 1% of circulating

immunoglobulins. Therefore, Old World primates are not immunotolerant toward grafts

expressing the α-gal epitope leading to an acute rejection of these grafts caused by natural

anti-Gal antibodies in a manner similar to humans. Three antigen-depleted naturally

derived porcine tendon scaffolds were produced using the method outlined above. In

addition, two native porcine patellar tendon grafts that did not undergo antigen-depletion

served as controls. The grafts were implanted in 5 male Bonnet macaques (Macaca radiata)

with a mean weight of 7.4 kg (range, 4.6 kg to 9.4 kg). Animals were sedated with 5-20

mg/kg intramuscular ketamine. The implantation site between the scapulae was shaved and

prepared in a sterile fashion using 70 % isopropyl alcohol and iodine. All procedures were

performed using standard aseptic technique including the use of sterilized surgical

instruments for each animal. An incision, approximately 3cm in length was made between

the scapulae. The native tendons or decellularized scaffolds were placed in a subcutaneous

103

pocket and secured with two 5-0 vicryl sutures. The wound was thoroughly irrigated with

sterile normal saline and closed with subdermal 2-0 vicryl sutures. The skin was closed

with a single 4-0 nylon suture. Native tendon and scaffold placement was confirmed by

ultrasound 2 weeks and 6 weeks following implantation.

Blood samples were obtained under ketamine sedation from all animals prior to the

surgical procedure, 2 days after the procedure and 1, 2, 3, 4, 8 and 13 weeks after

implantation. Whole blood samples (2ml per animal) were analyzed for white blood cell

counts (WBC). Approximately 4 ml of whole blood was collected in serum-separating

tubes that were centrifuged immediately following collection. Approximately 3 ml of

serum from each animal was obtained and transferred to sterile Eppendorf tubes and frozen

at -80°C for further processing.

Anti-α-gal activity in the serum of transplanted monkeys was assessed pre-operatively and

at weeks 2, 4, 8, and 13 post-operatively using an ELISA protocol adapted from Stone et

al. (19, 20) and Galili et al. (21). Briefly, 96-well plates (Nunc MaxiSorp, Thermo Fisher

Scientific, Waltham, MA) were coated with 50 µl of 10 µg/ml α-Gal-BSA (Dextra,

Redding, UK) for 2 hours at 37◦C and incubated overnight at 4◦C prior to being blocked

with 1% BSA. 50 µl serum aliquots diluted in 1% BSA in twofold serial dilutions from

1:50 to 1:6400 were added to wells in triplicate and incubated for 1 hour at room

temperature. Wells were aspirated and washed five times with sterile PBS. Polyclonal

rabbit anti-human antibody conjugated to horseradish peroxidase (HRP) (Dako,

Copenhagen, Denmark) diluted 1:1000 was then added to wells in 50 µl aliquots and

incubated for 1 hour at room temperature. HRP substrate (OPD, Sigma Aldrich, St Louis,

MO) was added for 30 minutes before halting color development by addition of 2N sulfuric

104

acid. Absorbance was measured at 492 nm on a microplate reader (SpectraMax, Molecular

Devices, Sunnyvale, CA). The fold change of the anti-α-Gal antibody was calculated

relative to pre-transplantation levels at each time point.

2.13. Statistical Analysis

Data from the in vitro experiments (MTS assay, neutral red assay, DNA content, α-

gal Epitope levels, TNF-α and IL-6, and residual PAA) were analyzed in triplicates. All

numerical data were averaged, the standard deviation of the mean was calculated, and a

comparison was made between appropriate groups using a two-tailed Student t-test with

equal variances for parametric data. The Mann-Whitney U test was used for analysis of

nonparametric data. For comparisons of multiple groups, one-way analysis of variance was

performed with post-hoct-testing using a Bonferroni correction. A p value of <0.05 was

considered significant.

105

RESULTS

3.1. Histological Analysis

Abundant cellular material, specifically nuclear material, was evident after H&E

(Figure 1A), Masson trichrome (Figure 1C) and DAPI staining (Figure 1E) of cross-

sectional sections of fresh-frozen native porcine patellar tendon prior to decellularization

and oxidation. Of note, there is minimal inter-fascicular and intra-fascicular space present

in the sections of the fresh-frozen native porcine patellar tendon prior to processing. After

decellularization and oxidation, no cells or condensed nuclear material were evident after

H&E staining (Figure 1B), Masson trichrome (Figure 1D), and DAPI staining (Figure 1F).

An increase in intra-fascicular and inter-fascicular space after oxidative treatment was also

observed.

3.2. Scanning Electron Microscopy (SEM) Analysis

Scanning electron microscopy demonstrated a dense micro-architecture observed

in the cross-sectional sections of fresh-frozen native porcine patellar tendon prior to

decellularization and oxidation (Figure 2A), as well as an increase in inter-fascicular and

intra-fascicular space which correlates with an increase in overall total porosity and pore

size after decellularization and oxidation (Figure 2B).

3.3. Mercury Intrusion Porosimetry

The mean total porosity measured by mercury intrusion porosimetry was 37.7% ±

4.25% for the native porcine patellar tendon and 68.43% ± 8.76% for the porcine tendon

scaffolds (Figure 3A). This difference was statistically different, p < 0.001. Similarly, the

mean pore size was 0.24 ± 0.11 μm for the native porcine patellar tendon compared with

2.32 ± 2.36 μm for the porcine tendon scaffolds (Figure 3B) (p < 0.001). These

106

measurements quantify the histological observation of increased in intra-fascicular and

inter-fascicular space after oxidative treatment.

3.4. Detection and Identification of Proteins with Mass spectrometry

Mass spectrometry analysis demonstrated the presence of collagens and

extracellular matrix proteins with high scores indicating minimal disruption to structural

characteristics that may significantly influence the mechanics and biomechanical tensile

properties of tendon and ligaments. Analysis revealed the presence of various types of

collagen. Through evolution, collagen has maintained a highly conserved amino acid

sequence which is the reason for its low antigenic potential. Other non-fibrillar components

such as proteoglycans decorin and fibromodulin and glycoproteins such as thrombospondin

and tenascin-C precursor were also detected. The mass spectrometry results from 3 distinct

tendon scaffolds are shown in Table 1. Only proteins with a score of ≥ 100 in a single

sample are shown.

3.5. Tensile Testing

No gross slippage or intermittent loss of load during elongation was observed

during testing. All specimens failed at the midsubstance and none were excluded from

analysis. The ultimate tensile load (UTL) at failure was 81.84 ± 41.60 N for the native

porcine patellar tendons compared to 95.31 ± 47.66 N for the porcine tendon scaffolds (p

= 0.372) (Table 2). The stiffness of the native tendons was 23.64 ± 13.70 N/mm compared

to 25.66 ± 13.28 N/mm for the tendon scaffolds (p = 0.657). The ultimate tensile stress at

failure was 4.60 ± 2.36 MPa for the native tendon and 5.45 ± 2.55 MPa for the tendon

scaffolds (p = 0.306). The calculated elastic modulus (E) of the native tendons was 33.65

± 18.52 MPa compared to 33.67 ± 17.13 MPa for the tendon scaffolds (p = 0.997). There

was no statistical difference between UTL, stiffness, and E between native porcine patellar

107

tendon and decellularized and oxidized scaffolds. The percent elongation at ultimate tensile

stress was 32.33% ± 5.28% for the native tendons and 53.70% ± 12.99% for the tendon

scaffolds (p < 0.001). This difference was likely due to the breaking of bonds between

proteins of the extracellular matrix during the decellularization and oxidation process.

3.6. DNA Content

The mean DNA content of the native porcine patellar tendon was 175.44 ± 57.25

ng DNA/mg tissue dry weight compared with 69.24 ± 20.43 ng DNA/mg tissue dry weight

for the decellularized and oxidized scaffold, p<0.001. The DNA content of the

decellularized and oxidized scaffolds was decreased by 61% compared to native tendon

(Figure 4).

3.7. Analysis of Residual Peracetic Acid

The PAA concentration after the 2 hours incubation period was 68.33 ± 12.91 ppm

and was reduced to 31.67 ± 7.53 ppm after the initial wash step (12 hours). None of the

scaffolds had residual PAA at the detection limit after the fourth (24 hour) and final wash

(36 hours) (Figure 5).

3.8. In Vitro Cytocompatibility of Scaffolds Assessed by Direct Contact

In vitro cytotoxicity of scaffolds was determined by the MTS assay. NIH 3T3 cells

were exposed to scaffold pieces and absorbance was measured at 490 nm providing an

estimation of mitochondrial activity of viable cell. Absorbance for cells exposed to scaffold

pieces was 0.268 ± 0.141, which was not significantly different to cells exposed to media

only 0.325 ± 0.135, p = 0.399. Similarly, theneutral reduptakeassaywasusedtoprovide

a quantitative estimation of the number of viable cells in a culture. The absorbance at 540

nm was 0.19 ± 0.07 for cells exposed to scaffold pieces compared to 0.260 ± 0.09 for NIH

3T3 cells exposed to media alone. This difference was not significant (p = 0.105). The

108

absorbance for cells exposed to the scaffolds and for cells exposed to media only (negative

controls) differed significantly from the absorbance for cells exposed to latex particles

(positive control) in both assays, p < 0.001.

3.9. Measurement of α-gal Epitope Levels

The elimination of α-Gal epitopes from native porcine patellar tendon using a

decellularization and oxidation process was assessed by binding of the monoclonal anti-

Gal M86 antibody to α-Gal epitopes of freezer-milled tendon using an established ELISA

inhibition assay. Untreated native porcine tendon displayed 50% inhibition of M86 binding

in the ELISA inhibition assay at 6.85 mg/ml tendon homogenate (Figure 6A).

Decellularization and oxidation treatment of the tendons resulted in a significantly lower

interaction of the anti-Gal M86 antibody with the scaffold homogenate with approximately

17% on inhibition at the same tissue concentration of 6.85 mg/ml (Figure 6A). This was a

reduction of approximately 66% of α-Gal epitopes expressed in the tissue. Overall, there

was a significant reduction but not complete elimination of α-Gal epitopes observed at all

tissue concentrations, p < 0.001.

Mouse myeloma SP2/0 cells express approximately 1 x 106 α-Gal epitopes per

cell.(14) A similar 50% inhibition of the M86 antibody was obtained with the mouse

myeloma cells SP2/0 at a concentration of 2.86 x 107 cells/ml (Figure 6B). Because the

mouse myeloma SP2/0 cells express approximately 1 x 106 α-Gal epitopes per cell, the

total concentration of α-Gal epitopes that induce 50% inhibition of M86 binding is 2.86 x

1013 α-Gal epitopes/ml. This suggests that 6.85 mg of native porcine patellar tendon

contains approximately 2.86 x 1013 α-Gal epitopes/ml. This measurement may not provide

109

for an accurate calculation of the number of α-Gal epitopes per cell, however, it does allow

for relative measurement of these epitopes and cross comparisons.

3.10. Induction of Tumor necrosis factor-α (TNF- α) and Interleukin 6 (IL-6)

The in vitro release of pro-inflammatory cytokines, tumor necrosis factor-α (TNF-

α) and interleukin 6 (IL-6), by THP-1 cells in response to exposure to either native porcine

patellar tendon or porcine patellar tendon scaffold was determined using ELISA and

compared to LPS stimulation. THP-1 cells exposed to native tendon released a mean of

250.69 ± 128.39 pg/ml of TNF-α compared to a mean of 82.59 ± 100.55 pg/ml in cells

exposed to tendon scaffold (Figure 7A). Stimulation of THP-1 cells with LPS 10ng/ml

yielded a mean release of 390.51 ± 48.85 pg/ml, respectively (Figure 7A). These findings

indicate that decellularization and oxidation resulted in significantly less TNF-α release, p

< 0.001. Similar results were attained when assaying for IL-6 release. THP-1 cells exposed

to native tendon released a mean of 4.15 ± 0.46 pg/ml of IL-6 compared to a mean of 3.59

± 0.39 pg/ml in cells exposed to tendon scaffold (Figure 7B). Interleukin-6 release in

response to LPS 10ng/ml stimulation was 5.58 ± 0.58 pg/ml (Figure 7B). Again, these

results demonstrate that decellularization and oxidation resulted in significantly less IL-6

release when compared to native porcine tendon and LPS stimulation, p < 0.001

2.14. Non-human primate in vivo experiments

Standard clinical hematological panels including white blood cell counts with

differentials did not yield any adverse findings in either monkeys receiving a native tendon

implant or monkeys receiving a porcine tendon scaffold. ELISA measurements of anti-Gal

response have been proven to be a sensitive measure for presence of α-gal epitopes on the

graft.(15, 20) The response is depicted as a fold-change from baseline (pre-implantation)

for anti-Gal antibody at 50% maximal binding (Figure 8). The anti-Gal titer in monkeys

110

who received native porcine patellar tendon grafts increased by an average of 47.45-fold

within 2 weeks post implantation compared to a 27.96-fold change in monkeys receiving

a tendon scaffold. A similar change was observed at 4 weeks post implantation with a

38.22-fold change for the titer in monkeys receiving native tendons and a 21.23-fold

change in titer for the monkeys receiving tendon scaffold. At the 8 and 13 week time points

post implantation, anti-Gal titers were attenuated in both monkeys receiving native porcine

tendons and decellularized and oxidized tendon scaffolds; 8.14-fold and 3.08-fold change

in titer for the monkeys who received tendon graft compared to 6.08-fold and 4.75-fold

change in titer for the monkeys who received decellularized and oxidized tendon scaffolds,

respectively. There was a greater than 40% increase in titer in the initial 2 to 4 week post

implantation period in the monkeys receiving native tendon as compared with the monkey

engrafted with tendon scaffold.

111

DISCUSSION

With increased popularity and usage of human allograft tissue in orthopaedic

surgery, the discrepancy between the demand and supply of allograft tissue for clinical use

as well as allograft safety remain major concerns.(3, 22) The development of xenograft-

based scaffolds for tendon and ligament repair may represent a potential approach to

addressing these concerns. Xenografts are highly attractive because they carry a small risk

of infectious disease transmission, are non-supply constrained, inexpensive, and are not

associated with donor site morbidity. The purpose of the present study was to develop a

decellularized xenograft-based scaffold suitable for tendon and ligament reconstruction.

The challenge was to decrease immunogenicity by removing any cellular material and

removing xenoantigens associated with graft rejection without compromising the

mechanical integrity.

In tissue engineering, numerous decellularization processes have been applied to

remove immunogenic material and produce scaffolds that preserve native anatomy and

biomechanical properties.(23, 24) The decellularization protocol used in the present study

is a combination of physical, chemical and enzymatic modalities. This protocol was

developed in our laboratories and has been used in tissue decellularization and sterilization

using various source materials including domestic chicken (Gallus gallusdomesticus) and

human allograft tendons.(12, 13) It includes sequential steps combining freeze-thaw

cycles, hypotonic solutions to lyse cells, enzymatic treatment with trypsin, extensive

washing, non-ionic detergent Triton X-100, peracetic acid, and lyophilization. Our study

results demonstrate near-complete removal of nuclear material as shown by DNA analysis

and histology. One of the biggest concerns with decellularization/oxidation of soft tissue

112

such as tendons and ligaments is damage to collagen, the major stress bearing component

of these tissues. Histological, SEM, and mass spectrometry analysis revealed preserved

microarchitecture and collagen content following decellularization. Therefore, this

increased inter-fascicular and intra-fascicular space may be beneficial and facilitate cell

infiltration and remodeling. Similar findings with regard to the microarchitecture have been

reported with other decellularization protocols.(25-27)

The biocompatibility of the scaffold was tested using in vitro direct contact

cytotoxicity assays.(17) Using a combination of Triton-X and peracetic acid in the

decellularization process, no cytotoxicity toward the scaffold or any residual material in

the scaffold matrix was found. This finding is important since other detergents such as

sodium dodecyl sulfate (SDS) have shown to negatively impact tissues.(28, 29) In a recent

study by Vavken et al, Triton-X was found to be most effective for decellularization

without detrimental effects to collagen, total protein content, and glycosaminoglycan

content.(30) Peracetic acid is a common sterilization reagent that is effective against

bacteria, viruses, and spores (31-33) and has been shown to be important in increasing

porosity and reseeding potential in tissue engineering.(12, 34) Scheffler et al. reported

deleterious effects of peracetic acid sterilization on soft tissue allografts with delayed

remodeling at 8 and 12 weeks.(35) This finding stands in contrast to our previous in vitro

and in vivo findings in which we were unable to detect any residual peracetic acid after

oxidation and did not see any effects on the remodeling potential in an animal model of

xenogenic ACL reconstruction.(12, 13, 36) Furthermore, peracetic acid is known to break

down into oxygen, water, and carbon dioxide leaving no harmful residues.(37, 38)

113

Hyperacute rejection of tissues transplanted from animals to humans is primarily

mediated by the recognition of the naturally occurring anti-Gal antibody in humans to the

xenoantigen α-gal on transplanted animal tissue.(8, 39) Thus, it is critically important to

ensure that during the production of xenogenic scaffolds that α-gal epitopes are removed.

Several decellularization processes using either a detergent such as SDS or an enzymatic

treatment with α-galactosidase have shown promising results for eliminating the α-gal

epitope.(20, 40) Despite the fact that we achieved a significant reduction in epitope

expression, our process does not completely eliminate the expression of α–Gal epitopes. In

addition, there was an attenuated response and a decrease in the release of pro-

inflammatory markers to decellularized and oxidized scaffolds in vitro when compared to

untreated native tendons. These findings were consistent with the non-human primate

experiments in which we saw an approximately 25-fold decrease in response in antibody

production when compared to native untreated tendons. However, without an established

rejection threshold of α-gal epitopes, it is unclear if a low-level of remaining α-gal epitopes

may result in a prolonged low-level anti-Gal antibody production and chronic

inflammatory response comparable to a long-term tissue rejection limiting the functional

integrity of xenograft scaffolds.(41, 42) Raeder at el studied the ability of naturally

occurring anti-Gal antibodies to influence the success of implantation and remodeling of

untreated porcine small intestine submucosa (SIS).(43) The authors implanted SIS in α-

Gal knockout mice and found that there was slight delay in SIS remodeling and prolonged

inflammation at the implant site and that these differences disappeared by day 35 post-

implantation when compared to controls. Based on these findings, the authors concluded

conclude that naturally occurring anti-Gal antibodies in humans have little influence on the

114

acceptance of xenogenic biomaterials.(43) Konakci et al investigated the sera of patients

that received various porcine valve prostheses and found that contained cells that expressed

the α–Gal epitope and elicit a specific cytotoxic immune response mediated by IgM.(44)

The authors suggested that any procedure diminishing the presence of the α–Gal epitope

might decrease the immune response against the xenograft. In our study, we did observe a

lessened immune response to the decellularized and oxidized tendons in vivo. However,

further studies are needed to determine whether scaffolds developed using this process can

circumvent rejection under a sustained low-level production of these antibodies.

One of the limitations of this study is that scaffolds were implanted subcutaneously

in Old World monkeys. Thus, based strictly on these in vivo studies, we are unable to draw

any conclusions regarding the use of this porcine-derived scaffold as a graft choice with

suitable tensile properties to replace an injured ligament such as the anterior cruciate

ligament in humans. Given the significant costs associated with non-human primate

studies, we do not have any long-term data analyzing if this scaffold is repopulated by

recipient’s cells and undergoes the process of ligamentization. While we did observe

repopulation and ligamentization in vivo using a different source material in an earlier

publication(12), we do not have sufficient data with the porcine-derived scaffold to draw

similar conclusions and additional long-term in vivo experiments are required to determine

the effects of the in vivo environment on scaffold remodeling and biomechanical properties.

115

CONCLUSIONS

The results presented here demonstrate that a novel process for decellularization

and oxidation of porcine patellar tendon is effective in reducing cellular material and

decreasing the expression of the α-Gal epitope. Both the removal of cellular material as

well as the reduced expression of the α-Gal epitope may reduce the immunogenicity of the

scaffolds. The chemical oxidation portion of our process resulted in increased porosity and

individual pore size without compromising the extracellular matrix so that the prepared

scaffold has the potential to serve as a template for cell infiltration and repopulation.

Despite these promising preliminary results, additional long-term studies are needed to

further delineate the host response before assuming that this porcine xenograft-derived

scaffold is a viable option that performs in an analogous manner when compared to

autografts and allografts.

116

REFERENCES

1. Hinsenkamp M, Muylle L, Eastlund T, Fehily D, Noel L, Strong DM. Adverse

reactions and events related to musculoskeletal allografts: reviewed by the World Health

Organisation Project NOTIFY. International orthopaedics. 2012 Mar;36(3):633-41.

PubMed PMID: 22048753. Pubmed Central PMCID: 3291755. Epub 2011/11/04. eng.

2. Mallick TK, Mosquera A, Zinderman CE, St Martin L, Wise RP. Reported

infections after human tissue transplantation before and after new Food and Drug

Administration (FDA) regulations, United States, 2001 through June, 2010. Cell and

tissue banking. 2012 Jun;13(2):259-67. PubMed PMID: 21479712. Epub 2011/04/12.

eng.

3. Mroz TE, Joyce MJ, Steinmetz MP, Lieberman IH, Wang JC. Musculoskeletal

allograft risks and recalls in the United States. The Journal of the American Academy of

Orthopaedic Surgeons. 2008 Oct;16(10):559-65. PubMed PMID: 18832599. Epub

2008/10/04. eng.

4. Varettas K. Micro-organisms isolated from cadaveric samples of allograft

musculoskeletal tissue. Cell and tissue banking. 2013 Jan 23. PubMed PMID: 23340929.

Epub 2013/01/24. Eng.

5. Pierson RN, 3rd, Dorling A, Ayares D, Rees MA, Seebach JD, Fishman JA, et al.

Current status of xenotransplantation and prospects for clinical application.

Xenotransplantation. 2009 Sep-Oct;16(5):263-80. PubMed PMID: 19796067. Pubmed

Central PMCID: 2866107. Epub 2009/10/03. eng.

6. Tisato V, Cozzi E. Xenotransplantation: an overview of the field. Methods Mol

Biol. 2012;885:1-16. PubMed PMID: 22565986. Epub 2012/05/09. eng.

117

7. Laurencin CT, El-Amin SF. Xenotransplantation in orthopaedic surgery. The

Journal of the American Academy of Orthopaedic Surgeons. 2008 Jan;16(1):4-8.

PubMed PMID: 18180387. Epub 2008/01/09. eng.

8. Galili U. The alpha-Gal epitope (Galalpha1-3Galbeta1-4GlcNAc-R) in

xenotransplantation. Biochimie. 2001 Jul;83(7):557-63. PubMed PMID: 11522383. Epub

2001/08/28. eng.

9. Scalea J, Hanecamp I, Robson SC, Yamada K. T-cell-mediated immunological

barriers to xenotransplantation. Xenotransplantation. 2012 Jan-Feb;19(1):23-30. PubMed

PMID: 22360750. Epub 2012/03/01. eng.

10. Rihn JA, Harner CD. The use of musculoskeletal allograft tissue in knee surgery.

Arthroscopy : the journal of arthroscopic & related surgery : official publication of the

Arthroscopy Association of North America and the International Arthroscopy

Association. 2003 Dec;19 Suppl 1:51-66. PubMed PMID: 14673420. Epub 2003/12/16.

eng.

11. Dion N, Sim FH. The use of allografts in musculoskeletal oncology. Instructional

course lectures. 2002;51:499-506. PubMed PMID: 12064140. Epub 2002/06/18. eng.

12. Whitlock PW, Seyler TM, Parks GD, Ornelles DA, Smith TL, Van Dyke ME, et

al. A novel process for optimizing musculoskeletal allograft tissue to improve safety,

ultrastructural properties, and cell infiltration. The Journal of bone and joint surgery

American volume. 2012 Aug 15;94(16):1458-67. PubMed PMID: 22786867. Epub

2012/07/13. eng.

13. Whitlock PW, Smith TL, Poehling GG, Shilt JS, Van Dyke M. A naturally

derived, cytocompatible, and architecturally optimized scaffold for tendon and ligament

118

regeneration. Biomaterials. 2007 Oct;28(29):4321-9. PubMed PMID: 17610948. Epub

2007/07/06. eng.

14. Galili U, LaTemple DC, Radic MZ. A sensitive assay for measuring alpha-Gal

epitope expression on cells by a monoclonal anti-Gal antibody. Transplantation. 1998

Apr 27;65(8):1129-32. PubMed PMID: 9583877. Epub 1998/05/16. eng.

15. Stone KR, Abdel-Motal UM, Walgenbach AW, Turek TJ, Galili U. Replacement

of human anterior cruciate ligaments with pig ligaments: a model for anti-non-gal

antibody response in long-term xenotransplantation. Transplantation. 2007 Jan

27;83(2):211-9. PubMed PMID: 17264818. Epub 2007/02/01. eng.

16. Uphill PF, Christopher DH. Developing a Positive Control for Cytotoxicity

Testing of Medical Device Materials. Medical Device Technology. 1990;Nov/Dec:24-7.

17. International Organization for Standardization I. ISO 10993-5:2009 Biological

evaluation of medical devices - Part 5: Tests for in vitro cytotoxicity. 2009:1-34.

18. Supronowicz P, Zhukauskas R, York-Ely A, Wicomb W, Thula T, Fleming L, et

al. Immunologic analyses of bovine bone treated with a novel tissue sterilization process.

Xenotransplantation. 2008 Nov-Dec;15(6):398-406. PubMed PMID: 19152668. Epub

2009/01/21. eng.

19. Stone KR, Ayala G, Goldstein J, Hurst R, Walgenbach A, Galili U. Porcine

cartilage transplants in the cynomolgus monkey. III. Transplantation of alpha-

galactosidase-treated porcine cartilage. Transplantation. 1998 Jun 27;65(12):1577-83.

PubMed PMID: 9665073. Epub 1998/07/17. eng.

20. Stone KR, Walgenbach AW, Turek TJ, Somers DL, Wicomb W, Galili U.

Anterior cruciate ligament reconstruction with a porcine xenograft: a serologic,

119

histologic, and biomechanical study in primates. Arthroscopy : the journal of arthroscopic

& related surgery : official publication of the Arthroscopy Association of North America

and the International Arthroscopy Association. 2007 Apr;23(4):411-9. PubMed PMID:

17418335. Epub 2007/04/10. eng.

21. Galili U, LaTemple DC, Walgenbach AW, Stone KR. Porcine and bovine

cartilage transplants in cynomolgus monkey: II. Changes in anti-Gal response during

chronic rejection. Transplantation. 1997 Mar 15;63(5):646-51. PubMed PMID: 9075832.

Epub 1997/03/15. eng.

22. Vangsness CT, Jr., Dellamaggiora RD. Current safety sterilization and tissue

banking issues for soft tissue allografts. Clinics in sports medicine. 2009 Apr;28(2):183-

9, vii. PubMed PMID: 19306728. Epub 2009/03/25. eng.

23. Gilbert TW. Strategies for tissue and organ decellularization. Journal of cellular

biochemistry. 2012 Jul;113(7):2217-22. PubMed PMID: 22415903. Epub 2012/03/15.

eng.

24. Gilbert TW, Sellaro TL, Badylak SF. Decellularization of tissues and organs.

Biomaterials. 2006 Jul;27(19):3675-83. PubMed PMID: 16519932. Epub 2006/03/08.

eng.

25. Ingram JH, Korossis S, Howling G, Fisher J, Ingham E. The use of ultrasonication

to aid recellularization of acellular natural tissue scaffolds for use in anterior cruciate

ligament reconstruction. Tissue engineering. 2007 Jul;13(7):1561-72. PubMed PMID:

17518726. Epub 2007/05/24. eng.

26. Scheffler SU, Scherler J, Pruss A, von Versen R, Weiler A. Biomechanical

comparison of human bone-patellar tendon-bone grafts after sterilization with peracetic

120

acid ethanol. Cell and tissue banking. 2005;6(2):109-15. PubMed PMID: 15909098.

Epub 2005/05/24. eng.

27. Huang Q, Dawson RA, Pegg DE, Kearney JN, Macneil S. Use of peracetic acid to

sterilize human donor skin for production of acellular dermal matrices for clinical use.

Wound repair and regeneration : official publication of the Wound Healing Society [and]

the European Tissue Repair Society. 2004 May-Jun;12(3):276-87. PubMed PMID:

15225206. Epub 2004/07/01. eng.

28. Bodnar E, Olsen EG, Florio R, Dobrin J. Damage of porcine aortic valve tissue

caused by the surfactant sodiumdodecylsulphate. The Thoracic and cardiovascular

surgeon. 1986 Apr;34(2):82-5. PubMed PMID: 2424134. Epub 1986/04/01. eng.

29. Cebotari S, Tudorache I, Jaekel T, Hilfiker A, Dorfman S, Ternes W, et al.

Detergent decellularization of heart valves for tissue engineering: toxicological effects of

residual detergents on human endothelial cells. Artificial organs. 2010 Mar;34(3):206-10.

PubMed PMID: 20447045. Epub 2010/05/08. eng.

30. Vavken P, Joshi S, Murray MM. TRITON-X is most effective among three

decellularization agents for ACL tissue engineering. Journal of orthopaedic research :

official publication of the Orthopaedic Research Society. 2009 Dec;27(12):1612-8.

PubMed PMID: 19504590. Pubmed Central PMCID: 2824567. Epub 2009/06/09. eng.

31. Pruss A, Baumann B, Seibold M, Kao M, Tintelnot K, von Versen R, et al.

Validation of the sterilization procedure of allogeneic avital bone transplants using

peracetic acid-ethanol. Biologicals : journal of the International Association of Biological

Standardization. 2001 Jun;29(2):59-66. PubMed PMID: 11580210. Epub 2001/10/03.

eng.

121

32. Pruss A, Hansen A, Kao M, Gurtler L, Pauli G, Benedix F, et al. Comparison of

the efficacy of virus inactivation methods in allogeneic avital bone tissue transplants. Cell

and tissue banking. 2001;2(4):201-15. PubMed PMID: 15256903. Epub 2004/07/17. eng.

33. Pruss A, Kao M, Kiesewetter H, von Versen R, Pauli G. Virus safety of avital

bone tissue transplants: evaluation of sterilization steps of spongiosa cuboids using a

peracetic acid-methanol mixture. Biologicals : journal of the International Association of

Biological Standardization. 1999 Sep;27(3):195-201. PubMed PMID: 10652175. Epub

2000/02/01. eng.

34. Woon CY, Pridgen BC, Kraus A, Bari S, Pham H, Chang J. Optimization of

human tendon tissue engineering: peracetic acid oxidation for enhanced reseeding of

acellularized intrasynovial tendon. Plastic and reconstructive surgery. 2011

Mar;127(3):1107-17. PubMed PMID: 21364414. Epub 2011/03/03. eng.

35. Scheffler SU, Gonnermann J, Kamp J, Przybilla D, Pruss A. Remodeling of ACL

allografts is inhibited by peracetic acid sterilization. Clinical orthopaedics and related

research. 2008 Aug;466(8):1810-8. PubMed PMID: 18491201. Pubmed Central PMCID:

2584264. Epub 2008/05/21. eng.

36. Whitlock PW, Seyler TM, Northam CN, Smith TL, Poehling GG, Koman LA, et

al. Effect of cyclic strain on tensile properties of a naturally derived, decellularized

tendon scaffold seeded with allogeneic tenocytes and associated messenger RNA

expression. Journal of surgical orthopaedic advances. 2013 Fall;22(3):224-32. PubMed

PMID: 24063799. Epub 2013/09/26. eng.

37. Centre of Disease Control. Guidelines for Disinfection and Sterilization in

Healthcare Facilities. 2008.

122

38. Malchesky P. Disinfection, Sterilization, and Preservation. 5th ed. Philadelphia,

PA: Lippincott Williams & Wilkins; 2000 December 15, 2000. 1504 pages p.

39. Sandrin MS, McKenzie IF. Gal alpha (1,3)Gal, the major xenoantigen(s)

recognised in pigs by human natural antibodies. Immunological reviews. 1994

Oct;141:169-90. PubMed PMID: 7532618. Epub 1994/10/01. eng.

40. Stapleton TW, Ingram J, Katta J, Knight R, Korossis S, Fisher J, et al.

Development and characterization of an acellular porcine medial meniscus for use in

tissue engineering. Tissue engineering Part A. 2008 Apr;14(4):505-18. PubMed PMID:

18370607. Epub 2008/03/29. eng.

41. Wang H, Yang YG. Innate cellular immunity and xenotransplantation. Current

opinion in organ transplantation. 2012 Apr;17(2):162-7. PubMed PMID: 22262106.

Pubmed Central PMCID: 3324331. Epub 2012/01/21. eng.

42. Galili U, Tibell A, Samuelsson B, Rydberg L, Groth CG. Increased anti-Gal

activity in diabetic patients transplanted with fetal porcine islet cell clusters.

Transplantation. 1995 Jun 15;59(11):1549-56. PubMed PMID: 7539957. Epub

1995/06/15. eng.

43. Raeder RH, Badylak SF, Sheehan C, Kallakury B, Metzger DW. Natural anti-

galactose alpha1,3 galactose antibodies delay, but do not prevent the acceptance of

extracellular matrix xenografts. Transplant immunology. 2002 Jun;10(1):15-24. PubMed

PMID: 12182460. Epub 2002/08/17. eng.

44. Konakci KZ, Bohle B, Blumer R, Hoetzenecker W, Roth G, Moser B, et al.

Alpha-Gal on bioprostheses: xenograft immune response in cardiac surgery. European

123

journal of clinical investigation. 2005 Jan;35(1):17-23. PubMed PMID: 15638815. Epub

2005/01/11. eng.

124

FIGURES AND TABLES

Figure 1. Histologic Analysis of native porcine patellar tendon and porcine patellar tendon-derived scaffolds. (A) H&E-stained cross section of native patellar tendon; (B) H&E-stained cross section of scaffold; (C) Masson trichrome-stained cross section of native patellar tendon; (D) Masson trichrome-stained cross section of scaffold; (E) DAPI-stained cross section of native patellar tendon; (F) DAPI-stained cross section of scaffold. Removal of nuclear material and an increase in intra-fascicular and inter-fascicular space after oxidative treatment was observed. Panels a through F at 200X magnification.

125

Figure 2. Scanning Electron Microscopy of native porcine patellar tendon and porcine patellar tendon-derived scaffolds. (A) Cross-section of native porcine patellar tendon; (B) Cross-section of porcine patellar tendon-derived scaffold. There was an increase in increase in inter-fascicular and intra-fascicular space. 50X magnification.

126

Figure 3. Mercury intrusion porosimetry demonstrated a statistically significant increase in total porosimetry (A) (p<0.001) and mean pore size (B) (p<0.001) in decellularized and oxidized scaffolds compared to native tendons.

127

Figure 4. DNA detection assay revealed a significant decreased of mean DNA content in the porcine tendon-derived scaffold by 61% when compared to native tendon, p<0.001. Data expressed as the mean and standard deviation.

128

Figure 5. Within 24 hours of the oxidation step of the decellularization protocol, the peracetic acid was removed to a level below detection limits.

129

Figure 6. ELISA inhibition assay performed with the monoclonal anti-Gal antibody M86. Data presented as % inhibition of M86 binding to α-gal-BSA in ELISA, after overnight incubation of M86 diluted 1/100. (A) Measuring α- gal epitope expression in native porcine patellar tendon [white circle] and porcine tendon-derived scaffold after decellularization and oxidation [black circle]. This was a reduction of approximately 66% of α-Gal epitopes expressed in the tissue at 50% inhibition of M86 binding. (B) As a standard to calculate epitope number, mouse myeloma SP2/0 cells at various concentrations were used because these cells to express approximately 106 α-gal epitopes per cell. Error bars represent standard deviation.

130

Figure 7. Tumor necrosis factor (TNF-a) (A) and IL-6 (B) release by human acute monocytic leukemia (THP-1) cells exposed to porcine tendon-derived scaffold and native tendon. Treatment of native tendon with a novel decellularization and oxidation process resulted in a significantly (p<0.001) lower production of TNF-a release compared with native tendon and LPS stimulation. Similar results were observed in IL-6 release. LPS was used as a reference in both experiments with at two concentrations (10ng/ml). Results are displayed as the mean and standard deviation for each group.

131

Figure 8. ELISA measurements of anti-Gal response in monkeys who received native porcine patellar tendon and monkeys who received porcine tendon-derived scaffolds. Response is depicted as fold-change from baseline (pre-implantation) for anti-Gal antibody at 50% maximal binding There was a greater than 40% increase in titer in the initial 2 to 4 week post implantation period in the monkeys receiving native tendon as compared with the monkey engrafted with tendon scaffold. No difference was noted at the 8 and 13 week time points. Error bars represent standard deviation.

132

Table 1. Mass spectrometry results listing specific mass, accession number, protein description and score per sample.

Table 1. Mass spectrometry

Accession # Score Scaffold #1 Score Scaffold #2 Score Scaffold #3 Mass Description

335303568 3597 3784 3060 322197 collagen alpha‐3(VI) chain isoform 2 [Sus scrofa]

343887367 3577 3394 3070 129652 collagen, type I, alpha 2 precursor [Sus scrofa]

350578415 1619 1331 1156 241355 collagen alpha‐1(XII) chain [Sus scrofa]

335300861 745 926 764 106542 collagen alpha‐2(VI) chain [Sus scrofa]

343168808 705 614 539 139788 collagen alpha‐1(III) chain precursor [Sus scrofa]

833798 665 ND ND 71362 albumin [Sus scrofa]

124257959 746 820 746 71550 albumin, partial [Sus scrofa]

350590450 548 562 1105 43557 collagen alpha‐1(I) chain [Sus scrofa]

350596326 114 108 118 144181 collagen alpha‐1(II) chain‐like, partial [Sus scrofa]

311264394 466 552 387 43570 fibromodulin‐like isoform 1 [Sus scrofa]

350581945 461 447 455 27343 mimecan‐like [Sus scrofa]

311253435 346 433 287 194518 collagen alpha‐1(XIV) chain [Sus scrofa]

311277155 291 323 292 41849 biglycan [Sus scrofa]

311249371 259 459 352 128704 cartilage intermediate layer protein 2 [Sus scrofa]

335293906 217 185 131 36169 annexin A5‐like [Sus scrofa]

809283 207 ND ND 16082 chain B, porcine hemoglobin [Sus scrofa]

335310813 181 155 132 42655 collagen alpha‐1(VI) chain‐like, partial [Sus scrofa]

4501885 178 215 121 42052 actin, cytoplasmic 1 [Homo sapiens]

335296459 153 165 77 53692 vimentin‐like [Sus scrofa]

92020086 147 105 112 10639 type VI collagen alpha‐1 chain [Sus scrofa]

47523544 146 281 111 196922 tenascin precursor [Sus scrofa]

54020966 146 134 ND 38795 annexin A2 [Sus scrofa]

350580885 134 133 ND 99696 thrombospondin‐4‐like [Sus scrofa]

343183420 127 165 148 39097 lumican precursor [Sus scrofa]

335293644 119 ND 77 54724 vitamin D‐binding protein [Sus scrofa]

122465 110 62 60 15087 hemoglobin subunit alpha [Sus scrofa]

311264396 108 154 121 44084 prolargin‐like [Sus scrofa]

6919844 ND 328 210 75042 transforming growth factor‐beta‐induced protein ig‐

h3 [Sus scrofa]

4929107 ND 122 109 36022 decorin [Sus scrofa]

89574051 ND 111 ND 47060 mitochondrial ATP synthase, H+ transporting F1 

complex beta subunit [Sus scrofa]

1165145 ND 101 47 38501 annexin I [Sus scrofa]

164507 117 ND 117 51886 immunoglobulin gamma‐chain [Sus scrofa]

ND = not detected

133

Table 2. Mass spectrometry results listing specific mass, accession number, protein description and score per sample.

Table 2. In vitro Tensile Testing

Biomechanical Property Native Tendon Tendon Scaffold P Value

UTL [N] 81.84 ± 41.60 95.31 ± 47.66 0.372Stiffness [N/mm] 23.64 ± 13.70 25.66 ± 13.28 0.657UTS [MPa] 4.60 ± 2.36 5.45 ± 2.55 0.306Elastic modulus [Mpa] 33.65 ± 18.52 33.67 ± 17.13 0.977Elongation at UTS (%) 32.33 ± 5.28 53.70 ± 12.99 < 0.001

UTL = ultimate tensile load. UTS = ultimate tensile stress. Values are given as the mean and the standard deviation of the mean. P values were calculated using Student t test (tw-tailed, equal variances) or Mann-Whitney Rank Sum test if normaility failed.

134

CHAPTER 5

THE EFFECT OF CYCLIC STRAIN ON THE TENSILE PROPERTIES OF A

NATURALLY-DERIVED, DECELLULARIZED TENDON SCAFFOLD SEEDED

WITH ALLOGENEIC TENOCYTES AND ASSOCIATED MRNA EXPRESSION

Patrick W. Whitlock 1,2, Thorsten M Seyler 1,2, Casey Northam1, Thomas L. Smith1,2,

Gary G. Poehling1,2, L. Andrew Koman1,2, Mark E. Van Dyke2

1Department of Orthopaedic Surgery, Wake Forest University Health Sciences, Medical

Center Boulevard, Winston-Salem, North Carolina, 27157

2Institute for Regenerative Medicine, Wake Forest University Health Sciences, Medical

Center Boulevard, Winston-Salem, North Carolina, 27157

135

The following manuscript was published in the Journal of Surgical Orthopaedic Advances

September 1st, 2013 (Vol. 22 #3). This chapter is reprinted with permission. Stylistic

variations are due to the requirements of the journal. Thorsten M. Seyler made substantial

contributions to the conception of the work and all experiments. Participated in the

acquisition, analysis, or interpretation of data for all experiments. Drafting the work and

revising it critically for important intellectual content including final approval of the

version to be published. He is accountable for all aspects of the work in ensuring that

questions related to the accuracy or integrity of any part of the work are appropriately

investigated and resolved. He shared first authorship with Dr. Whitlock who served as

corresponding author.

136

ABSTRACT

Background

Naturally-derived tendon scaffolds have the potential to improve the treatment of flexor

tendon injuries.

Methods

Seeded and unseeded tendon scaffolds were maintained in the presence or absence of

physiologic strain for seven days. After seven days, the tensile properties and associated

mRNA expression were compared.

Results

Seeded scaffolds maintained in the absence of strain had significantly lower tensile

properties than unseeded tendons and fresh-frozen tendons. The loss of tensile properties

was associated with elevated MMP-2 and Collagen III expression. Tensile properties of

seeded scaffolds maintained in the presence of strain for 7 days after seeding did not

differ from those of fresh-frozen tendons.

Conclusions

This study demonstrates that the tensile properties of seeded, naturally-derived tendon

scaffolds will degrade rapidly in the absence of cyclic strain. Seeded scaffolds used for

tendon reconstruction should be maintained under cyclic strain to maintain essential

tensile properties.

137

INTRODUCTION

Tendons are dense connective tissues connecting muscle to bone, which transmit

tensile loads generated by muscles to produce joint movement [1-4]. Acute tendon injuries

typically occur after trauma, whereas chronic injuries often arise from repetitive

mechanical stress and/or degeneration. With regard to flexor tendon injuries of the hand,

clinical outcomes following surgery remain suboptimal [5-8]. Severe flexor tendon injuries

are difficult to manage surgically and may require graft material to bridge large defects [9-

11]. A major problem in flexor tendon repair is the lack of suitable graft material for

surgical reconstruction [12]. Graft materials that have been used include autografts,

allografts, and prosthetic substitutes [13]. Whereas immune rejection remains a cause of

concern when using tendon allografts, most prosthetic substitutes also fail to achieve

satisfactory long-term tendon function and, therefore, autologous grafts continue to be the

“gold standard” [13]. However, autologous grafts are traditionally limited to the palmaris

longus, plantaris and foot extensor tendons. Tissue engineering seeks to address the

limitations of each of these graft materials by implanting biologically compatible substrates

with or without cells in place of injured tendons to eliminate the concerns associated with

the use of autografts (limited availability, donor site morbidity, increased surgical time),

allografts (immunogenicity, disease transmission, excess inflammation) and prosthetic

substitutes (functionality, cytotoxic breakdown products, limited longevity) [13-15].

Current tissue-engineering strategies for flexor digitorum profundus reconstruction have

relied predominantly upon scaffolds made from synthetic (polyglycolic acid) and natural

(collagen) materials to form the cell-scaffold construct. However, synthetic scaffolds

produce breakdown products that are potentially antimitotic and cytotoxic in vivo [16].

138

Furthermore, early range of motion is considered essential to the rehabilitation and success

of flexor tendon repairs. A delay in early range of motion is associated with subsequent

adhesion formation, decreased range of motion, and poor functional outcomes

[13,14,17,18]. Tissue-engineered tendons produced in vitro or via self-assembly are

significantly weaker than native adult tendons and would not be expected to withstand

current rehabilitation protocols after implantation [16,19,20].

Several genes have been implicated in the remodeling and maturation of tendons in

vivo and in vitro. Collagen I and III have been shown to be upregulated in cells seeded on

scaffolds, but this increase in expression was not observed until Day 14 [21]. MMP-2

(matrix metalloproteinase-2) has been implicated in the initiation and progression of

collagen fibril growth, matrix assembly, and tendon development [22]. The overexpression

of MMP-2 has been associated with observed altered collagen fibril structure and tendon

organization during development [23,24]. Tenomodulin, a type II trans-membrane protein,

has been identified as a late marker for tendon formation and tenocyte differentiation [25].

Therefore, the effect of cyclic strain upon the expression of these genes by cells seeded in

naturally-derived tendon scaffolds is of significant interest and may aid not only in the

optimization of structural and material properties of cell-seeded scaffolds, but also the

mechanisms involved in tendon injury, repair and healing.

Little is known about the effect of mechanical strain on the tensile properties and mRNA

expression of seeded, decellularized tendon scaffolds. We previously described the

development and characterization of a novel, architecturally-optimized, biocompatible,

decellularized and oxidized scaffold derived from avian flexor digitorum profundus tendon

[28]. The goal of this study was to determine the effect of the absence or presence of cyclic

139

strain upon the tensile properties of flexor digitorum profundus (FDP)-derived tendon

scaffolds seeded with allogeneic tenocytes, and the mRNA expression of genes of interest

by the seeded cells. We hypothesized that the tensile properties (ultimate tensile stress,

modulus, stiffness) of seeded scaffolds maintained in the presence of strain would be

greater than those of seeded scaffolds maintained in the absence of strain and that the

difference would be associated with a difference in the mRNA expression by seeded cells.

140

MATERIAL & METHODS

The feet of 56-day-old Leghorn chickens were provided at no cost by Wayne Farms

(Dobson, NC), placed on ice immediately after receipt, and stored at 4°C until dissection.

The feet were cleaned and disinfected using aseptic techniques. The intrasynovial portion

of the FDP tendon (ca. 60 mm x 3 mm x1 mm) was harvested from the proximal portion

of the proximal phalanx to its distal insertion on the distal phalanx of the long digit and

prepared according to the previously published protocol [26]. Briefly, immediately after

harvest, FDP tendons were transferred under aseptic conditions to individual clean,

autoclaved, 100 ml glass, screw-top bottles (Gibco, Grand Island, NY). One hundred

milliliters of DNase-free/RNase-free, distilled water was added to each sample. The bottle

was placed onto a rotating shaker (Barnstead MaxQ400, Dubuque, IA) at 200 r.p.m., 37°C,

for 24 h. After 24 h, the water was discarded and the cycle was repeated. At the conclusion

of the second cycle, the water was discarded and 100 ml of 0.05% trypsin-EDTA (Gibco,

Grand Island, NY) was added. The sample was placed onto the rotating shaker at 200

r.p.m., 37°C for 1 h. At the end of the cycle, the trypsin solution was discarded and 100 ml

of Dulbecco's modified Eagle's medium (DMEM) high-glucose (Gibco, Grand Island, NY)

containing 10% fetal bovine serum (FBS) (Valley Labs, Winchester, VA) and 100 IU/ml

penicillin, 100 μg/ml streptomycin, 0.25 μg/ml amphotercin B (Gibco, Grand Island, NY)

was added in order to halt trypsin digestion of the sample and maintain an aseptic

preparation. The sample was placed back onto the rotary shaker at 200 r.p.m., 37°C, for 24

h. After 24 h, the DMEM–FBS solution was discarded and 100 ml of the DNase-

free/RNase-free distilled water was added and the sample was placed onto the rotary shaker

at 200 r.p.m., 37°C for 24 h. The water wash was discarded and 100 ml of 1.5% peracetic

141

acid (Sigma, St. Louis, MO) solution with 2.0% Triton X-100 (Sigma, St. Louis, MO) in

distilled, deionized water (diH2O) was added and the sample was placed onto the rotary

shaker at 200 r.p.m., 37°C for 4 h. The solution was discarded and two 100 ml washes with

diH2O were performed, each for 12 h at 37°C and 200 r.p.m. on the rotary shaker. At the

end of the second wash, the sample was removed and placed into a clean, sterile 15 ml

conical tube and frozen for 24 h at −80°C. The scaffold then was lyophilized (Labconco,

Freeze Dry System, Kansas City, MO) for 24 h before being returned to the freezer and

stored at −80°C until further use. These samples are hereafter referred to as scaffolds.

Three fresh long digit tendons, one each from three Leghorn chickens were

aseptically harvested, and all of the soft tissue attachments and synovium were debrided.

The tendons then were placed immediately into sterile Dulbecco’s phosphate buffered

saline (DPBS, Sigma, St. Louis, MO) containing 100 IU/ml penicillin, 100 µg/ml

streptomycin, and 0.25 µg/ml amphotercin B solution on ice. Tendons then were minced

and transferred to 10 cm tissue culture plastic plates. Tenocyte culture media (DMEM high-

glucose, 10% FBS, 1% L-glutamine, 100 IU/ml penicillin, 100 µg/ml streptomycin, 0.25

µg/ml amphotercin B, and 25 mg of L-ascorbic acid/500 ml media) then was added to the

minced tendon. The tissue culture plates were covered and left undisturbed in an incubator

at 37°C, 5% CO2 for seven days, during which a mixed population of allogeneic tendon-

derived cells, containing tenocytes, migrated out of the fresh tissue and adhered to the

bottom of the plates. After 7 days, cells from the three plates were trypsinized with 0.05%

Trypsin-EDTA (Sigma, St. Louis, MO) at 37°C, 5% CO2 for five minutes. The minced

tissue was removed and the resulting cell suspension was centrifuged at 1,500 x g for 5

minutes. The supernatant was discarded and the cell pellet was re-suspended in 12 ml of

142

tenocyte culture media and re-plated onto 15 cm tissue culture plastic plates and allowed

to grow to 75% confluency. This process was performed for a total of 4 passages in order

to produce sufficient cells for seeding of scaffolds. Passage four cells from all three donors

were pooled prior to seeding of scaffolds. Six scaffolds, prepared as previously described,

were placed in individual sterile plastic tissue culture plates. Scaffolds then were

equilibrated in 70% ethanol in diH2O at 4°C for 24 hours to improve surface

wettability/decrease the surface tension on/within the scaffolds in order to minimize

agglomeration and maximize penetration of seeded cells. Un-seeded scaffolds were

prepared similarly. Scaffolds then were rinsed copiously with media, transferred to fresh

sterile tissue culture plates and covered with tenocyte media and equilibrated at 37°C, 5%

CO2 for 24 hours. Scaffolds then were removed and placed in fresh sterile tissue culture

plates and allowed to stand at room temperature for approximately one hour to remove

excess media. Scaffolds were seeded using a pipette (Eppendorf, Research 2100 Model

1000 µl, Westbury, NY) with 300 µl of a 10.0 X 106 cells/ml solution of the cells in media

applied in a dropwise fashion along the length of the scaffolds. The scaffolds then were

completely wetted by the media and allowed to equilibrate for 120 minutes at 37°C, 5%

CO2. The scaffolds then were rotated 180 degrees upon the circumferential axis of the

tendon and seeded with an additional 300 µl of the same cell suspension on their opposite

side. The seeded scaffolds were incubated at 37°C, 5% CO2 for 48 hours, undisturbed.

Following 48 hours of incubation, the seeded scaffolds were trimmed to a length of 30 mm.

A total of six seeded (SEEDED T7 No Strain) and six unseeded scaffolds (UNSEEDED T7

No Strain) were maintained in a 15 cm tissue culture dish in the absence of strain. Six

seeded scaffolds were maintained under cyclic mechanical strain in a commercially

143

available bioreactor (SEEDED T7 5% Strain BioRx). Media was changed in all samples

every 48 hours. Six unseeded control scaffolds were maintained under cyclic mechanical

strain to serve as controls to determine if application of cyclic strain alone would cause a

change in tensile properties due to fber alignment and loss of the “toe” region of the load-

elongation curve (UNSEEDED T7 5% Strain BioRx).

A commercially available DynaGen™ bioreactor system (Tissue Growth

Technologies, Minnetonka, MN) was used to apply cyclic strain to the seeded scaffolds.

Briefly, the DynaGen™ bioreactor system consisted of a TC-20 single axis displacement

simulator that can apply a maximum force of 20N and three L30-1C LigaGen™ bioreactor

chambers. Seeded scaffolds were held in place by two customized mechanical clamps. The

position of the upper clamp was controlled by the motor while the lower clamp was

stationary. GrowthWorks™ software was used to control the TC-20 single axis

displacement actuators and collect data. In the experimental setting, all bioreactor

specimens (5% Strain BioRx) were exposed to a constant, sinusoidal 5% mechanical strain

at 1Hz for one hour per day, for a total of seven days, in a stable environment of 37oC,

95% relative humidity, 5% CO2.

Tensile testing of fresh-frozen avian FDP tendons (n=8), unseeded scaffolds

maintained in the absence of strain (UNSEEDED T7 No Strain), (n=6), scaffolds seeded

with allogeneic cells and maintained in the absence of strain (SEEDED T7 No Strain),

(n=6), unseeded scaffolds maintained in the bioreactor under cyclic mechanical strain

(UNSEEDED T7 5% Strain BioRx), (n=6), as well as scaffolds seeded with the same

allogeneic cells and maintained in the bioreactor under cyclic mechanical strain (SEEDED

T7 5% Strain BioRx), (n=6) was performed. Additionally, un-seeded (n=6) and seeded

144

scaffolds (n=6) were tested after their initial 48h incubation (UNSEEDED T0 No Strain [48

hrs], SEEDED T0 No Strain [48 hrs]) to determine the baseline tensile properties of both,

the un-seeded scaffolds, and the seeded scaffolds 48h after seeding. Fresh-frozen FDP

tendons were removed from storage at −80°C and allowed to equilibrate in sterile DPBS

at 37°C for 30 minutes. Immediately prior to testing, fresh-frozen tendons and un-

seeded/seeded scaffolds were cut into “dogbone” specimens using a custom made punch

(Freeman, Fremont, OH), their ends were wrapped with saline-soaked gauze which

completely eliminated slippage at the tendon-grip interface, and they were placed into

custom made grips on an uniaxial load frame (Instron 5544, Needham, MA) for tensile

testing. Samples were measured prior to testing with digital calipers to determine the

individual cross-sectional area of each sample (~1 mm2) and length (15 mm). Tendons

were pre-loaded to 0.1 N, and then pre-conditioned 10 times to 0.1mm extension prior to

being loaded to failure at a rate of 10 mm/min (strain rate of 1.11%/s). Tendons were kept

moist during testing with a mist of DPBS. Ultimate tensile stress (UTS, MPa) at break was

calculated using the maximal load (N) recorded during each test and cross-sectional area

of each specimen measured immediately prior to testing. Young’s modulus (Y, MPa) and

Stiffness (S, N/mm) were calculated from the linear portion of stress–strain curves

generated from the load and displacement data obtained from the instrument and the cross-

sectional area of each specimen measured immediately prior to testing.

At T0 [48 hrs], and after 7 days in the presence or absence of cyclic strain (T7),

seeded scaffolds were rinsed with fresh media, dried briefly, then frozen in liquid nitrogen,

and the total RNA was extracted with Trireagent (Ambion, Austin, TX). Quantity and

purity of the RNA was assessed with a NanoDrop ND-1000 spectrophotometer (Nano-

145

Drop Technologies, Wilmington, DE). The mRNA was transcribed to cDNA using random

hexamers (Invitrogen) and Superscript II (Invitrogen). Taqman1 gene expression assays

(Applied Biosystems, Foster City, CA) were used to perform quantitative real-time

polymerase chain reaction (qRTPCR). Probed sequences included gallus gallus Collagen

I, Collagen III, MMP-2, and Tenomodulin. Relative quantification (DD-CT) RTPCR was

performed on an Applied Biosystems 7900HT. GAPDH was used as the endogenous

control and was constant throughout samples. Data were normalized to the average of the

T0 [48 hrs] samples for simplification of presentation.

The results of tensile testing and mRNA expression were collected and recorded in

a spreadsheet. For statistical analysis, the mean and standard error of the mean (SEM) of

investigated variables were calculated. To test if any statistical differences existed between

the tensile properties among the different groups, a one-way ANOVA and post hoc testing

using the Holm-Sidak method was used. To test if any statistical differences in mRNA

expression existed among the different groups, an unpaired Student’s t-test with unequal

variance was used. A p-value of < 0.05 was considered statistically significant. All

calculations were performed using SigmaStat Version 3.5 (Systat Software, San Jose, CA).

Results

Tensile Testing

The failure site of all samples during tensile testing occurred within the

midsubstance portion of the specimen and no disruption or slippage occurred at the

interface between the clamp and tendon. Therefore, none of the specimens had to be

excluded from the tensile testing. Ultimate tensile stress (UTS), Young’s modulus (Y), and

stiffness (S) were calculated for all samples (Table 1). UTS, Y, and S of the un-seeded

146

scaffolds maintained in the absence of strain for 48 hours (UNSEEDED T0 No Strain [48

hrs]) was not significantly different than that of either the un-seeded scaffolds maintained

in the absence of strain for 7 days (UNSEEDED T7 No Strain), the un-seeded scaffolds

maintained under cyclic strain in the bioreactor for 7 days (UNSEEDED T7 5% Strain

BioRx), or the fresh-frozen FDP tendons (Figure 1A, C, E). There was also no significant

difference in tensile properties between all unseeded scaffolds and the seeded scaffolds

maintained under cyclic strain at 7 days (SEEDED T7 5% Strain BioRx). Similarly, no

significant difference in the observed modulus or the observed stiffness of seeded scaffolds

maintained under strain for 7 days (SEEDED T7 5% Strain BioRx) was observed when

compared to fresh-frozen FDP tendons (Figure 1B, D). A significantly lower ultimate

tensile stress was observed for seeded scaffolds maintained under cyclic strain for 7 days

(SEEDED T7 5% Strain BioRx) when compared to fresh-frozen FDP tendons (Figure 1F).

A significant difference was found between the UTS, modulus, and stiffness of both the

seeded scaffolds maintained in the absence of cyclic strain after 48h of incubation

(SEEDED T0 No Strain [48 hrs]), and the seeded scaffolds maintained in the absence of

cyclic strain after 7 days (SEEDED T7 No Strain) when compared to the fresh-frozen FDP

tendons and the seeded scaffolds maintained under cyclic strain after 7 days in culture

(SEEDED T7 5% Strain BioRx), (Figure 1B, D, F).

mRNA Expression

No significant difference in the expression of Collagen I by seeded cells was

observed among any group at any time point (Figure 2A). Collagen III expression was

significantly lower in seeded scaffolds maintained under cyclic strain at day 7 (SEEDED

T7 5% Strain BioRx) when compared to seeded scaffolds at T0 (Figure 2B). Expression of

147

Collagen III in seeded scaffolds after 7 days in the absence of cyclic strain (SEEDED T7

No Strain) was also greater than that observed for seeded scaffolds maintained under cyclic

strain at day 7 (SEEDED T7 5% Strain BioRx), but the difference was not statistically

significant (Figure 2B). There was no significant difference in Collagen III expression of

seeded scaffolds at T0 (SEEDED T0 No Strain [48 hrs]) and seeded scaffolds analyzed after

7 days in the absence of cyclic strain (SEEDED T7 No Strain), (Figure 2B). MMP-2

expression was significantly higher in seeded scaffolds at T0 (SEEDED T0 No Strain [48

hrs]) when compared to seeded scaffolds after 7 days in the absence of cyclic strain

(SEEDED T7 No Strain), (Figure 2C). No significant difference was observed for MMP-2

expression between seeded scaffolds maintained under cyclic strain at day 7 (SEEDED T7

5% Strain BioRx) and seeded scaffolds after 7 days in the absence of cyclic strain

(SEEDED T7 No Strain), (Figure 2C). A lower level of MMP-2 expression was observed

in seeded scaffolds maintained under cyclic strain at day 7 (SEEDED T7 5% Strain BioRx)

when compared to seeded scaffolds at T0 (SEEDED T0 No Strain [48 hrs]), p=0.08 (Figure

2C). A significant increase in tenomodulin expression in seeded scaffolds maintained under

cyclic strain at day 7 (SEEDED T7 5% Strain BioRx) was observed in comparison to seeded

scaffolds at T0 (SEEDED T0 No Strain [48 hrs]), (Figure 2D). No significant difference in

tenomodulin expression was observed between seeded scaffolds at T0 (SEEDED T0 No

Strain [48 hrs]) and seeded scaffolds after 7 days in the absence of cyclic strain (SEEDED

T7 No Strain), (Figure 2D). No significant difference was observed for tenomodulin

expression between seeded scaffolds maintained under cyclic strain at day 7 (SEEDED T7

5% Strain BioRx) and seeded scaffolds after 7 days in the absence of cyclic strain

(SEEDED T7 No Strain), (Figure 2C).

148

DISCUSSION

Currently, there is no optimal graft choice for the repair of flexor tendon injuries,

especially those requiring multiple tendon reconstructions. We have previously described

the development of a decellularized, biocompatible, architecturally-optimized scaffold

derived from avian flexor digitorum profundus tendon suitable for flexor tendon

reconstruction [26]. The present study describes the seeding of allogeneic tenocytes onto

that scaffold and the resulting tensile properties of seeded scaffolds and associated cellular

mRNA expression of seeded cells after culture of seeded scaffolds in the presence or

absence of cyclic strain.

The tensile properties of seeded scaffolds decreased significantly as quickly as 48

hours after seeding in the absence of strain. A significantly higher level of MMP-2

expression was observed in seeded scaffolds at 48 hours compared to 7 days when scaffolds

were maintained without cyclic strain. Scaffolds in the presence of cyclic strain exhibited

a lower level of MMP-2 expression, albeit not significantly lower (p=0.08), after 7 days in

comparison to seeded scaffolds 48 hours after seeding. No difference in MMP-2 expression

was observed between seeded scaffolds maintained in the presence or absence of cyclic

strain after 7 days. MMP-2 is a matrix metalloproteinase that has a well-established role

in the remodeling and development of tendons [22-24]. Therefore, increased expression of

this gelatinase may play a role in the initial degradation of tensile properties observed in

scaffolds in the first 48 hours following seeding. Others have found that after an initial

degradation in tensile properties in “under-stimulated” tenocyte seeded scaffolds, peak

stresses can be restored by the addition of protease inhibitors or mechanical loading [27].

Similarly, our results suggest a protease-mediated degradation of tensile properties that

149

occurs quickly after seeding in the setting of inadequate mechanical stimulation. However,

a firm conclusion cannot be made that elevated MMP-2 expression in the absence of

mechanical strain is directly responsible, as differences in expression between the seeded

scaffolds maintained in the absence of cyclic strain for 7 days and that of the seeded

scaffolds after maintenance under cyclic strain at 7 days did not reach statistical

significance. Rather we postulate that there is an initial increase in MMP-2 expression

within 48 hours after seeding in the absence of mechanical stimulation that takes place and

is possibly responsible for the initial degradation of tensile properties.

An increased level of Collagen III expression was observed in scaffolds 48 hours

after seeding when compared to scaffolds maintained under cyclic strain after 7 days.

Collagen III is associated with scar formation during tendon healing as well as the

decreased tensile properties associated with scar tissue [28]. No difference in the level of

Collagen I expression was observed between any group, consistent with prior studies [21].

Therefore, the higher level of Collagen III expression may represent a shift towards the

formation of scar initially after seeding which is decreased significantly after exposure to

cyclic strain, leading to the observed recovery of tensile properties similar to those

observed by others [28]. Tensile properties observed for seeded scaffolds maintained in the

absence of cyclic strain for 7 days were unchanged from those observed 48 hours after

seeding. Again, these results support the initial formation of inferior tissue in the absence

of strain, or rather degradation of the scaffold, which does not appear to be reversed in the

absence of strain after 7 days, consistent with prior results for other scaffolds [27].

The tensile properties of the seeded scaffolds were restored so that no statistically

significant difference was observed when compared to unseeded scaffolds. Similarly, the

150

modulus and stiffness of seeded scaffolds maintained under cyclic strain for 7 days were

not significantly different from those of fresh-frozen tendons. Tenomodulin is a marker for

tenocytes and late tendon formation [25]. A significantly higher level of tenomodulin

expression by cells seeded in scaffolds maintained under cyclic strain for 7 days was

observed in comparison to cells 48 hours after seeding in the absence of cyclic strain. The

increased expression of this gene, in association with the observed restoration of tensile

properties, provides evidence that seeded cells and seeded scaffolds undergo a process that

results in a tissue-engineered construct that more closely resembles native tendon when

compared to seeded scaffolds at early time points and cultured for longer time points in the

absence of strain. Again, a firm conclusion cannot be made that elevated tenomodulin

expression is a direct result of the presence of mechanical strain. Differences in expression

between the seeded scaffolds maintained in the absence and presence of cyclic strain for 7

days did not reach statistical significance. However, an elevated level of expression was

observed in seeded scaffolds after exposure to mechanical strain that was not observed in

the seeded scaffolds maintained in its absence. Therefore, it is possible that, after seeding

in an unstrained environment, seeded cells exposed to subsequent strain began to express

the specific tenocyte marker, tenomodulin, indicating a possible cellular shift in gene

expression toward a more tendon-like phenotype temporally associated with a restoration

of tensile properties. Further studies investigating the cellular and molecular events

associated with remodeling of seeded, decellularized scaffolds are warranted to fully

elucidate this process.

One limitation of the present study is that we focused on the tensile properties of

graft remodeling to the exclusion of compositional changes. However, ongoing

151

investigations in our laboratory are analyzing changes in collagen, proteoglycans and ultra-

structure of seeded scaffolds. The allogeneic cell population used for seeding likely

contains cells other than tenocytes and may have influenced the observed expression of

genes of interest. Cell contamination by mixed-population cells isolated from tendon

explants cultures and phenotypic drift of isolated cells may have also contributed to the

observed pattern of mRNA expression by seeded cells. Similarly, plausible that the

transduction of mechanical signals may decrease as strain decreases from its maximum at

cells seeded within and on the immediate surface of the scaffold to those at its periphery.

Thus, the contribution of the mechanically-stimulated cells in the matrix and on the surface

of the scaffold may be affected by the cells at the periphery which receive diminished

mechanical signals necessary for altered gene expression.

Another limitation of the study is that all specimens were subjected to one cyclic

stretching regimen. This regimen was based on an extensive literature review and was

designed to closely approximate in vivo conditions associated with loads experienced in

vivo by the donor species and by the loads present in the linear portion of the stress-strain

curve of the scaffold [26, 29-32]. Physiologic strain of tendons and ligaments has been

shown to occur between 2-5% at a level that is equivalent to less than 25% of the ultimate

tensile stress [33,34]. As previously stated, it is likely that not all cells are subjected to the

same strain. Therefore, we chose to base our regimen on 125% of the maximal allowable

physiologic strain that could be applied without theoretical damage to tendon fiber bundles

in order to produce an effect on seeded cells [35]. Lastly, it has been noted that the accuracy

of cross-sectional measurement by digital calipers may be less than that of non-contact

methods such as a laser micrometry. Woo et al. have shown that for homogenous,

152

rectangular specimens, such as those analyzed in our study, the accuracy of digital calipers

is acceptable when compared to that obtained using laser micrometer or other non-contact

methods [36].

We have previously shown that our scaffold is decellularized to decrease

inflammation, has increased porosity to promote cellular infiltration and influx of nutrients,

is cyto-compatible in vitro and in vivo, and can be easily derived from a readily available

source [26]. Such a scaffold, possibly derived from human allograft or xenograft sources,

would be potentially useful for the treatment of tendon injuries, especially those associated

with trauma or tumor where sufficient quantities of autograft may not be available for

reconstruction.

Absence of mechanical strain and exercise in patients are known to be associated

with a loss of tensile properties in vivo. The present study shows that seeded tendon

scaffolds are associated with an initial loss of tensile properties soon after seeding in vitro.

However, tensile properties can be restored in the presence of cyclic strain. Therefore,

seeded and unseeded scaffolds that are to be implanted in vivo should be maintained in the

presence of cyclic strain to prevent degradation by either seeded cells or infiltrating host

cells. Failure to initiate early motion may result in a delay in return of function or possible

implant failure.

Lastly, although no significant improvement in the tensile properties of our scaffold was

observed after seeding with allogeneic cells and mechanical strain in vitro, bioreactor

preconditioning may still prove useful once constructs are implanted in vivo. Bioreactor

preconditioning may allow for preservation or even optimization of the biologic and tensile

153

properties of autologous cell-seeded scaffolds prior to implantation, potentially allowing

more rapid in vivo incorporation and remodeling.

154

CONCLUSIONS

The effect of cyclic strain upon the tensile properties and mRNA expression of a

cell-seeded tendon scaffold potentially suitable for flexor tendon reconstruction was

investigated. The observed results demonstrate that tensile properties of cell-seeded

scaffolds decrease significantly within 48 hours after introducing the cells, in the absence

of mechanical strain. The decrease is associated with elevated MMP-2 and collagen III

expression by seeded cells. MMP-2 is associated with tendon degradation and collagen III

is associated with inferior tensile properties when compared to collagen I, the dominant

collagen in tendons. Tensile properties were restored in the presence of physiologic

mechanical strain, but not in its absence. The restoration of tensile properties was

associated with increased levels of tenomodulin expression by seeded cells. Tenomodulin

is a specific marker for tenocytes and thus, based on mRNA expression and tensile testing,

the construct more closely resembles native tendon after mechanical stimulation.

In the absence of mechanical strain, tensile properties of seeded, decellularized

tendon scaffolds degrade rapidly and are never recovered. However, in the presence of

mechanical strain, the seeded scaffolds are able to re-gain their tensile properties and more

closely resembles native tendon. Therefore, if decellularized tendon scaffolds are to be

used in vivo in the presence of autologous cells, scaffolds should be maintained in the

presence of mechanical strain in vitro and in vivo to optimize function after implantation.

155

COMPETING INTERESTS

PWW and MEV have been issued an allowance of claims on US Patent Application No.

11/738,258. This patent describes the method of production of the acellular scaffold used

in this manuscript. GGP is a consultant for MTF®. An unrestricted, educational grant from

Smith & Nephew® was provided to PWW, TMS and GGP during completion of this

manuscript.

156

ACKNOWLEDGEMENTS

The authors and this work were supported by an educational grant by Smith & Nephew

(PWW, TMS, GGP), a Resident Clinician Scientist Training Grant from the Orthopaedic

Research and Education Foundation (PWW, LAK), and a Resident Clinician Scientist

Training Grant from the Orthopaedic Research and Education Foundation to Dr. Seyler.

Use of the DynaGen bioreactor system was generously provided by Tissue Growth

Technologies.

157

REFERENCES

1. Sharma P, Maffulli N: Tendon injury and tendinopathy: healing and repair. J Bone

Joint Surg Am 2005, 87:187-202

2. Lin TW, Cardenas L, Soslowsky LJ: Biomechanics of tendon injury and repair. J

Biomech 2004, 37: 865-877

3. Benjamin M, and Ralphs JR: The cell and developmental biology of tendons and

ligaments. Int Rev Cytol 2000, 196:85-130

4. Kannus P, Josza L, and Jarvinnen M: Basic science of tendons. In Principles and

Practice of Orthopaedic Sports Medicine. Edited by Garrett WE, Speer KP, and

Kirkendall DT. Philadelphia, Pennsylvania: Lippincott Williams and Wilkins;

2000:21-37.

5. Strickland JW: Flexor tendon repair. Hand Clin 1985, 155-168

6. Strickland JW: Development of flexor tendon surgery: twenty-five years of

progress. J Hand Surg Am 2000, 25:214-235

7. Tang JB: Clinical outcomes associated with flexor tendon repair. Hand Clin 2005,

21:199-210

8. Hasslund S, Jacobson JA, Dadali T, Basile P, Ulrich-Vinther M, Soballe K, Schwarz

EM, O'Keefe RJ, Mitten DJ, and Awad HA: Adhesions in a murine flexor tendon

graft model: Autograft versus allograft reconstruction. J Orthop Res 2008, 26:824-

833

9. Leversedge FJ, Zelouf D, Williams C, Gelberman RH, and Seiler JG III: Flexor

tendon grafting to the hand: an assessment of the intrasynovial donor tendon-A

preliminary single-cohort study. J Hand Surg Am 2000, 25:721-730

158

10. Strickland JW: Flexor tendon surgery. Part 2: Free tendon grafts and tenolysis. J

Hand Surg Br 1989, 14:368-382

11. Zhang AY, and Chang J: Tissue engineering of flexor tendons. Clin Plast Surg

2003, 30:565-572

12. Costa MA, Wu C, Pham BV, Chong AK, Pham HM, and Chang J: Tissue

engineering of flexor tendons: optimization of tenocyte proliferation using growth

factor supplementation. Tissue Eng 2006, 12:1937-1943

13. Cao Y, Liu Y, Liu W, Shan Q, Buonocore SD, and Cui L: Bridging tendon defects

using autologous tenocyte engineered tendon in a hen model. Plast Reconstr Surg

2002, 110:1280-1289

14. Liu W, Chen B, Deng D, Xu F, Cui L, and Cao Y: Repair of tendon defect with

dermal fibroblast engineered tendon in a porcine model. Tissue Eng 2006, 12:775-

788

15. Derwin KA, Baker AR, Spragg RK, Leigh DR, Iannotti JP: Commercial

extracellular matrix scaffolds for rotator cuff tendon repair: Biomechanical,

biochemical and cellular properties. J Bone Joint Surg Am 2006, 88: 2665-2672

16. Garvin J, Qi J, Maloney M, Banes AJ: Novel system for engineering of bioartificial

tendons and application of mechanical load. Tissue Eng 2003, 9: 967-979

17. Buckwalter JA: Effects of early range of motion on healing of musculoskeletal

tissues. Hand Clin 1996, 12:13-24

18. Buckwalter JA: vs. rest in the treatment of bone, soft tissue and joint injuries. Iowa

Orthop J 1995, 15:29-42

159

19. Cao D, Wei L, Wei X, Xu F, Cui L, Cao Y: In vitro tendon engineering with avian

tenocytes and polyglycolic acids: a preliminary report. Tissue Eng 2006, 12: 1369-

1377

20. Calve S, Dennis RG, Kosnik PE 2nd, Baar K, Grosh K, Arruda EM: Engineering

of functional tendon. Tissue Eng 2004, 10:755-761

21. Altman GH, Horan RL, Martin I, Farhadi J, Stark PR, Volloch V, Richmond JC,

Vunjak-Novakovic G, Kaplan DL: Cell differentiation by mechanical stress.

FASEB J 2002, 16:270-272

22. Jung JC, Wang PX, Zhang G, Ezura Y, Fini ME, Birk DE: Collagen fibril growth

during chicken tendon development:matrix metalloproteinase-2 and its activation.

Cell Tissue Res 2009, 336:79-89

23. Kyriakides TR, Zhu YH, Smith LT, Bain SD, Yang Z, Lin MT,Danielson KG,

Iozzo RV, LaMarca M, McKinney CE, Ginns EI, Bornstein P: Mice that lack

thrombospondin 2 display connective tissue abnormalities that are associated with

disordered collagen fibrillogenesis, an increased vascular density, and a bleeding

diathesis. J Cell Biol 1998, 140:419-430

24. Yang Z, Kyriakides TR, Bornstein P: Matricellular proteins as modulators of cell-

matrix interactions: adhesive defect in thrombospondin 2-null fibroblasts is a

consequence of increased levels of matrix metalloproteinase-2. Mol Biol Cell 2000,

11:3353-3364

25. Shukunami C, Takimoto A, Oro M, Hiraki Y: Scleraxis positively regulates the

expression of tenomodulin, a differentiation marker of tenocytes. Developmental

Biology 2006, 298: 234–247

160

26. Whitlock PW, Smith TL, Poehling GG, Shilt JS, and Van Dyke ME: A naturally

derived, cytocompatible, and architecturally optimized scaffold for tendon and

ligament regeneration. Biomaterials 2007, 28:4321-4329

27. Cousineau-Pelletier P, Langelier E: Relative contributions of mechanical

degradation, enzymatic degradation, and repair of the extracellular matrix on the

response of tendons to under- and over-stimultaions in vitro. J Orthop Res 2009,

28: 204-210

28. Birk DE, and Mayne R: Localization of collagen types I, III and V during tendon

development. Changes in collagen types I and III are correlated with changes in

fibril diameter. Eur J Cell Biol 1997, 72:352-361

29. Butler DL, Goldstein SA, Guilak F: Functional tissue engineering: the role of

biomechanics. J Biomech Eng 2000, 122:570-575

30. Butler DL, Juncosa-Melvin N, Boivin GP, Galloway MT, Shearn JT, Gooch C,

Awad H: Functional tissue engineering for tendon repair: A multidisciplinary

strategy using mesenchymal stem cells, bioscaffolds, and mechanical stimulation.

J Orthop Res 2008, 26:1-9

31. Freed LE, Guilak F, Guo XE, Gray ML, Tranquillo R, Holmes JW, Radisic M,

Sefton MV, Kaplan D, and Vunjak-Novakovic G: Advanced tools for tissue

engineering: scaffolds, bioreactors, and signaling. Tissue Eng 2006, 12: 3285-3305

32. Vunjak-Novakovic G, Altman G, Horan R, Kaplan DL: Tissue engineering of

ligaments. Annu Rev Biomed Eng 2004, 6:131-156

33. Butler DL, Grood ES, Noyes FR: Biomechanics of ligaments and tendons. Exerc

Sport Sci Rev 1978, 6: 125-181

161

34. Fung YCB. Biomechanics: Mechanical Properties of Living Tissues. New York:

Springer-Verlag; 1981:222.

35. Kear M, Smith RN: A method for recording tendon strain in sheep during

locomotion. Acta Orthop Scand 1975, 46: 896-905

36. Woo SL, Danto MI, Ohland KJ, Lee TQ, Newton PO: The use of laser micrometer

system to determine the cross-sectional shape and area of ligaments: a comparative

study with two existing methods. J Biomech Eng 1990, 112:426-431

162

FIGURES AND TABLES

Figure 1. Tensile Properties (A), Young’s modulus of UNSEEDED scaffolds after 48 hour initial culture (T0 No Strain [48 hrs]), 7 days in culture in the absence of cyclic strain (T7 No Strain), 7 days in culture in the presence of cyclic strain (T7 5% Strain BioRx) and fresh-frozen FDP tendons; (C), Stiffness of UNSEEDED scaffolds after 48 hour initial culture (T0 No Strain [48 hrs]), 7 days in culture in the absence of cyclic strain (T7 No Strain), 7 days in culture in the presence of cyclic strain (T7 5% Strain BioRx) and fresh-frozen FDP tendons; (E), Ultimate Tensile Stress of UNSEEDED scaffolds after 48 hour initial culture (T0 No Strain [48 hrs]), 7 days in culture in the absence of cyclic strain (T7 No Strain), 7 days in culture in the presence of cyclic strain (T7 5% Strain BioRx) and fresh-frozen FDP tendons. (B), Young’s modulus of SEEDED scaffolds after 48 hour initial culture (T0 No Strain [48 hrs]), 7 days in culture in the absence of cyclic strain (T7 No Strain), 7 days in culture in the presence of cyclic strain (T7 5% Strain BioRx) and fresh-frozen FDP tendons; (D), Stiffness of SEEDED scaffolds after 48 hour initial culture (T0 No Strain [48 hrs]), 7 days in culture in the absence of cyclic strain (T7 No Strain), 7 days in culture in the presence of cyclic strain (T7 5% Strain BioRx) and fresh-frozen FDP tendons; (F), Ultimate Tensile Stress of SEEDED scaffolds after 48 hour initial culture (T0 No Strain [48 hrs]), 7 days in culture in the absence of cyclic strain (T7 No Strain), 7 days in culture in the presence of cyclic strain (T7 5% Strain BioRx) and fresh-frozen FDP tendons. Bars represent significant differences, p<0.05. Cyclic strain parameters: f=1 Hz, duration=1 hour per day, time=7 days.

163

164

Figure 2. mRNA Expression Relative gene expression by cells of seeded scaffolds after 48 hour initial culture (T0 No Strain [48 hrs]), 7 days in culture in the absence of cyclic strain (T7 No Strain), 7 days in culture in the presence of cyclic strain (T7 5% Strain BioRx). (A) Collagen I, (B) Collagen III, (C) MMP-2 (matrix metalloproteinase-2), (D) Tenomodulin. Gene expression was normalized. Bars represent significant differences, p<0.05. Cyclic strain parameters: f=1 Hz, duration=1 hour per day, time=7 days.

165

Table 1. Tensile Properties

Table 1. Tensile Properties

Sample UTS [Mpa] Y [MPA] S [N/mm]

UNSEEDED T0 No Strain[48 hrs] 40.54 ± 6.94 639.29 ± 58.90 18.82 ± 1.73

UNSEEDED T7 No Strain 60.20 ± 5.06 697.04 ± 26.99 27.24 ± 1.86

UNSEEDED T7 5% Strain BioRx 59.43 ± 13.98 672.68 ± 101.00 26.16 ± 3.98

Fresh-Frozen FDP Tendon 76.97 ± 5.99 725.05 ± 24.20 28.66 ± 1.65

SEEDED T7 5% Strain BioRx 56.37 ± 4.34 728.93 ± 39.50 24.83 ± 1.22

SEEDED T7 No Strain 31.46 ± 6.63 437.25 ± 54.59 14.36 ± 2.08

SEEDED T0 No Strain[48 hrs] 33.15 ± 6.97 412.84 ± 52.22 15.63 ± 2.08

Ultimate Tensile Stress (UTS), Young’s modulus (Y), and Stiffness (S) of fresh-frozen FDP tendons, and seeded and un-seeded scaffolds after 48 hours (T0) and 7 days (T7) in culture in the presence (5% Strain BioRx) or absence (No Strain) of cyclic strain. Bars represent significant differences, p<0.05. Cyclic strain parameters: f=1 Hz, duration=1 hour per day, time=7 days.

166

CHAPTER 6

SUMMARY, CLINICAL RELEVANCE, & FUTURE DIRECTIONS

Thorsten M. Seyler, M.D.

167

SUMMARY OF DOCTORAL THESIS

Decellularized tissues have been used successfully in several areas of tissue

engineering. Numerous decellularization protocols have been utilized contributing to the

knowledge of techniques that can be used to adequately decellularize tissues.36,62,67,70 The

data presented in this thesis suggest that a novel decellularization and oxidation protocol

that combines physical, chemical, and enzymatic modalities is effective for decellularizing

tendons from various sources including chicken flexor digitorum profundus tendons,

human Achilles tendon, and pig patellar tendons.43,66,69 This protocol is effective in

reducing cellular material and lipids in general and decreasing the expression of antigenic

material such as the α-Gal epitope in xenogenic sources. Both the removal of cellular nad

nuclear material as well as the reduced expression of the α-Gal epitope may reduce the

immunogenicity of scaffolds.9,20,36,59,62 Exposure to enzymatic treatment with trypsin and

oxidation with peracetic acid disrupts cell-matrix interactions and increases total tissue

porosity and pore size without compromising biomechanical properties of the source

tissue.43,66,69 The retention of the extracellular matrix and the modified scaffold structure

facilitate re-population by host cells.66 This process is critically important because the

maintenance of the biomechanical properties allows for the slow in vivo remodeling

process while preserving function.69 The incorporation of peracetic acid and lyophilization

steps in the protocol also have shown to be effective in virus inactivation and tissue

sterilization making this protocol a potential method for the preparation of human allograft

tissue.66

168

In summary, the tissue-engineered scaffolds produced using the novel

decellularization and oxidation protocol demonstrate:

(1) Decreased DNA content and amount of cellular material leading to reduced

immunogenic potential

(2) Non-cytotoxic and biocompatible in vitro and in vivo which make it an ideal

candidate for clinical translation

(3) Alterations in the micro-architecture including increased porosity and pore size

promoting cell infiltration and re-population

(4) Preservation of tensile properties which offers rapid functionality and suitability

for clinical translation

(5) Preservation of extracellular matrix serves as a structural and logistic template

for cell attachment which also facilitates site-specific differentiation and

remodeling

(6) Can be successfully seeded with progenitor cells and subsequently may permit

formation of functional tissues

(7) Mechanical loading of seeded scaffolds improves and maintains the tensile

properties which ultimately enhances the rate of tissue development

(8) Virus inactivation and tissue sterilization reducing the risk of disease

transmission

(9) Reduction of xenoantigens such as the α-gal epitope decreasing the risk for

xenogenic tissue rejection

169

CLINICAL RELEVANCE TO ORTHOPAEDIC SURGERY

Tendon and ligament injuries place a significant burden on the United States

economy, accounting for nearly 32 million orthopaedic musculoskeletal injuries

annually.50 These ligamentous and tendinous injuries include tears to the rotator cuff and

anterior cruciate ligament; tendon injuries of the hand and foot; and damage to the

medial/lateral collateral ligament of the elbow. The goals of orthopaedic surgery are to

restore normal joint motion, reduce pain, return patients to full function, and prevent further

degenerative disease. Despite many recent surgical advances, surgeons continue to seek

alternative reconstruction techniques and graft choices in an attempt to meet the patient’s

functional demands, reduce complications, and improve clinical outcomes.

The popularity and use of musculoskeletal allograft tissue in a wide variety of

orthopaedic procedures has dramatically increased over the last 20 years. In a statement

from 2011, the American Academy of Orthopaedic Surgeons (AAOS) believes that for

appropriate patients, musculoskeletal allografts represent a therapeutic alternative.61

Potential advantages of allografts include decreased donor site morbidity, immediate

availability of various graft sizes (off-the-shelf), less postoperative pain, better

postoperative function, and lower incidence of postoperative arthrofibrosis.

The greatest disadvantage of allografts is the potential risk of disease transmission.

Contamination of allografts can occur while the tissue remains confined to the donor’s

corpus and during or after tissue removal because of improper tissue processing

procedures.46,68

In the past, the two methods employed to avoid disease transmission were graft

sterilization using ethylene oxide treatment or gamma irradiation. Ethylene oxide is no

170

longer used because of associated post-operative synovitis and intra-articular graft

destruction. Gamma irradiation is thought to create free radicals and modify nucleic acids

leading to virus and bacterial destruction. However, it has been also associated with

disruption of the structural integrity of the graft.24 Initial concerns of graft weakness

associated with using ethylene oxide or high dose gamma irradiation sterilization have been

addressed by employing a multimodal process that involves sterile harvesting of tissue

from the donor, rigorous donor screening/testing, antibiotic soaking, low dose irradiation

(1.2-1.8 Mrad), and use of a final preservation technique such as lyophilization or

freezing.32,34 Despite these advances in graft sterilization, one of the major drawbacks of

using allografts is slow incorporation into host bone and graft remodeling, which may lead

to reduced graft strength resulting in early failure.11

Naturally derived extracellular-matrix scaffolds have evolved as a viable option for

tendon and ligament repair21. However, some materials, such as small intestinal

submucosa, lack the initial strength to adequately replace tissue structures; instead they

have been utilized successfully to augment soft tissue repairs.16,35,44,49 A decellularization

and oxidation protocol is presented that can produce a tissue-engineered scaffold that

possesses mechanical properties similar to the native tissue and can maintain its mechanical

properties until remodeling has occurred and improved functionality. Application of this

procedure may allow patients to have an expedited return to work and recreational activities

after undergoing orthopaedic ligament and tendon reconstruction surgery.

171

FUTURE DIRECTION FOR RESEARCH

Our research efforts are centered on addressing clinical problems, and ultimately,

to improve patient care by translating this basic-science knowledge into clinical practice.

Tendon and ligament injuries pose a significant clinical problem that is inadequately

addressed by current orthopaedic interventions. Tissue-engineered scaffolds offer a

promising alternative to current graft options by delivering a graft with favorable

mechanical properties to the site of injury, providing functional utilization of reconstructed

tendons and ligaments immediately after surgery, as well as graft integration and healing

in the months to years following surgery. The experiments in this thesis demonstrated that

a novel decellularization and oxidation protocol that combines physical, chemical, and

enzymatic modalities can be used to produce naturally derived scaffolds with significant

clinical potential. Research using a small animal study and an ectopic model of

xenotransplantation in hon-human primates has been successful. Although this represents

a critical step in building a foundation for future studies and demonstrating the feasibility

of using a tissue-engineered scaffold for ligament and tendon reconstruction, several

challenges must be addressed prior to translation of this method into the clinic. Among

these future studies, scaffold processing and exploration of tissue sources are among the

most important variables that impact the host response and remodeling outcome. For

clinical translation, many hurdles remain to be overcome such a defining the optimal donor

source and addressing the key concern when using xenograft tissues; immunogenicity and

its impact on graft rejection and long-term graft survival.

172

REFERENCES

1. Weiss, A. B.; Blazina, M. E.; Goldstein, A. R.; and Alexander, H.: Ligament

replacement with an absorbable copolymer carbon fiber scaffold--early clinical experience.

Clin Orthop Relat Res, (196): 77-85, 1985.

2. Bolton, C. W., and Bruchman, W. C.: The GORE-TEX expanded

polytetrafluoroethylene prosthetic ligament. An in vitro and in vivo evaluation. Clin Orthop

Relat Res, (196): 202-13, 1985.

3. Nikolaou, P. K.; Seaber, A. V.; Glisson, R. R.; Ribbeck, B. M.; and Bassett, F. H.,

3rd: Anterior cruciate ligament allograft transplantation. Long-term function, histology,

revascularization, and operative technique. Am J Sports Med, 14(5): 348-60, 1986.

4. Olson, E. J.; Kang, J. D.; Fu, F. H.; Georgescu, H. I.; Mason, G. C.; and Evans, C.

H.: The biochemical and histological effects of artificial ligament wear particles: in vitro

and in vivo studies. Am J Sports Med, 16(6): 558-70, 1988.

5. Lukianov, A. V.; Richmond, J. C.; Barrett, G. R.; and Gillquist, J.: A multicenter

study on the results of anterior cruciate ligament reconstruction using a Dacron ligament

prosthesis in "salvage" cases. Am J Sports Med, 17(3): 380-5; discussion 385-6, 1989.

6. Schindhelm, K.; Rogers, G. J.; Milthorpe, B. K.; Hall, P. J.; Howlett, C. R.; Sekel,

R.; Goldberg, J.; and Viglione, W.: Autograft and Leeds-Keio reconstructions of the ovine

anterior cruciate ligament. Clin Orthop Relat Res, (267): 278-93, 1991.

7. Bigliani, L. U.; Cordasco, F. A.; McIlveen, S. J.; and Musso, E. S.: Operative

treatment of failed repairs of the rotator cuff. J Bone Joint Surg Am, 74(10): 1505-15, 1992.

173

8. McCarthy, D. M.; Tolin, B. S.; and Schwenderman, L.: In Anterior cruciate

ligament: Current and future concepts, pp. 343-356. Edited, 343-356, New York, Raven,

1993.

9. Cooper, D. K.; Good, A. H.; Koren, E.; Oriol, R.; Malcolm, A. J.; Ippolito, R. M.;

Neethling, F. A.; Ye, Y.; Romano, E.; and Zuhdi, N.: Identification of alpha-galactosyl and

other carbohydrate epitopes that are bound by human anti-pig antibodies: relevance to

discordant xenografting in man. Transpl Immunol, 1(3): 198-205, 1993.

10. Margevicius, K. J.; Claes, L. E.; Durselen, L.; and Hanselmann, K.: Identification

and distribution of synthetic ligament wear particles in sheep. J Biomed Mater Res, 31(3):

319-28, 1996.

11. Jackson, D. W.; Corsetti, J.; and Simon, T. M.: Biologic incorporation of allograft

anterior cruciate ligament replacements. Clin Orthop Relat Res, (324): 126-33, 1996.

12. Frank, C. B., and Jackson, D. W.: The science of reconstruction of the anterior

cruciate ligament. J Bone Joint Surg Am, 79(10): 1556-76, 1997.

13. Laurencin, C. T.; Ambrosio, A. M.; Borden, M. D.; and Cooper, J. A., Jr.: Tissue

engineering: orthopedic applications. Annu Rev Biomed Eng, 1: 19-46, 1999.

14. Gottlob, C. A.; Baker, C. L., Jr.; Pellissier, J. M.; and Colvin, L.: Cost effectiveness

of anterior cruciate ligament reconstruction in young adults. Clin Orthop Relat Res, (367):

272-82, 1999.

15. Lin, V. S.; Lee, M. C.; O'Neal, S.; McKean, J.; and Sung, K. L.: Ligament tissue

engineering using synthetic biodegradable fiber scaffolds. Tissue Eng, 5(5): 443-52, 1999.

16. Butler, D. L., and Awad, H. A.: Perspectives on cell and collagen composites for

tendon repair. Clin Orthop Relat Res, (367 Suppl): S324-32, 1999.

174

17. Buckwalter, J. A., and Grodzinsky, A. J.: Loading of healing bone, fibrous tissue,

and muscle: implications for orthopaedic practice. J Am Acad Orthop Surg, 7(5): 291-9,

1999.

18. Butler, D. L.; Goldstein, S. A.; and Guilak, F.: Functional tissue engineering: the

role of biomechanics. J Biomech Eng, 122(6): 570-5, 2000.

19. Galatz, L. M.; Griggs, S.; Cameron, B. D.; and Iannotti, J. P.: Prospective

longitudinal analysis of postoperative shoulder function : a ten-year follow-up study of

full-thickness rotator cuff tears. J Bone Joint Surg Am, 83-A(7): 1052-6, 2001.

20. Galili, U.: The alpha-Gal epitope (Galalpha1-3Galbeta1-4GlcNAc-R) in

xenotransplantation. Biochimie, 83(7): 557-63, 2001.

21. Badylak, S. F.; Park, K.; Peppas, N.; McCabe, G.; and Yoder, M.: Marrow-derived

cells populate scaffolds composed of xenogeneic extracellular matrix. Exp Hematol,

29(11): 1310-8, 2001.

22. Altman, G. H.; Horan, R. L.; Martin, I.; Farhadi, J.; Stark, P. R.; Volloch, V.;

Richmond, J. C.; Vunjak-Novakovic, G.; and Kaplan, D. L.: Cell differentiation by

mechanical stress. FASEB J, 16(2): 270-2, 2002.

23. Min, B. H.; Han, M. S.; Woo, J. I.; Park, H. J.; and Park, S. R.: The origin of cells

that repopulate patellar tendons used for reconstructing anterior cruciate ligaments in man.

J Bone Joint Surg Br, 85(5): 753-7, 2003.

24. Vangsness, C. T., Jr.; Garcia, I. A.; Mills, C. R.; Kainer, M. A.; Roberts, M. R.; and

Moore, T. M.: Allograft transplantation in the knee: tissue regulation, procurement,

processing, and sterilization. Am J Sports Med, 31(3): 474-81, 2003.

175

25. Chang, S. K.; Egami, D. K.; Shaieb, M. D.; Kan, D. M.; and Richardson, A. B.:

Anterior cruciate ligament reconstruction: allograft versus autograft. Arthroscopy, 19(5):

453-62, 2003.

26. Goutallier, D.; Postel, J. M.; Gleyze, P.; Leguilloux, P.; and Van Driessche, S.:

Influence of cuff muscle fatty degeneration on anatomic and functional outcomes after

simple suture of full-thickness tears. J Shoulder Elbow Surg, 12(6): 550-4, 2003.

27. Garvin, J.; Qi, J.; Maloney, M.; and Banes, A. J.: Novel system for engineering

bioartificial tendons and application of mechanical load. Tissue Eng, 9(5): 967-79, 2003.

28. Vunjak-Novakovic, G.; Altman, G.; Horan, R.; and Kaplan, D. L.: Tissue

engineering of ligaments. Annu Rev Biomed Eng, 6: 131-56, 2004.

29. Galatz, L. M.; Ball, C. M.; Teefey, S. A.; Middleton, W. D.; and Yamaguchi, K.:

The outcome and repair integrity of completely arthroscopically repaired large and massive

rotator cuff tears. J Bone Joint Surg Am, 86-A(2): 219-24, 2004.

30. Sherman, O. H., and Banffy, M. B.: Anterior cruciate ligament reconstruction:

which graft is best? Arthroscopy, 20(9): 974-80, 2004.

31. Laurencin, C. T., and Freeman, J. W.: Ligament tissue engineering: an evolutionary

materials science approach. Biomaterials, 26(36): 7530-6, 2005.

32. Poehling, G. G.; Curl, W. W.; Lee, C. A.; Ginn, T. A.; Rushing, J. T.; Naughton,

M. J.; Holden, M. B.; Martin, D. F.; and Smith, B. P.: Analysis of outcomes of anterior

cruciate ligament repair with 5-year follow-up: allograft versus autograft. Arthroscopy,

21(7): 774-85, 2005.

176

33. Webb, K.; Hitchcock, R. W.; Smeal, R. M.; Li, W.; Gray, S. D.; and Tresco, P. A.:

Cyclic strain increases fibroblast proliferation, matrix accumulation, and elastic modulus

of fibroblast-seeded polyurethane constructs. J Biomech, 39(6): 1136-44, 2006.

34. Musculoskeletal Transplant Foundation, M. T. F.: MTF Allograft Tissue

Instructions for Use. 2006.

35. Hoffmann, A., and Gross, G.: Tendon and ligament engineering: from cell biology

to in vivo application. Regen Med, 1(4): 563-74, 2006.

36. Gilbert, T. W.; Sellaro, T. L.; and Badylak, S. F.: Decellularization of tissues and

organs. Biomaterials, 27(19): 3675-83, 2006.

37. Laurencin, C. T.; Khan, Y.; Kofron, M.; El-Amin, S.; Botchwey, E.; Yu, X.; and

Cooper, J. A., Jr.: The ABJS Nicolas Andry Award: Tissue engineering of bone and

ligament: a 15-year perspective. Clin Orthop Relat Res, 447: 221-36, 2006.

38. Iannotti, J. P.; Codsi, M. J.; Kwon, Y. W.; Derwin, K.; Ciccone, J.; and Brems, J.

J.: Porcine small intestine submucosa augmentation of surgical repair of chronic two-

tendon rotator cuff tears. A randomized, controlled trial. J Bone Joint Surg Am, 88(6):

1238-44, 2006.

39. Gentleman, E.; Livesay, G. A.; Dee, K. C.; and Nauman, E. A.: Development of

ligament-like structural organization and properties in cell-seeded collagen scaffolds in

vitro. Ann Biomed Eng, 34(5): 726-36, 2006.

40. Gimbel, J. A.; Van Kleunen, J. P.; Lake, S. P.; Williams, G. R.; and Soslowsky, L.

J.: The role of repair tension on tendon to bone healing in an animal model of chronic

rotator cuff tears. J Biomech, 40(3): 561-8, 2007.

177

41. Stone, K. R.; Walgenbach, A. W.; Turek, T. J.; Somers, D. L.; Wicomb, W.; and

Galili, U.: Anterior cruciate ligament reconstruction with a porcine xenograft: a serologic,

histologic, and biomechanical study in primates. Arthroscopy, 23(4): 411-9, 2007.

42. Stone, K. R.; Abdel-Motal, U. M.; Walgenbach, A. W.; Turek, T. J.; and Galili, U.:

Replacement of human anterior cruciate ligaments with pig ligaments: a model for anti-

non-gal antibody response in long-term xenotransplantation. Transplantation, 83(2): 211-

9, 2007.

43. Whitlock, P. W.; Smith, T. L.; Poehling, G. G.; Shilt, J. S.; and Van Dyke, M.: A

naturally derived, cytocompatible, and architecturally optimized scaffold for tendon and

ligament regeneration. Biomaterials, 28(29): 4321-9, 2007.

44. Cancedda, R.; Giannoni, P.; and Mastrogiacomo, M.: A tissue engineering

approach to bone repair in large animal models and in clinical practice. Biomaterials,

28(29): 4240-50, 2007.

45. Whitlock, P. W.; Smith, T. L.; Shilt, J. S.; Van Dyke, M. E.; and Poehling, G. G.:

A Novel Scaffold for Tendon and Ligament Regeneration Derived from Human Allograft

Tissue Combined Meeting of the International Orthopaedic Research Societies, Honolulu

HI, 2007.

46. Suarez, L. S., and Richmond, J. C.: Overview of procurement, processing, and

sterilization of soft tissue allografts for sports medicine. Sports Med Arthrosc, 15(3): 106-

13, 2007.

47. Dines, J. S.; Grande, D. A.; and Dines, D. M.: Tissue engineering and rotator cuff

tendon healing. J Shoulder Elbow Surg, 16(5 Suppl): S204-7, 2007.

178

48. Cole, B. J.; McCarty, L. P., 3rd; Kang, R. W.; Alford, W.; Lewis, P. B.; and Hayden,

J. K.: Arthroscopic rotator cuff repair: prospective functional outcome and repair integrity

at minimum 2-year follow-up. J Shoulder Elbow Surg, 16(5): 579-85, 2007.

49. Aurora, A.; McCarron, J.; Iannotti, J. P.; and Derwin, K.: Commercially available

extracellular matrix materials for rotator cuff repairs: state of the art and future trends. J

Shoulder Elbow Surg, 16(5 Suppl): S171-8, 2007.

50. Butler, D. L.; Juncosa-Melvin, N.; Boivin, G. P.; Galloway, M. T.; Shearn, J. T.;

Gooch, C.; and Awad, H.: Functional tissue engineering for tendon repair: A

multidisciplinary strategy using mesenchymal stem cells, bioscaffolds, and mechanical

stimulation. Journal of Orthopaedic Research, 26(1): 1-9, 2008.

51. Butler, D. L.; Juncosa-Melvin, N.; Boivin, G. P.; Galloway, M. T.; Shearn, J. T.;

Gooch, C.; and Awad, H.: Functional tissue engineering for tendon repair: A

multidisciplinary strategy using mesenchymal stem cells, bioscaffolds, and mechanical

stimulation. J Orthop Res, 26(1): 1-9, 2008.

52. Joyce, M. J.; Greenwald, A. S.; Boden, S.; Brubaker, S.; and Heim, C. S.:

Musculoskeletal Allograft Tissue Safety American Academy of Orthopaedic Surgeons

Committee on Biological Implants Tissue Work Group, San Francisco CA, 2008.

53. Whitlock, P. W.; Van Dyke, M.; Poehling, G. G.; Smith, T. L.; Marker, D. R.;

Koman, L. A.; and Seyler, T. M.: Functional Tissue Engineering of Tendons and Ligaments

American Academy of Orthopaedic Surgeons Annual Meeting, Las Vegas NV, 2009.

54. Oh, J. H.; Kim, S. H.; Ji, H. M.; Jo, K. H.; Bin, S. W.; and Gong, H. S.: Prognostic

factors affecting anatomic outcome of rotator cuff repair and correlation with functional

outcome. Arthroscopy, 25(1): 30-9, 2009.

179

55. Ide, J.; Kikukawa, K.; Hirose, J.; Iyama, K.; Sakamoto, H.; and Mizuta, H.:

Reconstruction of large rotator-cuff tears with acellular dermal matrix grafts in rats. J

Shoulder Elbow Surg, 18(2): 288-95, 2009.

56. Neri, B. R.; Chan, K. W.; and Kwon, Y. W.: Management of massive and

irreparable rotator cuff tears. J Shoulder Elbow Surg, 18(5): 808-18, 2009.

57. Cousineau-Pelletier, P., and Langelier, E.: Relative contributions of mechanical

degradation, enzymatic degradation, and repair of the extracellular matrix on the response

of tendons when subjected to under- and over- mechanical stimulations in vitro. J Orthop

Res, 28(2): 204-10, 2010.

58. Tischer, T.; Aryee, S.; Wexel, G.; Steinhauser, E.; Adamczyk, C.; Eichhorn, S.;

Milz, S.; Martinek, V.; Gansbacher, B.; Imhoff, A. B.; and Vogt, S.: Tissue engineering of

the anterior cruciate ligament-sodium dodecyl sulfate-acellularized and revitalized tendons

are inferior to native tendons. Tissue Eng Part A, 16(3): 1031-40, 2010.

59. Nagata, S.; Hanayama, R.; and Kawane, K.: Autoimmunity and the clearance of

dead cells. Cell, 140(5): 619-30, 2010.

60. AAOS: Bone and soft-tissue allografts processing and safety. AAOS Now, May

2011, 2011.

61. American Academy of Orthopaedic Surgeons. Information Statement - Use of

Musculoskeletal Tissue Allografts.

(http://www.aaos.org/about/papers/advistmt/1011.asp), 2011.

62. Crapo, P. M.; Gilbert, T. W.; and Badylak, S. F.: An overview of tissue and whole

organ decellularization processes. Biomaterials, 32(12): 3233-43, 2011.

180

63. Whitlock, P. W.; Seyler, T. M.; Northam, C.; Smith, T. L.; Van Dyke, M. E.;

Poehling, G. G.; and Koman, L. A.: The Effect of Cyclic Strain on the Tensile Properties

of a Naturally-Derived, Decellularized Tendon Scaffold Seeded with Allogeneic Tenocytes

and Associated mRNA Expression. J Surg Orthop Adv, Accepted for publication, 2012.

64. Rathbone, S.; Maffulli, N.; and Cartmell, S. H.: Most british surgeons would

consider using a tissue-engineered anterior cruciate ligament: a questionnaire study. Stem

Cells Int, 2012: 303724, 2012.

65. Schubert, T.; Bigare, E.; Van Isacker, T.; Gigi, J.; Delloye, C.; and Cornu, O.:

Analysis of predisposing factors for contamination of bone and tendon allografts. Cell

Tissue Bank, 13(3): 421-9, 2012.

66. Whitlock, P. W.; Seyler, T. M.; Parks, G. D.; Ornelles, D. A.; Smith, T. L.; Van

Dyke, M. E.; and Poehling, G. G.: A novel process for optimizing musculoskeletal allograft

tissue to improve safety, ultrastructural properties, and cell infiltration. J Bone Joint Surg

Am, 94(16): 1458-67, 2012.

67. Gilbert, T. W.: Strategies for tissue and organ decellularization. J Cell Biochem,

113(7): 2217-22, 2012.

68. Hinsenkamp, M.; Muylle, L.; Eastlund, T.; Fehily, D.; Noel, L.; and Strong, D. M.:

Adverse reactions and events related to musculoskeletal allografts: reviewed by the World

Health Organisation Project NOTIFY. Int Orthop, 36(3): 633-41, 2012.

69. Whitlock, P. W.; Seyler, T. M.; Northam, C. N.; Smith, T. L.; Poehling, G. G.;

Koman, L. A.; and Van Dyke, M. E.: Effect of cyclic strain on tensile properties of a

naturally derived, decellularized tendon scaffold seeded with allogeneic tenocytes and

associated messenger RNA expression. J Surg Orthop Adv, 22(3): 224-32, 2013.

181

70. Arenas-Herrera, J. E.; Ko, I. K.; Atala, A.; and Yoo, J. J.: Decellularization for

whole organ bioengineering. Biomed Mater, 8(1): 014106, 2013.

182

CURRICULUM VITAE

Thorsten M. Seyler, M.D.

EDUCATION

Wake Forest University, Graduate School of Arts and Sciences, Winston-Salem, NC Ph.D., Molecular Medicine and Translational Science Graduate Program: 2007 – 2012

Emory University School of Medicine, Atlanta, GA Visiting Student, International Exchange Scholarship Program, Ruprecht-Karls-University, School of Medicine, 2004

Mayo Medical and Graduate School of Medicine, Rochester MN Visiting Student, International Exchange Scholarship Program, Ruprecht-Karls-University, School of Medicine, 2002-2003

Ruprecht-Karls-University, School of Medicine, Heidelberg, Germany M.D., 2000 – 2005 Semmelweis University for Medical Science, Budapest, Hungary

Premedical Sciences, 1998 – 2000

183

POST GRADUATE EDUCATION & TRAINING

Wake Forest University, Department of Orthopaedic Surgery, Winston-Salem, NC Resident, Orthopaedic Surgery: July 2007 – present Program Director: Jason E. Lang, M.D. Wake Forest University, Department of General Surgery, Winston-Salem, NC

Resident (Intern), General Surgery: July 2009 – June 2010 Program Director: Jason E. Lang, M.D. Wake Forest University, Graduate School of Arts and Sciences, Winston-Salem,

NC Graduate Student (Ph.D.), Molecular Medicine and Translational Science Graduate Program: August 2007 – December 2012 Program Directors: Richard F. Loeser, Jr., M.D. & Kathleen B. Brosnihan, Ph.D.

Sinai Hospital of Baltimore, Rubin Institute for Advanced Orthopedics, Baltimore, MD Research Fellow, Center for Joint Preservation and Reconstruction: July 2005 – June 2007 Program Director: Michael A. Mont, M.D.

184

MEDICAL LICENSURES

Resident training license, State of North Carolina, 2007 (active) Full license, Federal Republic of Germany, 2005 (active)

185

PROFESSIONAL SOCIETY MEMBERSHIPS

Orthopaedic Research Society: Associate Member 007 - present Southern Orthopaedic Association: Resident Member

2007 - present American Academy of Orthopaedic Surgeons: Resident Member

2007 - present American Association of Hip and Knee Surgeons: Resident Member

2012 - present

186

HONORS & AWARDS

2003 Nominated for the ACR/ARHP Student Achievement Award at the 67th American College of Rheumatology Annual Scientific Meeting, Orlando, FL (Lead-author)

2006 Award for Best Resident/Fellow Clinical Research Paper, 65th Annual Meeting Maryland Orthopaedic Association, Baltimore, MD (Co-author)

2008 Award for Best Basic Science Research Paper, Resident Research Day, Department of Orthopaedic Surgery, Wake Forest University School of Medicine, Winston-Salem, NC (Lead-author)

2009 Award of Excellence for Scientific Exhibits, 76th Annual Meeting of the American Academy of Orthopaedic Surgeons, Las Vegas, NV (Co-author)

2012 Runner-up Best Clinical Science Resident Research Paper, 32nd Annual Oscar Miller Day Symposium, Charlotte, NC (Lead-author)

2012 Best Clinical Science Resident Research Paper, 32nd Annual Oscar Miller Day Symposium, Charlotte, NC (Co-author)

2012 Best Research Paper, 100th Annual Meeting of The Clinical Orthopaedic Society (COS), The Palmer House, Chicago, IL (Co-author)

2012 Best Research Paper, Annual Meeting North Carolina Orthopaedic Association, Pinehurst Resort, Village of Pinehurst, NC (Co-author)

187

EDITORIAL ACTIVITIES

Editorial Advisory Board Member, Open Bone Journal 2009 - present

Associate Editor, Bone & Joint Research 2012 - present

Consultant Reviewer, Clinical Orthopaedics and Related Research 2008 - present

Consultant Reviewer, Journal of Joint and Bone Surgery Am. Vol. 2006 - present

Consultant Reviewer, Journal of Arthroplasty 2006 - present

Consultant Reviewer, Musculoskeletal Disorders 2006 – present

Consultant Reviewer, Orthopaedic Knowledge Online Journal 2009 - present

Guest Editor, Proceedings of the 1st Annual United States Comprehensive Course on Total Hip Resurfacing Arthroplasty, Journal of Bone and Joint Surgery Am. Vol., Volume 90-A, Supplement 3, Pages 1-148, 2008

Guest Editor, Proceedings of the 2nd Annual United States Comprehensive Course on Total Hip Resurfacing Arthroplasty, Bulletin of the NYU Hospital for Joint Diseases, Volume 62 (2), Pages 101-192, 2009

Guest Editor, Symposium on Advanced Techniques for Rehabilitation after Total Hip and Knee Arthroplasty, Clinical Orthopaedics and Related Research, Volume 476 (6), Pages 1395-1500, 2009

188

COMMITTEES

Program Director, First Annual U.S. Comprehensive Course on Total Hip Resurfacing Arthroplasty, November 8-10, 2007, Annapolis, MD

Program Director, Second Annual U.S. Comprehensive Course on Total Hip Arthroplasty, October 24-25, 2008, Los Angeles, CA

Program Director, Third Annual U.S. Comprehensive Course on Total Hip Arthroplasty, September 4-5, 2009, Baltimore, MD

Program Director, Fourth Annual Hip Joint Course, Treatment for Young Patients with Hip Arthritis, August 27-29, 2010, Baltimore, MD

Program Director, Sixth Annual Joint Course, New Technology for the Treatment of Adult Hip and Knee Disorders, August 24-25, 2012, Baltimore, MD

Voting Delegate, International Consensus Meeting on Periprosthetic Joint Infection, August 1st, 2013, Philadelpia, PA

189

PEER-REVIEWED PUBLICATIONS

1. Marulanda, G. A.; Ragland, P. S.; Seyler, T. M.; and Mont, M. A.: Reductions in blood loss with use of a bipolar sealer for hemostasis in primary total knee arthroplasty. Surg Technol Int, 14: 281-6, 2005.

2. Seyler, T. M.; Bonutti, P. M.; Ragland, P. S.; Marulanda, G. A.; and Mont, M. A.: Minimally invasive lateral approach to total knee arthroplasty. Seminars in Arthroplasty, 16(3): 223-226, 2005.

3. Weyand, C. M.; Seyler, T. M.; and Goronzy, J. J.: B cells in rheumatoid synovitis. Arthritis Res Ther, 7 Suppl 3: S9-12, 2005.

4. Park, Y. W.; Pryshchep, S.; Seyler, T. M.; Goronzy, J. J.; and Weyand, C. M.: B cells as a therapeutic target in autoimmune diseases. Expert Opin Ther Targets, 9(3): 431-45, 2005.

5. Seyler, T. M.; Park, Y. W.; Takemura, S.; Bram, R. J.; Kurtin, P. J.; Goronzy, J. J.; and Weyand, C. M.: BLyS and APRIL in rheumatoid arthritis. J Clin Invest, 115(11): 3083-92, 2005.

6. Mont, M. A.; Bonutti, P. M.; Seyler, T. M.; Plate, J. F.; Delanois, R. E.; and Kester, M.: The Future of High Performance Total Knee Arthroplasty. Seminars in Arthroplasty, 17(2): 80-87, 2006.

7. Mont, M. A.; Bonutti, P. M.; Seyler, T. M.; Plate, J. F.; Delanois, R. E.; and Kester, M.: The Future of High Performance Total Hip Arthroplasty. Seminars in Arthroplasty, 17(2): 88-92, 2006.

8. Seyler, T. M.; Etienne, G.; Plate, J. F.; Fisher, P.; and Mont, M. A.: Use of modular large femoral heads without liners in hip arthroplasty. Surg Technol Int, 15: 217-20, 2006.

9. Seyler, T. M.; Marulanda, G. A.; Delanois, R. E.; and Mont, M. A.: Limited Approach Surface Replacement Total Hip Arthroplasty. Oper Tech Orthop, 16(2): 120-125, 2006.

10. Seyler, T. M.; Mont, M. A.; Ragland, P. S.; Kachwala, M. M.; and Delanois, R. E.: Sports activity after total hip and knee arthroplasty: specific recommendations concerning tennis. Sports Med, 36(7): 571-83, 2006.

11. Mont, M. A.; Ragland, P. S.; Etienne, G.; Seyler, T. M.; and Schmalzried, T. P.: Hip resurfacing arthroplasty. J Am Acad Orthop Surg, 14(8): 454-63, 2006.

12. Leadbetter, W. B.; Seyler, T. M.; Ragland, P. S.; and Mont, M. A.: Indications, contraindications, and pitfalls of patellofemoral arthroplasty. J Bone Joint Surg Am, 88 Suppl 4: 122-37, 2006.

13. Diaz-Borjon, A.; Seyler, T. M.; Chen, N. L.; and Lim, S. S.: Bisphosphonate-associated arthritis. J Clin Rheumatol, 12(3): 131-3, 2006.

14. Marulanda, G.; Seyler, T. M.; Sheikh, N. H.; and Mont, M. A.: Percutaneous drilling for the treatment of secondary osteonecrosis of the knee. J Bone Joint Surg Br, 88(6): 740-6, 2006.

15. Bonutti, P. M.; Seyler, T. M.; Kester, M.; McMahon, M.; and Mont, M. A.: Minimally invasive revision total knee arthroplasty. Clin Orthop Relat Res, 446: 69-75, 2006.

190

16. Mont, M. A.; Seyler, T. M.; Marulanda, G. A.; Delanois, R. E.; and Bhave, A.: Surgical treatment and customized rehabilitation for stiff knee arthroplasties. Clin Orthop Relat Res, 446: 193-200, 2006.

17. Bonutti, P. M.; Seyler, T. M.; Delanois, R. E.; McMahon, M.; McCarthy, J. C.; and Mont, M. A.: Osteonecrosis of the knee after laser or radiofrequency-assisted arthroscopy: treatment with minimally invasive knee arthroplasty. J Bone Joint Surg Am, 88 Suppl 3: 69-75, 2006.

18. Mont, M. A.; Seyler, T. M.; Marker, D. R.; Marulanda, G. A.; and Delanois, R. E.: Use of metal-on-metal total hip resurfacing for the treatment of osteonecrosis of the femoral head. J Bone Joint Surg Am, 88 Suppl 3: 90-7, 2006.

19. Mont, M. A.; Seyler, T. M.; Plate, J. F.; Delanois, R. E.; and Parvizi, J.: Uncemented total hip arthroplasty in young adults with osteonecrosis of the femoral head: a comparative study. J Bone Joint Surg Am, 88 Suppl 3: 104-9, 2006.

20. Seyler, T. M.; Bonutti, P. M.; Shen, J.; Naughton, M.; and Kester, M.: Use of an alumina-on-alumina bearing system in total hip arthroplasty for osteonecrosis of the hip. J Bone Joint Surg Am, 88 Suppl 3: 116-25, 2006.

21. Mont, M. A.; Marulanda, G. A.; Seyler, T. M.; Plate, J. F.; and Delanois, R. E.: Core decompression and nonvascularized bone grafting for the treatment of early stage osteonecrosis of the femoral head. Instr Course Lect, 56: 213-20, 2007.

22. Akbar, M.; Seyler, T. M.; Abel, R.; and Gerner, H. J.: [Heterotope Ossifikation bei Querschnittlähmung und Schädel-Hirn-Trauma]. Phys Rehab Kur Med, 17(3): 156-171, 2007.

23. Seyler, T. M.; Cui, Q.; Mihalko, W. M.; Mont, M. A.; and Saleh, K. J.: Advances in hip arthroplasty in the treatment of osteonecrosis. Instr Course Lect, 56: 221-33, 2007.

24. Mont, M. A.; Jones, L. C.; Seyler, T. M.; Marulanda, G. A.; Saleh, K. J.; and Delanois, R. E.: New treatment approaches for osteonecrosis of the femoral head: an overview. Instr Course Lect, 56: 197-212, 2007.

25. Childress III, L.; Ulrich, S. D.; Seyler, T. M.; Delanois, R. E.; Marker, D. R.; and Mont, M. A.: Role of Metal-on-Metal Resurfacing in the Treatment of Late Stage Osteonecrosis. Seminars in Arthroplasty, 18(3): 216-219, 2007.

26. Diaz Jr, L. A.; Foss, C. A.; Thornton, K.; Nimmagadda, S.; Endres, C. J.; Uzuner, O.; Seyler, T. M.; Ulrich, S. D.; Conway, J.; Bettegowda, C.; Agrawal, N.; Cheong, I.; Zhang, X.; Ladenson, P. W.; Vogelstein, B. N.; Mont, M. A.; Zhou, S.; Kinzler, K. W.; Vogelstein, B.; and Pomper, M. G.: Imaging of musculoskeletal bacterial infections by [124I]FIAU-PET/CT. PLoS ONE, 2(10), 2007.

27. Delanois, R. E.; Seyler, T. M.; Essner, A.; Schmidig, G.; and Mont, M. A.: Cementation of a polyethylene liner into a metal shell. J Arthroplasty, 22(5): 732-7, 2007.

28. Boyd, H. S.; Ulrich, S. D.; Seyler, T. M.; Marulanda, G. A.; and Mont, M. A.: Resurfacing for Perthes disease: an alternative to standard hip arthroplasty. Clin Orthop Relat Res, 465: 80-5, 2007.

29. Mont, M. A.; Seyler, T. M.; Ulrich, S. D.; Beaule, P. E.; Boyd, H. S.; Grecula, M. J.; Goldberg, V. M.; Kennedy, W. R.; Marker, D. R.; Schmalzried, T. P.; Sparling, E. A.; Vail, T. P.; and Amstutz, H. C.: Effect of changing indications and techniques on total hip resurfacing. Clin Orthop Relat Res, 465: 63-70, 2007.

191

30. Mont, M. A.; Seyler, T. M.; Ragland, P. S.; Starr, R.; Erhart, J.; and Bhave, A.: Gait analysis of patients with resurfacing hip arthroplasty compared with hip osteoarthritis and standard total hip arthroplasty. J Arthroplasty, 22(1): 100-8, 2007.

31. Mont, M. A.; Marker, D. R.; Seyler, T. M.; Gordon, N.; Hungerford, D. S.; and Jones, L. C.: Knee arthroplasties have similar results in high- and low-activity patients. Clin Orthop Relat Res, 460: 165-73, 2007.

32. Ulrich, S. D.; Mont, M. A.; Bonutti, P. M.; Seyler, T. M.; Marker, D. R.; and Jones, L. C.: Scientific evidence supporting computer-assisted surgery and minimally invasive surgery for total knee arthroplasty. Expert Rev Med Devices, 4(4): 497-505, 2007.

33. Mont, M. A.; Ulrich, S. D.; and Seyler, T. M.: Role of thrombotic and fibrinolytic alterations in the pathogenesis and treatment of osteonecrosis. J Rheumatol, 34(3): 466-8, 2007.

34. Ulrich, S. D.; Bhave, A.; Marker, D. R.; Seyler, T. M.; and Mont, M. A.: Focused rehabilitation treatment of poorly functioning total knee arthroplasties. Clin Orthop Relat Res, 464: 138-45, 2007.

35. Ulrich, S. D.; Bonutti, P. M.; Seyler, T. M.; Marker, D. R.; Jones, L. C.; and Mont, M. A.: Outcomes-based evaluations supporting computer-assisted surgery and minimally invasive surgery for total hip arthroplasty. Expert Rev Med Devices, 4(6): 873-83, 2007.

36. Seyler, T. M.; Bonutti, P. M.; Ulrich, S. D.; Fatscher, T.; Marker, D. R.; and Mont, M. A.: Minimally invasive lateral approach to total knee arthroplasty. J Arthroplasty, 22(7 Suppl 3): 21-6, 2007.

37. Marker, D. R.; Seyler, T. M.; Jinnah, R. H.; Delanois, R. E.; Ulrich, S. D.; and Mont, M. A.: Femoral neck fractures after metal-on-metal total hip resurfacing: a prospective cohort study. J Arthroplasty, 22(7 Suppl 3): 66-71, 2007.

38. Seyler, T. M.; Marker, D. R.; Bhave, A.; Plate, J. F.; Marulanda, G. A.; Bonutti, P. M.; Delanois, R. E.; and Mont, M. A.: Functional problems and arthrofibrosis following total knee arthroplasty. J Bone Joint Surg Am, 89 Suppl 3: 59-69, 2007.

39. Akbar, M.; Abel, R.; Seyler, T. M.; Bedke, J.; Haferkamp, A.; Gerner, H. J.; and Mohring, K.: Repeated botulinum-A toxin injections in the treatment of myelodysplastic children and patients with spinal cord injuries with neurogenic bladder dysfunction. BJU Int, 100(3): 639-45, 2007.

40. Bhave, A.; Marker, D. R.; Seyler, T. M.; Ulrich, S. D.; Plate, J. F.; and Mont, M. A.: Functional problems and treatment solutions after total hip arthroplasty. J Arthroplasty, 22(6 Suppl 2): 116-24, 2007.

41. Kolisek, F. R.; Seyler, T. M.; Ulrich, S. D.; Marker, D. R.; Jessup, N. M.; and Mont, M. A.: A Comparison of the Minimally Invasive Dual-Incision versus Posterolateral Approach in Total Hip Arthroplasty. Surg Technol Int, 17: 253-8, 2008.

42. Mont, M. A.; Ulrich, S. D.; Seyler, T. M.; Smith, J. M.; Marker, D. R.; McGrath, M. S.; Hungerford, D. S.; and Jones, L. C.: Bone scanning of limited value for diagnosis of symptomatic oligofocal and multifocal osteonecrosis. J Rheumatol, 35(8): 1629-34, 2008.

43. Seyler, T. M.; Lai, L. P.; Sprinkle, D. I.; Ward, W. G.; and Jinnah, R. H.: Does computer-assisted surgery improve accuracy and decrease the learning curve in hip resurfacing? A radiographic analysis. J Bone Joint Surg Am, 90 Suppl 3: 71-80, 2008.

192

44. Bonutti, P. M.; McGrath, M. S.; Ulrich, S. D.; McKenzie, S. A.; Seyler, T. M.; and Mont, M. A.: Static progressive stretch for the treatment of knee stiffness. Knee, 15(4): 272-6, 2008.

45. Mont, M. A.; McGrath, M. S.; Ulrich, S. D.; Seyler, T. M.; Marker, D. R.; and Delanois, R. E.: Metal-on-metal total hip resurfacing arthroplasty in the presence of extra-articular deformities or implants. J Bone Joint Surg Am, 90 Suppl 3: 45-51, 2008.

46. McGrath, M. S.; Desser, D. R.; Ulrich, S. D.; Seyler, T. M.; Marker, D. R.; and Mont, M. A.: Total hip resurfacing in patients who are sixty years of age or older. J Bone Joint Surg Am, 90 Suppl 3: 27-31, 2008.

47. Robinson, Y.; Heyde, C. E.; Tschoke, S. K.; Mont, M. A.; Seyler, T. M.; and Ulrich, S. D.: Evidence supporting the use of bone morphogenetic proteins for spinal fusion surgery. Expert Rev Med Devices, 5(1): 75-84, 2008.

48. Delanois, R. E.; McGrath, M. S.; Ulrich, S. D.; Marker, D. R.; Seyler, T. M.; Bonutti, P. M.; and Mont, M. A.: Results of total knee replacement for isolated patellofemoral arthritis: when not to perform a patellofemoral arthroplasty. Orthop Clin North Am, 39(3): 381-8, vii, 2008.

49. Akbar, M.; Mont, M. A.; Heisel, C.; Marker, D. R.; Ulrich, S. D.; and Seyler, T. M.: [Resurfacing for osteonecrosis of the femoral head]. Orthopade, 37(7): 672-8, 2008.

50. Bonutti, P. M.; Seyler, T. M.; Bianco, P. D.; Ulrich, S. D.; and Mont, M. A.: Inventing in orthopaedics: from idea to marketed device. J Bone Joint Surg Am, 90(6): 1385-92, 2008.

51. Marulanda, G. A.; Ulrich, S. D.; Seyler, T. M.; Delanois, R. E.; and Mont, M. A.: Reductions in blood loss with a bipolar sealer in total hip arthroplasty. Expert Rev Med Devices, 5(2): 125-31, 2008.

52. Stiehl, J. B.; Ulrich, S. D.; Seyler, T. M.; Bonutti, P. M.; Marker, D. R.; and Mont, M. A.: Bone morphogenetic proteins in total hip arthroplasty, osteonecrosis and trauma surgery. Expert Rev Med Devices, 5(2): 231-8, 2008.

53. Marker, D. R.; Seyler, T. M.; Ulrich, S. D.; Srivastava, S.; and Mont, M. A.: Do modern techniques improve core decompression outcomes for hip osteonecrosis? Clin Orthop Relat Res, 466(5): 1093-103, 2008.

54. Seyler, T. M.; Marker, D. R.; Ulrich, S. D.; Fatscher, T.; and Mont, M. A.: Nonvascularized bone grafting defers joint arthroplasty in hip osteonecrosis. Clin Orthop Relat Res, 466(5): 1125-32, 2008.

55. Bonutti, P. M.; Dethmers, D.; Ulrich, S. D.; Seyler, T. M.; and Mont, M. A.: Computer navigation-assisted versus minimally invasive TKA: benefits and drawbacks. Clin Orthop Relat Res, 466(11): 2756-62, 2008.

56. Marker, D. R.; Seyler, T. M.; McGrath, M. S.; Delanois, R. E.; Ulrich, S. D.; and Mont, M. A.: Treatment of early stage osteonecrosis of the femoral head. J Bone Joint Surg Am, 90 Suppl 4: 175-87, 2008.

57. McGrath, M. S.; Ulrich, S. D.; Bonutti, P. M.; Smith, J. M.; Seyler, T. M.; and Mont, M. A.: Evaluation of static progressive stretch for the treatment of wrist stiffness. J Hand Surg [Am], 33(9): 1498-504, 2008.

58. Seyler, T. M.; Smith, B. P.; Marker, D. R.; Ma, J.; Shen, J.; Smith, T. L.; Mont, M. A.; Kolaski, K.; and Koman, L. A.: Botulinum neurotoxin as a therapeutic modality

193

in orthopaedic surgery: more than twenty years of experience. J Bone Joint Surg Am, 90 Suppl 4: 133-45, 2008.

59. Sikes, C. V.; Lai, L. P.; Schreiber, M.; Mont, M. A.; Jinnah, R. H.; and Seyler, T. M.: Instability after total hip arthroplasty: treatment with large femoral heads vs constrained liners. J Arthroplasty, 23(7 Suppl): 59-63, 2008.

60. Ulrich, S. D.; Seyler, T. M.; Bennett, D.; Delanois, R. E.; Saleh, K. J.; Thongtrangan, I.; Kuskowski, M.; Cheng, E. Y.; Sharkey, P. F.; Parvizi, J.; Stiehl, J. B.; and Mont, M. A.: Total hip arthroplasties: what are the reasons for revision? Int Orthop, 32(5): 597-604, 2008.

61. Mont, M. A.; Marker, D. R.; Seyler, T. M.; Jones, L. C.; Kolisek, F. R.; and Hungerford, D. S.: High-impact sports after total knee arthroplasty. J Arthroplasty, 23(6 Suppl 1): 80-4, 2008.

62. Seyler, T. M.; Jinnah, R. H.; Koman, L. A.; Marker, D. R.; Mont, M. A.; Ulrich, S. D.; and Bhave, A.: Botulinum toxin type A injections for the management of flexion contractures following total knee arthroplasty. J Surg Orthop Adv, 17(4): 231-8, 2008.

63. Mont, M. A.; Schmalzried, T. P.; Zywiel, M. G.; McGrath, M. S.; and Seyler, T. M.: Perceptions concerning hip resurfacing from attendees at the Second Annual U.S. Comprehensive Course on Total Hip Resurfacing Arthroplasty. Bull NYU Hosp Jt Dis, 67(2): 102-7, 2009.

64. Bhave, A.; Zywiel, M. G.; Ulrich, S. D.; McGrath, M. S.; Seyler, T. M.; Marker, D. R.; Delanois, R. E.; and Mont, M. A.: Botulinum toxin type A injections for the management of muscle tightness following total hip arthroplasty: a case series. J Orthop Surg Res, 4: 34, 2009.

65. Marker, D. R.; Mont, M. A.; Seyler, T. M.; McGrath, M. S.; Kolisek, F. R.; and Bonutti, P. M.: Does functional improvement following TKA correlate to increased sports activity? Iowa Orthop J, 29: 11-6, 2009.

66. Shields, J. S.; Seyler, T. M.; Maguire, C.; and Jinnah, R. H.: Computer-assisted navigation in hip resurfacing arthroplasty - a single-surgeon experience. Bull NYU Hosp Jt Dis, 67(2): 164-7, 2009.

67. Kolisek, F. R.; McGrath, M. S.; Marker, D. R.; Jessup, N.; Seyler, T. M.; Mont, M. A.; and Lowry Barnes, C.: Posterior-stabilized versus posterior cruciate ligament-retaining total knee arthroplasty. Iowa Orthop J, 29: 23-7, 2009.

68. Seyler, T. M.; Mont, M. A.; Lai, L. P.; Xie, J.; Marker, D. R.; Zywiel, M. G.; and Bonutti, P. M.: Mid-term results and factors affecting outcome of a metal-backed unicompartmental knee design: a case series. J Orthop Surg Res, 4: 39, 2009.

69. Maguire, C. M.; Seyler, T. M.; Boyd, H. S.; Lai, L. P.; Delanois, R. E.; and Jinnah, R. H.: Hip resurfacing-keys to success. Bull NYU Hosp Jt Dis, 67(2): 142-5, 2009.

70. Strassmair, M.; Mont, M. A.; Seyler, T. M.; Bosebeck, H.; Marker, D. R.; and Laporte, D. M.: The use of a type-I lyophilisate collagen as an osteoinductive factor in pseudarthroses of the forearm. Surg Technol Int, 18: 213-8, 2009.

71. Zywiel, M. G.; McGrath, M. S.; Seyler, T. M.; Marker, D. R.; Bonutti, P. M.; and Mont, M. A.: Osteonecrosis of the knee: a review of three disorders. Orthop Clin North Am, 40(2): 193-211, 2009.

194

72. Marker, D. R.; Mont, M. A.; Seyler, T. M.; Laporte, D. M.; and Frassica, F. J.: Current literature: an educational tool to study osteonecrosis for the orthopaedic in-training examination? Orthop Clin North Am, 40(2): 299-304, 2009.

73. Bonutti, P. M.; Zywiel, M. G.; Seyler, T. M.; Lee, S. Y.; McGrath, M. S.; Marker, D. R.; and Mont, M. A.: Minimally invasive total knee arthroplasty using the contralateral knee as a control group: a case-control study. Int Orthop, 2009.

74. Marulanda, G. A.; Krebs, V. E.; Bierbaum, B. E.; Goldberg, V. M.; Ries, M.; Ulrich, S. D.; Seyler, T. M.; and Mont, M. A.: Hemostasis using a bipolar sealer in primary unilateral total knee arthroplasty. Am J Orthop (Belle Mead NJ), 38(12): E179-83, 2009.

75. Marulanda, G. A.; Minniti, C. P.; Ulrich, S. D.; Seyler, T. M.; and Mont, M. A.: Perioperative management for orthopaedic patients with sickle cell anaemia. J Orthop Surg (Hong Kong), 17(3): 346-50, 2009.

76. Kolisek, F. R.; Mont, M. A.; Seyler, T. M.; Marker, D. R.; Jessup, N. M.; Siddiqui, J. A.; Monesmith, E.; and Ulrich, S. D.: Total knee arthroplasty using cementless keels and cemented tibial trays: 10-year results. Int Orthop, 33(1): 117-21, 2009.

77. Saenz, C. L.; McGrath, M. S.; Marker, D. R.; Seyler, T. M.; Mont, M. A.; and Bonutti, P. M.: Early failure of a unicompartmental knee arthroplasty design with an all-polyethylene tibial component. Knee, 17(1): 53-6, 2009.

78. McGrath, M. S.; Suda, A. J.; Bonutti, P. M.; Zywiel, M. G.; Marker, D. R.; Seyler, T. M.; and Mont, M. A.: Techniques for managing anatomic variations in primary total knee arthroplasty. Expert Rev Med Devices, 6(1): 75-93, 2009.

79. McGrath, M. S.; Marker, D. R.; Seyler, T. M.; Ulrich, S. D.; and Mont, M. A.: Surface replacement is comparable to primary total hip arthroplasty. Clin Orthop Relat Res, 467(1): 94-100, 2009.

80. Harreld, K. L.; Marulanda, G. A.; Ulrich, S. D.; Marker, D. R.; Seyler, T. M.; and Mont, M. A.: Small-diameter percutaneous decompression for osteonecrosis of the shoulder. Am J Orthop (Belle Mead NJ), 38(7): 348-54, 2009.

81. Mont, M. A., and Seyler, T. M.: Advanced techniques for rehabilitation after total hip and knee arthroplasty. Clin Orthop Relat Res, 467(6): 1395, 2009.

82. Mont, M. A.; McGrath, M. S.; Bonutti, P. M.; Ulrich, S. D.; Marker, D. R.; Seyler, T. M.; and Suda, A. J.: Anatomic and physiologic rationale for various technologies for primary total hip arthroplasty. Expert Rev Med Devices, 6(2): 169-86, 2009.

83. Ulrich, S. D.; Bonutti, P. M.; Seyler, T. M.; Marker, D. R.; Morrey, B. F.; and Mont, M. A.: Restoring range of motion via stress relaxation and static progressive stretch in posttraumatic elbow contractures. J Shoulder Elbow Surg, 19(2): 196-201, 2009.

84. Akbar, M.; Bresch, B.; Seyler, T. M.; Wenz, W.; Bruckner, T.; Abel, R.; and Carstens, C.: Management of orthopaedic sequelae of congenital spinal disorders. J Bone Joint Surg Am, 91 Suppl 6: 87-100, 2009.

85. Marker, D. R.; LaPorte, D. M.; Seyler, T. M.; Ulrich, S. D.; McGrath, M. S.; Frassica, F. J.; and Mont, M. A.: Orthopaedic journal publications and their role in the preparation for the orthopaedic in-training examination. J Bone Joint Surg Am, 91 Suppl 6: 59-66, 2009.

86. Seyler, T. M.; Marker, D. R.; Boyd, H. S.; Zywiel, M. G.; McGrath, M. S.; and Mont, M. A.: Preoperative evaluation to determine candidates for metal-on-metal hip resurfacing. J Bone Joint Surg Am, 91 Suppl 6: 32-41, 2009.

195

87. Marker, D. R.; Seyler, T. M.; Bhave, A.; Zywiel, M. G.; and Mont, M. A.: Does commitment to rehabilitation influence clinical outcome of total hip resurfacing arthroplasty? J Orthop Surg Res, 5: 20, 2010.

88. Marker, D. R.; Zywiel, M. G.; Johnson, A. J.; Seyler, T. M.; and Mont, M. A.: Are component positioning and prosthesis size associated with hip resurfacing failure? BMC Musculoskelet Disord, 11: 227, 2010.

89. Akbar, M.; Almatrod, S.; Furstenberg, C. H.; Hemmer, S.; Kretzer, J. P.; Abel, R.; Seyler, T. M.; Bruckner, T.; Carstens, C.; and Wiedenhofer, B.: [Kyphectomy in myelomeningocele patients. Longterm results, complications and risk analysis]. Orthopade, 39(8): 792-800, 2010.

90. Clohisy, J. C.; Oryhon, J. M.; Seyler, T. M.; Wells, C. W.; Liu, S. S.; Callaghan, J. J.; and Mont, M. A.: Function and fixation of total hip arthroplasty in patients 25 years of age or younger. Clin Orthop Relat Res, 468(12): 3207-13, 2010.

91. Akbar, M.; Balean, G.; Brunner, M.; Seyler, T. M.; Bruckner, T.; Munzinger, J.; Grieser, T.; Gerner, H. J.; and Loew, M.: Prevalence of rotator cuff tear in paraplegic patients compared with controls. J Bone Joint Surg Am, 92(1): 23-30, 2010.

92. Marulanda, G. A.; McGrath, M. S.; Ulrich, S. D.; Seyler, T. M.; Delanois, R. E.; and Mont, M. A.: Percutaneous drilling for the treatment of atraumatic osteonecrosis of the ankle. J Foot Ankle Surg, 49(1): 20-4, 2010.

93. Bonutti, P. M.; Zywiel, M. G.; Ulrich, S. D.; Stroh, D. A.; Seyler, T. M.; and Mont, M. A.: A comparison of subvastus and midvastus approaches in minimally invasive total knee arthroplasty. J Bone Joint Surg Am, 92(3): 575-82, 2010.

94. LaPorte, D. M.; Marker, D. R.; Seyler, T. M.; Mont, M. A.; and Frassica, F. J.: Educational resources for the Orthopedic In-Training Examination. J Surg Educ, 67(3): 135-8, 2010.

95. Akbar, M.; Bresch, B.; Raiss, P.; Furstenberg, C. H.; Bruckner, T.; Seyler, T. M.; Carstens, C.; and Abel, R.: Fractures in myelomeningocele. J Orthop Traumatol, 11(3): 175-82, 2010.

96. Maguire, C. M.; Seyler, T. M.; Jinnah, R. H.; and Ward, W. G.: Hip resurfacing with retention of existing hardware - case report. Bull NYU Hosp Jt Dis, 69 Suppl 1: S98-102, 2011.

97. Seyler, T. M.; Johnson, A. J.; Marker, D. R.; Mont, M. A.; and Bonutti, P. M.: Arthroscopic-assisted minimally invasive total knee arthroplasty. Arthroscopy, 27(2): 290-3, 2011.

98. Lang, J. E.; Mannava, S.; Floyd, A. J.; Goddard, M. S.; Smith, B. P.; Mofidi, A.; Seyler, T. M.; and Jinnah, R. H.: Robotic systems in orthopaedic surgery. J Bone Joint Surg Br, 93(10): 1296-9, 2011.

99. Mannava, S.; Plate, J. F.; Whitlock, P. W.; Callahan, M. F.; Seyler, T. M.; Koman, L. A.; Smith, T. L.; and Tuohy, C. J.: Evaluation of in vivo rotator cuff muscle function after acute and chronic detachment of the supraspinatus tendon: an experimental study in an animal model. J Bone Joint Surg Am, 93(18): 1702-11, 2011.

100. Bracey, D. N.; Seyler, T. M.; Shields, J. S.; Leng, X.; Jinnah, R. H.; and Lang, J. E.: A Comparison of Acetate and Digital Templating for Hip Resurfacing. Am J Orth, Under Review, 2012.

196

101. Plate, J. F.; Seyler, T. M.; Halvorson, J. J.; Santago, A. C.; and Lang, J. E.: Non-Anatomic Closure of a Standard Parapatellar Knee Arthrotomy Leads to Patella Mal-Tracking and Decreased Range of Motion. Knee Surg Sports Traumatol Arthrosc, 2013 Jan 19. [Epub ahead of print]

102. Plate, J. F.; Seyler, T. M.; Stroh, A.; Akbar, M.; and Mont, M. A.: Risk of dislocation using large- vs. small-diameter femoral heads in total hip arthroplasty BMC Res Notes. Oct 5:553, 2012.

103. Schweppe, M. L.; Seyler, T. M.; Plate, J. F.; Swenson, R. D.; and Lang, J. E.: Does Surgical Approach in Total Hip Arthroplasty Affect Rehabilitation, Discharge Disposition, and Readmission Rate? Surg Technol Int, XXIII, 2013.

104. Seyler, T. M.; Whitlock, P. W.; Northam, C. N.; Smith, T. L.; Poehling, G. G.; Koman, L. A.; and Van Dyke, M. E.: The Effect of Cyclic Strain on the Tensile Properties of a Naturally-Derived, Decellularized Tendon Scaffold Seeded with Allogeneic Tenocytes and Associated mRNA Expression. J Surg Orthop Adv, 22,3:224-232, 2013.

105. Whitlock, P. W.; Seyler, T. M.; Parks, G. D.; Ornelles, D. A.; Smith, T. L.; Van Dyke, M. E.; and Poehling, G. G.: A Novel Process for Optimizing Musculoskeletal Allograft Tissue to Improve Safety, Ultrastructural Properties, and Cell Infiltration. J Bone Joint Surg Am, 94,16:1458-1467, 2012.

106. Mannava, S.; Plate, J. F.; Tuohy, C. J.; Seyler, T. M.; Whitlock, P. W.; Curl, W. W.; Smith, T. L.; and Saul, K. R.: The science of rotator cuff tears: translating animal models to clinical recommendations using simulation analysis. Knee Surg Sports Traumatol Arthrosc, 21,7:1610-1619, 2013.

107. Plate, J. F.; Mofidi, A.; Mannava, S.; Lorentzen, C. M.; Smith, B. P.; Seyler, T. M.; McTighe, T.; and Jinnah, R. H.: Unicompartmental Knee Arthroplasty: Past, Present, Future. Reconstructive Review, 2(2): 52-62, 2012.

108. Seyler, T. M.; Lorentzen, C. M.; and Jinnah, R. H. Femoro-acetabular Impingement: Prevalence and Importance in Patients Undergoing Hip Resurfacing. J Surg Orthop Adv, Under review, 2013.

109. Plate, J. F.; Pace, L. A.; Seyler, T. M.; Moreno, R. J.; Smith, T. L.; Tuohy, C. J.; and Mannava, S. Age-related Changes Affect Rat Rotator Cuff Muscle Function. J Shoulder Elbow Surg, 23,1:91-98, 2014.

110. Brown, P. J.; Sikes, C. V.; Mannava, S; Seyler, T. M.; Lang, J. E.; Stitzel, J. D. Biomechanical Comparison of A Single Large Diameter and Multiple Small Diameter Femoral Head Core Decompression Techniques. J Bone Joint Surg Am, Under Review, 2013.

197

BOOK CHAPTERS

1. Frey, M. E.; O'Young, B. J.; Marulanda, G. A.; Mont, M. A.; and Seyler, T. M.: The Knee: Anatomy, Pathology, Diagnosis, Treatment, and Rehabilitation. In Physical Medicine and Rehabilitation, pp. 376-391. Edited by O'Young, B. J.; Young, M. A.; and Stiens, S. A., 376-391, Philadelphia, PA, Elsevier Mosby Saunders 2007.

2. Mont, M. A.; Marulanda, G. A.; Delanois, R. E.; Seyler, T. M.; and Friedman, A.: The Hip: Anatomy, Pathology, Diagnosis, Treatment, and Rehabilitation. In Physical Medicine and Rehabilitation, pp. 365-375. Edited by O'Young, B. J.; Young, M. A.; and Stiens, S. A., 365-375, Philadelphia, PA, Elsevier Mosby Saunders, 2007.

3. Seyler, T. M.; Marker, D. R.; and Mont, M. A.: Osteonecrosis. In Primer on the Rheumatic Diseases, pp. 565-572. Edited by Klippel, J. H.; Stone, J. H.; Crofford, L. J.; and White, P. H., 565-572, New York, NY, Springer Science + Business Media, LLC, 2007.

4. Seyler, T. M.; Mont, M. A.; Plate, J. F.; and Bonutti, P. M.: Valgus Approach to Total Knee Arthroplasty. In Navigation and MIS in Orthopaedic Surgery, pp. 240-247. Edited by Stiehl, J. B.; Konermann, W. H.; Haaker, R. G.; and DiGioia III, A. M., 240-247, Heidelberg, Germany, Springer Medizin Verlag, 2007.

5. Ulrich, S. D.; Mont, M. A.; Marker, D. R.; and Seyler, T. M.: Minimally Invasive Approach to Metal-on-Metal Total Hip Resurfacing Arthroplasty. In Minimally Invasive Surgery in Orthopedics, pp. 195-204. Edited by Scuderi, G. R., and Tria, A. J., 195-204, New York, NY, Springer Science + Business Media, LLC, 2010.

6. Lai, L. P.; Seyler, T. M.; and Mont, M. A.: Osteonecrosis of the Hip. In Total Hip Arthroplasty: The Essentials. Edited by Parvizi, J., and Klatt, B. A., Thorofare, NJ, SLACK Incorporated, 2012.

7. Marker, D. R.; Seyler, T. M.; Mont, M. A.; and McCarthy, E. F.: Osteonecrosis and Transient Osteoporosis: Diagnosis, Etiology, Treatment Options. In Surgery of the Hip. Edited by Berry, D. M., and Lieberman, J. R., Philadelphia, PA, Elsevier Mosby Saunders, 2012.

8. Whitlock, P. W.; Seyler, T. M.; Mannava, S.; and Poehling, G. G.: A Tissue-Engineered Approach to Tendon and Ligament Reconstruction. In Sports Injuries. Prevention, Diagnosis, Treatment and Rehabilitation. Edited by Doral, M. M.; Tandoğan, R. N.; Mann, G.; and Verdonk, R., Heidelberg, Germany, Springer Medizin Verlag, 2012

198

MEETING PRESENTATIONS

1. Seyler, T. M.; Takemura, S.; Kang, Y. M.; Bram, R. J.; Kurtin, P. J.; Trousdale, R. T.; Goronzy, J. J.; and Weyand, C. M.: Biological Functions of BLyS and APRIL in Rheumatoid Synovitis. Arthritis & Rheumatism, 2003, Volume 48, Number 9, 67th Annual Scientific Meeting American College of Rheumatology Orlando, FL, 2003.

2. Seyler, T. M.; Park, Y. W.; Takemura, S.; Kang, Y. M.; Bram, R. J.; Goronzy, J. J.; and Weyand, C. M.: BLyS and APRIL in Rheumatoid Synovitis. Clinical and Investigative Medicine, 2004, Volume 27, Number 4, 12th International Congress of Immunology and 4th Annual Conference of FOCIS, Montreal, Canada, 2004.

3. Ragland, P. S.; Mont, M. A.; Marulanda, G. A.; Delanois, R. E.; and Seyler, T. M.: Use of Metal-on-metal Resurfacing Arthroplasty For Avascular Necrosis of the Hip. Podium Presentation, 13th Annual Meeting of Association Research Circulation Osseous (ARCO), Imperial College, London, United Kingdom, 2005.

4. Mont, M. A.; Ragland, P. S.; Marulanda, G. A.; Delanois, R. E.; Flowers, N. B.; and Seyler, T. M.: Core Decompression For Secondary Osteonecrosis Of the Knee Using a Small-Diameter Drilling Technique. Podium Presentation, 13th Annual Meeting of Association Research Circulation Osseous (ARCO), Imperial College, London, United Kingdom, 2005.

5. Ragland, P. S.; Mont, M. A.; Marulanda, G. A.; Delanois, R. E.; Flowers, N. B.; and Seyler, T. M.: Use of a Proximally Hydroxyapatite-Coated Tapered Cementless Stem for Avascular Necrosis of the Hip. Podium Presentation, 13th Annual Meeting of Association Research Circulation Osseous (ARCO), Imperial College, London, United Kingdom, 2005.

6. Marulanda, G. A.; Seyler, T. M.; Delanois, R. E.; and Mont, M. A.: Percutaneous Drilling for the Treatment of Secondary Osteonecrosis of the Knee. Podium Presentation, 65th Annual Meeting Maryland Orthopaedic Society, Curtis National Hand Center, The Union Memorial Hospital, Baltimore, MD, 2006.

7. Marker, D. R.; Mont, M. A.; Seyler, T. M.; Hungerford, D. S.; and Jones, L. C.: Total Knee Arthroplasty in High Activity Patients. Best Resident/Fellow Research Award Maryland Orthopaedic Association, 65th Annual Meeting Maryland Orthopaedic Society, Curtis National Hand Center, The Union Memorial Hospital, Baltimore, MD, 2006.

8. Bhave, A.; Mont, M. A.; Seyler, T. M.; Marker, D. R.; and Delanois, R. E.: Functional Problems and Treatment Solutions Following Total Hip Arthroplasty. Podium Presentation., 16th Annual Fall Meeting American Association of Hip and Knee Surgeons, Gaylord Texan Resort, Dallas, TX, 2006.

9. Bonutti, P. M.; Seyler, T. M.; McMahon, M.; Marker, D. R.; and Mont, M. A.: Unicompartmental Knee Arthroplasty: Implant Survival & Risk Factors at 11-year Follow-up. Poster Presentation, 25th Annual Meeting Mid-America Orthopaedic Association, Boca Raton Resort & Club, Boca Raton, FL, 2007.

10. Bonutti, P. M.; Seyler, T. M.; and Mont, M. A.: Comparison of a Midvastus and Subvastus Approach to Total Knee Arthroplasty. Poster Presentation, 25th Annual Meeting Mid-America Orthopaedic Association, Boca Raton Resort & Club, Boca Raton, FL, 2007.

199

11. Ulrich, S. D.; Mont, M. A.; Marker, D. R.; and Seyler, T. M.: Incidence and Reasons for Wasting Components in the Operative Room. Can this Problem be Reduced? Podium Presentation, 66th Annual Meeting Maryland Orthopaedic Society, University of Maryland Medical Center, Baltimore, MD, 2007.

12. Marker, D. R.; Seyler, T. M.; Ulrich, S. D.; and Mont, M. A.: The Effectiveness of Gram Stains From Multiple Mediums in Total Joint Arthroplasty. Podium Presentation, 66th Annual Meeting Maryland Orthopaedic Society, University of Maryland Medical Center, Baltimore, MD, 2007.

13. Wenz, W.; Akbar, M.; Annefeld, M.; Marker, D. R.; Mont, M. A.; and Seyler, T. M.: Glucosamine Sulfate Therapy Treatment for Articular Damage in Canine Model. Poster Presentation, 53rd Annual Meeting Orthopaedic Research Society, San Diego, CA, 2007.

14. Lancin, P.; Essner, A.; Seyler, T. M.; Mont, M. A.; Wang, A.; and Delanois, R. E.: Effect of Acetabular Cup Abduction Angle on UHMWPE Wear in Hip Simulator Testing. Poster Presentation, 53rd Annual Meeting Orthopaedic Research Society, San Diego, CA, 2007.

15. Lee, D. K.; Hwang, S.; Choi, K. B.; Lee, H. Y.; Yi, Y.; Mont, M. A.; Seyler, T. M.; Meschter, C.; Copeland, O.; and Lee, K. H.: Preclinical Studies of Retrovirally-transduced Human Chondrocytes Expressing TGF-Beta1 (TG-C). Podium Presentation, 53rd Annual Meeting Orthopaedic Research Society, San Diego, CA, 2007.

16. Strassmair, M.; Bosebeck, H.; Mont, M. A.; and Seyler, T. M.: The Use of a Type I Lyophilisate Collagen as Osteoinductive Factor in Pseudoarthroses of the Forearm. Podium Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

17. Bhave, A.; Delanois, R. E.; Mont, M. A.; Paley, D.; Plate, J. F.; and Seyler, T. M.: Peroneal Nerve Dysfunction after TKA: Combined Surgical and Physical Therapy Protocol. Poster Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

18. Bhave, A.; Mont, M. A.; Starr, R.; Delanois, R. E.; Marker, D. R.; and Seyler, T. M.: Gait and Strength Analysis for Successful Correction of Functional Problems Following THA. Poster Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

19. Bhave, A.; Seyler, T. M.; Starr, R.; Plate, J. F.; and Mont, M. A.: Analysis and Correction of Pathological Gait Patterns and Functional Problems after TKA. Podium Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

20. Mont, M. A.; Delanois, R. E.; Seyler, T. M.; Plate, J. F.; and Bhave, A.: Botulinum Toxin Type A Injections for the Management of Flexion Contractures Following TKA. Podium Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

21. Marulanda, G. A.; Mont, M. A.; Seyler, T. M.; and Delanois, R. E.: Percutaneous Perforations for the Treatment of Osteonecrosis of the Ankle. Podium Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

200

22. Akbar, M.; Balean, G.; Seyler, T. M.; Mont, M. A.; Ludwig, K.; Gerner, H. J.; and Loew, M.: Do Paraplegic Patients Walk on Their Arms? A Matched Comparison of Paraplegic Patients and Controls. Podium Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

23. Marulanda, G. A.; Mont, M. A.; Seyler, T. M.; and Delanois, R. E.: Technique and Results of Percutaneous Perforations for Avascular Necrosis of the Shoulder. Podium Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

24. Bonutti, P. M.; Seyler, T. M.; McMahon, M.; Marker, D. R.; and Mont, M. A.: Unicompartmental Knee Arthroplasty: Implant Survival and Risk Factors at 11-year Follow-up. Poster Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

25. Kolisek, F. R.; Monesmith, E. A.; Jessup, N.; Davis, K.; Seyler, T. M.; and Mont, M. A.: Total Knee Arthroplasty Utilizing Cementless Keels and Cemented Tibial Trays: Ten Year Results. Poster Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

26. Clohisy, J. C.; Seyler, T. M.; Kolisek, F. R.; Delanois, R. E.; Plate, J. F.; and Mont, M. A.: Total Hip Arthroplasty in Patients Younger Than 25 Years of Age. Poster Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

27. Delanois, R. E.; Seyler, T. M.; Plate, J. F.; Marker, D. R.; and Mont, M. A.: Selection Criteria for Joint Aspirations as a Diagnositic Test for Infection After Joint Arthroplasty. Podium Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

28. Ketterl, R.; Seyler, T. M.; and Mont, M. A.: Treatment of Bone Defects Using a Collagen Based Osteoinductive Scaffold. Poster Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

29. Seyler, T. M.; Bhave, A.; Plate, J. F.; Marker, D. R.; Marulanda, G. A.; Delanois, R. E.; and Mont, M. A.: Functional Problems and Arthrofibrosis Following Total Knee Arthroplasty. Scientific Exhibit Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

30. Bonutti, P. M.; Seyler, T. M.; Marker, D. R.; Plate, J. F.; and Mont, M. A.: Inventing Orthopaedics: From Design to Working Product. Scientific Exhibit Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

31. Seyler, T. M.; Bonutti, P. M.; Kester, M. A.; and Mont, M. A.: The Use of an Alumina-on-alumina Bearing System in THA for Avascular Necrosis of the Hip. Poster Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

32. Plate, J. F.; Seyler, T. M.; Marker, D. R.; Delanois, R. E.; and Mont, M. A.: Are Gram Stains Necessary for the Diagnosis of Infected TKA? Poster Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

33. Mont, M. A.; Seyler, T. M.; Marker, D. R.; Marulanda, G. A.; and Delanois, R. E.: Metal-on-metal Hip Resurfacing in Avascular Necrosis of the Femoral Head. Poster

201

Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

34. Marker, D. R.; Plate, J. F.; Seyler, T. M.; Delanois, R. E.; and Mont, M. A.: Diagnosing Total Joint Arthroplasty Infections: A Standardized Protocol. Scientific Exhibit Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

35. Mont, M. A.; Perez, O. A.; Moskal, J. T.; Hozack, W. J.; Steinberg, M. E.; Stiehl, J. B.; Hungerford, D. S.; Marulanda, G. A.; and Seyler, T. M.: Hip Surgery Complications: Multicenter Study of Femoral Stem Fractures after Total Hip Arthroplasty. Poster Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

36. Marker, D. R.; Seyler, T. M.; Delanois, R. E.; Plate, J. F.; and Mont, M. A.: The Sensitivity of Gram Stains from Multiple Sites in Patients with Infected Total Hip Arthroplasty. Poster Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

37. Mont, M. A.; Bonutti, P. M.; Marker, D. R.; Plate, J. F.; Marulanda, G. A.; and Seyler, T. M.: Minimally Invasive Knee Arthroplasty: Early Complications and Difficulties. Scientific Exhibit Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

38. Mont, M. A.; Delanois, R. E.; Plate, J. F.; and Seyler, T. M.: Femoral Neck Fractures Following Metal-on-metal Total Hip Resurfacing. Podium Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

39. Mont, M. A.; Amstutz, H. C.; Boyd, H. S.; Schmalzried, T. P.; Vail, T. P.; Sparling, E. A.; Kennedy, W. R.; Seyler, T. M.; and Goldberg, V. M.: Modern Generation Metal-on-metal Total Hip Resurfacings: Results of a Prospective FDA-IDE Study. Poster Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

40. Mont, M. A.; Jones, L. C.; Marulanda, G. A.; Seyler, T. M.; and Hungerford, D. S.: Effect of Epoetin Alfa on Blood Transfusions During Total Knee and Hip Arthroplasty. Poster Presentation, 74th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2007.

41. Bonutti, P. M.; Mont, M. A.; Plate, J. F.; and Seyler, T. M.: Osteonecrosis Of The Knee After Laser And Radiofrequency-Assisted Arthroscopy: Treatment With Minimally Invasive Knee Arthroplasty. E-poster Presentation, 6th Biennial Congress International Society of Arthroscopy, Knee Surgery & Orthopaedic Sports Medicine, Fortezza da Basso, Florence, Italy, 2007.

42. Bonutti, P. M.; Mont, M. A.; McMahon, M.; Ragland, P. S.; and Seyler, T. M.: Minimally Invasive Total Knee Arthroplasty: Complications and Pitfalls. Podium Presentation, 6th Biennial Congress International Society of Arthroscopy, Knee Surgery & Orthopaedic Sports Medicine, Fortezza da Basso, Florence, Italy, 2007.

43. Seyler, T. M.; Bonutti, P. M.; Ragland, P. S.; and Mont, M. A.: Minimally Invasive Valgus Approach to Total Knee Arthroplasty: A comparative Study. E-poster Presentation, 6th Biennial Congress International Society of Arthroscopy, Knee Surgery & Orthopaedic Sports Medicine, Fortezza da Basso, Florence, Italy, 2007.

44. Mont, M. A.; Bonutti, P. M.; Sharkey, P. F.; Hozack, W. J.; Zelicof, S. B.; Kolisek, F. R.; Purtill, J. J.; Rothman, R. H.; and Seyler, T. M.: Clinical Experience Using A

202

Minimally Invasive Surgical Approach for TKA: Early Results of a Prospective Randomized Multicenter Study compared to a Standard Approach. E-poster Presentation, 6th Biennial Congress International Society of Arthroscopy, Knee Surgery & Orthopaedic Sports Medicine, Fortezza da Basso, Florence, Italy, 2007.

45. Mont, M. A.; Seyler, T. M.; and Ragland, P. S.: Functional Limits in Patients with Successful Total Knee Arthroplasty. E-poster Presentation, 6th Biennial Congress International Society of Arthroscopy, Knee Surgery & Orthopaedic Sports Medicine, Fortezza da Basso, Florence, Italy, 2007.

46. Mont, M. A.; Marker, D. R.; Ulrich, S. D.; and Seyler, T. M.: The Effect of High Impact Sports on Total Knee Arthroplasties. Podium Presentation, 17th Annual Fall Meeting American Association of Hip and Knee Surgeons, Gaylord Texan Resort, Dallas, TX, 2007.

47. Bonutti, P. M.; Ulrich, S. D.; Marker, D. R.; Mont, M. A.; and Seyler, T. M.: Unicompartmental Knee Arthroplasty: Implant Survival and Risk Factors for Implant Failure at 11-year Follow-Up. Poster Presentation., 17th Annual Fall Meeting American Association of Hip and Knee Surgeons, Gaylord Texan Resort, Dallas, TX, 2007.

48. Seyler, T. M.; Ward, W. G.; Sprinkle, D. I.; and Jinnah, R. H.: Does Computer-assisted Surgery Aid in Shortening the Learning Curve in Metal-on-metal Surface Replacement? Poster Presentation, 20th Annual Congress of the International Society for Technology in Arthroplasty, Paris, France, 2007.

49. Marker, D. R.; Seyler, T. M.; Shilt, J. S.; LaPorte, D. M.; Mont, M. A.; and Frassica, F. J.: Current Literature: An Educational Tool to Study Osteonecrosis for the Orthopaedic In-Training Examination. Podium Presentation, 14th Annual Meeting of Association Research Circulation Osseous (ARCO), Baltimore, MD, 2007.

50. Boes, L.; Bosebeck, H.; Ulrich, S. D.; Mont, M. A.; and Seyler, T. M.: Collagen-Based Osteoinductive Bone Void Filler for MIS-Treatment in Osteochondritis dissecans and Osteonecrosis. Podium Presentation, 14th Annual Meeting of Association Research Circulation Osseous (ARCO), Baltimore, MD, 2007.

51. Marker, D. R.; Seyler, T. M.; Ulrich, S. D.; Srivastava, S.; and Mont, M. A.: Do Modern Techniques Improve Core Decompression Outcomes for Hip Osteonecrosis? Podium Presentation, 14th Annual Meeting of Association Research Circulation Osseous (ARCO), Baltimore, MD, 2007.

52. Ketterl, R.; Denzel, C.; Bosebeck, H.; Mont, M. A.; Seyler, T. M.; and Ulrich, S. D.: Treatment of Fractures and Nonunions Using Type-I Lyophilisate Collagen as an Osteoinductive Factor. Poster Presentation, 23rd Annual Meeting Orthopaedic Trauma Association, Boston, MA, 2007.

53. Lee, R. K.; Lancin, P.; Bhatt, M.; Essner, A.; Wang, A.; Mont, M. A.; and Seyler, T. M.: Effect of Lubricant Protein Concentration on the Wear of UHMWPE Tibial Inserts in Knee Simulation. Poster Presentation, 6th Combined Meeting of the Orthopaedic Research Societies, Honolulu, HI, 2007.

54. Seyler, T. M.; Jinnah, R. H.; Marker, D. R.; and Mont, M. A.: Accuracy Of Diagnosing Periprosthetic Infections By Both Cultures And Tissue Sampling. Podium Presentation, 25th Annual Meeting Southern Orthopaedic Association, The Homestead, Hot Spring, VA, 2008.

203

55. Seyler, T. M.; Marker, D. R.; Jinnah, R. H.; and Mont, M. A.: Influence Of Athletic Activities On Metal-On-Metal Total Hip Resurfacing Arthroplasty Outcomes. Podium Presentation, 25th Annual Meeting Southern Orthopaedic Association, The Homestead, Hot Spring, VA, 2008.

56. Seyler, T. M.; Jinnah, R. H.; Lai, L. P.; Sprinkle, D. I.; and Ward, W. G.: Does Computer-Assisted Navigation Aid In Resident Education And Shorten The Learning Curve n Hip Resurfacing? Podium Presentation, 25th Annual Meeting Southern Orthopaedic Association, The Homestead, Hot Spring, VA, 2008.

57. Whitlock, P. W.; Seyler, T. M.; Smith, T. L.; Poehling, G. G.; and Koman, L. A.: Effect of Strain on a Novel Cell-Seeded Scaffold for Tendon and Ligament Reconstruction. Poster Presentation, 54th Annual Meeting Orthopaedic Research Society, San Francisco, CA, 2008.

58. Bonutti, P. M.; Seyler, T. M.; Marker, D. R.; Rudert, L.; Dethmers, D.; Ulrich, S. D.; and Mont, M. A.: Navigation-Assisted TKA versus Minimally Invasive TKA: Benefits and Drawbacks. Podium Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

59. Kolisek, F. R.; Mont, M. A.; Jessup, N.; Marker, D. R.; Seyler, T. M.; and Barnes, C. L.: A Prospective Randomized Study of Posterior-Stabilized vs. Posterior Cruciate Ligament-Retaining TKA. Poster Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

60. Marker, D. R.; Seyler, T. M.; Lee, M.; Ulrich, S. D.; Delanois, R. E.; Akbar, M.; and Mont, M. A.: Does Metal-on-Metal Resurfacing Provide Comparable Results to Standard THA? Podium Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

61. Marker, D. R.; Seyler, T. M.; Ulrich, S. D.; Martimbianco, A. L.; Delanois, R. E.; and Mont, M. A.: Influence of Athletic Activities on Metal-on-Metal Hip Resurfacing Arthroplasty Outcomes. Podium Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

62. Marker, D. R.; Seyler, T. M.; Ulrich, S. D.; Martimbianco, A. L.; Srivastava, S.; and Mont, M. A.: High Impact Sports After THA: Is the Bearing Type an Independent Predictor of Activity Level? Podium Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

63. Marker, D. R.; Seyler, T. M.; Delanois, R. E.; Ulrich, S. D.; Srivastava, S.; and Mont, M. A.: Treatment of Early-Stage Osteonecrosis of the Femoral Head. Scientific Exhibit Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

64. Mont, M. A.; Seyler, T. M.; Kolisek, F. R.; Ulrich, S. D.; Bonutti, P. M.; and Marker, D. R.: Is Functional Improvement After TKA Related to Increased Sports Activity in Osteoarthritis Patients? Poster Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

65. Mont, M. A.; Seyler, T. M.; Vogelstein, B.; Diaz, L. A., Jr.; Conway, J. D.; Kinzler, K. W.; Foss, C. A.; Ulrich, S. D.; and Pomper, M. G.: Imaging of Musculoskeletal Bacterial Infections by [124]FIAU-PET/CT. Poster Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

66. Mont, M. A.; Ulrich, S. D.; Delanois, R. E.; Marker, D. R.; Akbar, M.; and Seyler, T. M.: Accuracy of Cultures and Tissue Sampling in Diagnosing Periprosthetic

204

Infections? Poster Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

67. Srivastava, S.; Mont, M. A.; Marker, D. R.; Ulrich, S. D.; Delanois, R. E.; and Seyler, T. M.: Clinical Efficacy of rhBMP-2 and rhBMP-7: Meta-Analysis of the Literature. Scientific Exhibit Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

68. Seyler, T. M.; Marker, D. R.; Ulrich, S. D.; Akbar, M.; and Mont, M. A.: Nonvascularized Bone Grafting for Avascular Necrosis of the Femoral Head. Paper Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

69. Seyler, T. M.; Smith, B. P.; Marker, D. R.; Ma, J.; Shen, J.; Smith, T. L.; Mont, M. A.; and Koman, L. A.: Botulinum Neurotoxin as a Therapeutic Modality in Orthopaedics: 20 Years of Experience. Scientific Exhibit Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

70. Srivastava, S.; Ulrich, S. D.; Marker, D. R.; Mont, M. A.; and Seyler, T. M.: Scientific Evidence for the Use of rhBMP-2 and rhBMP-7. Paper Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

71. Ulrich, S. D.; Bonutti, P. M.; Marker, D. R.; Dethmers, D. A.; Reinhold, J. H.; Robinson, Y.; Jones, L. C.; Seyler, T. M.; and Mont, M. A.: Computer-Assisted Surgery in Total Joint Arthroplasty: Complications, Pitfalls, and Solutions. Scientific Exhibit Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

72. Ulrich, S. D.; Childress III, L.; Marker, D. R.; Seyler, T. M.; and Mont, M. A.: Intra-Articular Injections of the Knee: A Prospective, Randomized Study to Evaluate Pain Intensity. Podium Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

73. Ulrich, S. D.; Mont, M. A.; Seyler, T. M.; Marker, D. R.; Morrey, B. F.; and Bonutti, P. M.: Evaluation of a 30-Minute Protocol to Restore Range of Motion in Posttraumatic Elbow contractures. Podium Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

74. Ulrich, S. D.; Mont, M. A.; Seyler, T. M.; Marker, D. R.; Robinson, Y.; Delanois, R. E.; and Bonutti, P. M.: Stress Relaxation and Static Progressive Stretch in the Treatment of Shoulder Contractures. Poster Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

75. Bonutti, P. M.; Marker, D. R.; Ulrich, S. D.; McMahon, M.; Rudert, L.; Seyler, T. M.; and Mont, M. A.: Evaluation of an All-Polyethylene Tibial Component Design in UKA. Poster Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

76. Bonutti, P. M.; Mont, M. A.; Seyler, T. M.; Childress III, L.; Marker, D. R.; Delanois, R. E.; and Ulrich, S. D.: The Use of Stress Relaxation and Static Progressive Stretch in Postoperative Wrist Contractures. Poster Presentation, 75th Annual Meeting American Academy of Orthopaedic Surgeons, San Francisco, CA, 2008.

77. Akbar, M.; Bresch, B.; Seyler, T. M.; Bruckner, T.; Carstens, C.; and Abel, R. F.: Fractures in Myelomeningocele: A Trend and Risk Analysis. Podium Presentation,

205

76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

78. McGrath, M. S.; Ulrich, S. D.; Bonutti, P. M.; Smith, J.; Seyler, T. M.; and Mont, M. A.: Evaluation of Static Progressive Stretch For the Treatment of Wrist Stiffness. Podium Presentation, 76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

79. Ulrich, S. D.; Bhave, A.; McGrath, M. S.; Seyler, T. M.; Marker, D. R.; Delanois, R. E.; and Mont, M. A.: Botulinum Toxin Type A Injections for the Management of Muscle Tightness Following THA. Podium Presentation, 76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

80. Akbar, M.; Bresch, B.; Seyler, T. M.; Wentz, W.; Bruckner, T.; Abel, R. F.; and Carstens, C.: Management of Orthopaedic Sequelae of Congenital Spinal Disorders. Scientific Exhibit Presentation, 76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

81. Marker, D. R.; LaPorte, D. M.; Seyler, T. M.; McGrath, M. S.; Ulrich, S. D.; and Mont, M. A.: Orthopaedic Journal Publications and Their Role in the Preparation for the OITE. Scientific Exhibit Presentation, 76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

82. Akbar, M.; Brunner, M.; Seyler, T. M.; Grieser, T.; Gerner, H. J.; and Loew, M.: Risk Factors for Rotator Cuff Tears in Wheelchair Users? A Case Control Study. Podium Presentation, 76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

83. Bonutti, P. M.; Ulrich, S. D.; Marker, D. R.; Seyler, T. M.; McGrath, M. S.; McMahon, M.; and Mont, M. A.: A Comparison of Subvastus and Midvastus Approaches in Total Knee Arthroplasty. Podium Presentation, 76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

84. Marker, D. R.; Ulrich, S. D.; McGrath, M. S.; Seyler, T. M.; Delanois, R. E.; and Mont, M. A.: Factors that Influence Weight Gain or Loss Following Total Hip Arthroplasty. Podium Presentation, 76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

85. Seyler, T. M.; Marker, D. R.; Boyd, H. S.; McGrath, M. S.; and Mont, M. A.: Pre-operative Evaluation to Determine Surgical Candidates for Metal-on-metal Resurfacing. Scientific Exhibit Presentation, 76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

86. Bonutti, P. M.; Seyler, T. M.; Ulrich, S. D.; Delanois, R. E.; Marker, D. R.; McGrath, M. S.; Capello, W. N.; D'Antonio, J. A.; and Mont, M. A.: Effect of Obesity on Clinical Success and Patient Satisfaction in Total Hip Arthroplasty. Poster Presentation, 76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

87. Marker, D. R.; Seyler, T. M.; Blackett, J. W.; McGrath, M. S.; and Mont, M. A.: Patient Expectations Regarding Activity Level Following Total Hip Resurfacing Arthroplasty. Poster Presentation, 76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

88. Marker, D. R.; Seyler, T. M.; Bhave, A.; McGrath, M. S.; Ulrich, S. D.; and Mont, M. A.: Does Commitment to Rehabilitation Influence Clinical Outcome of Total Hip

206

Resurfacing Arthroplasty? Podium Presentation, 76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

89. McGrath, M. S.; Bonutti, P. M.; McKenzie, S. A.; Ulrich, S. D.; Seyler, T. M.; and Mont, M. A.: Static Progressive Stretch for the Treatment of Knee Stiffness Following Total Knee Arthroplasty. Podium Presentation, 76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

90. Whitlock, P. W.; Van Dyke, M. W.; Poehling, G. G.; Smith, T. L.; Marker, D. R.; Koman, L. A.; and Seyler, T. M.: Functional Tissue Engineering of Tendon and Ligaments. Scientific Exhibit Presentation, 76th Annual Meeting American Academy of Orthopaedic Surgeons, Las Vegas, NV, 2009.

91. Seyler, T. M.; Maguire, C. M.; Jinnah, R. H.; and Reichard, S.: Metal-on-metal Hip Resurfacing in Young Patients: The Role of Femoroacetabular Impingement. Poster Presentation, 26th Annual Meeting Southern Orthopaedic Association, Amelia Island Plantation, Amelia Island, FL, 2009.

92. Maguire, C. M.; Seyler, T. M.; and Jinnah, R. H.: Metal-on-metal Hip Resurfacing in Young Patients: The Role of Femoroacetabular Impingement. Podium Presentation, 22nd Annual Congress of the International Society for Technology in Arthroplasty, Big Island, HI, 2009.

93. Shields, J. S.; Seyler, T. M.; Maguire, C. M.; and Jinnah, R. H.: Computer-Assisted Navigation in Hip Resurfacing: Learning Curve in the Hands of an Experienced Hip Surgeon. Podium Presentation, 22nd Annual Congress of the International Society for Technology in Arthroplasty, Big Island, HI, 2009.

94. Smith, T. L.; Seyler, T. M.; Van Dyke, M. W.; Poehling, G. G.; and Whitlock, P. W.: Development, Characterization and Cell Seeding of a Naturally-Derived Scaffold for Anterior Cruciate Ligament Reconstruction. Poster Presentation, Annual Meeting Biomedical Engineering Society, Pittsburgh, PA, 2009.

95. Smith, T. L.; Whitlock, P. W.; Koman, L. A.; Van Dyke, M. W.; and Seyler, T. M.: Effect of Strain on a Novel Cell-Seeded Scaffold for Tendon and Ligament Reconstruction. Podium Presentation, Annual Meeting Biomedical Engineering Society, Pittsburgh, PA, 2009.

96. Mannava, S.; Callahan, M. F.; Koman, L. A.; Plate, J. F.; Seyler, T. M.; Smith, T. L.; and Tuohy, C. J.: Novel Use of Electromyography (EMG) to Evaluate In Vivo Rotator Cuff Muscle Function After Acute and Chronic Detachment of the Supraspinatus Tendon: An Experimental Study in an Animal Model Poster Presentation, 27th Annual Meeting Southern Orthopaedic Association, El Conquistador, Fajardo, Puerto Rico, 2010.

97. Mannava, S.; Callahan, M. F.; Koman, L. A.; Plate, J. F.; Seyler, T. M.; Smith, T. L.; and Tuohy, C. J.: Evaluation of In Vivo Rotator Cuff Muscle Function After Acute and Chronic Detachment of the Supraspinatus Tendon: An Experimental Study in an Animal Model. Podium Presentation, 27th Annual Meeting Southern Orthopaedic Association, El Conquistador, Fajardo, Puerto Rico, 2010.

98. Seyler, T. M., and Jinnah, R. H.: Clinical Outcomes of Lateral UKA Using an All-Polyethylene Design. Podium Presentation, 27th Annual Meeting Southern Orthopaedic Association, El Conquistador, Fajardo, Puerto Rico, 2010.

207

99. Seyler, T. M.; Jinnah, R. H.; and Plate, J. F.: The Effect of Bearing Type on Postoperative Activity Level After THA. Poster Presentation, 27th Annual Meeting Southern Orthopaedic Association, El Conquistador, Fajardo, Puerto Rico, 2010.

100. Seyler, T. M.; Bosebeck, H.; Buchner, H.; Vogt, S.; Hofmann, R.; Walter, G.; Hirschberger, W.; Stemberger, A.; and Kiokekli, M.: Preclinical and Clinical Experience of a Novel Calcium Sulfate/Carbonate Based Bone Substitute with Antibiotic Release. Poster Presentation, 56th Annual Meeting Orthopaedic Research Society, New Orleans, LA, 2010.

101. Marker, D. R.; Zywiel, M. G.; Stroh, A.; Strimbu, K.; McGrath, M. S.; Seyler, T. M.; and Mont, M. A.: The Use of Erythrocyte Sedimentation Rates and C-Reactive Protein Levels to Confirm TKA Infection. Poster Presentation, 77th Annual Meeting American Academy of Orthopaedic Surgeons, New Orleans, LA, 2010.

102. Akbar, M.; Doustdar, S.; Hemmer, S.; Seyler, T. M.; Furstenberg, C. H.; Lehner, B.; and Wiedenhofer, B.: Operative Treatment of Spinal Infections: Evaluation of Treatment Concepts from 1996-2008. Scientific Exhibit Presentation, 77th Annual Meeting American Academy of Orthopaedic Surgeons, New Orleans, LA, 2010.

103. Seyler, T. M.; Marker, D. R.; Bonutti, P. M.; Kolisek, F. R.; McGrath, M. S.; Zywiel, M. G.; Johnson, A. J.; and Mont, M. A.: Athletic Activity Following Total Hip Arthroplasty: Assessment and Outcomes. Scientific Exhibit Presentation, 77th Annual Meeting American Academy of Orthopaedic Surgeons, New Orleans, LA, 2010.

104. Hemmer, S.; Furstenberg, C. H.; Seyler, T. M.; Putz, C.; Ewerbeck, V.; Lehner, B.; Wiedenhofer, B.; and Akbar, M.: Primary and Metastatic Spinal Tumors: Evidence-based Treatment Strategies. Scientific Exhibit Presentation, 77th Annual Meeting American Academy of Orthopaedic Surgeons, New Orleans, LA, 2010.

105. Akbar, M.; Zastrow, R.; Bruckner, T.; and Seyler, T. M.: Upper Extremity Sequelae in Spinal Cord Injuries: An Epidemiological Survey Study of 670 Patients. Podium Presentation, 77th Annual Meeting American Academy of Orthopaedic Surgeons, New Orleans, LA, 2010.

106. Seyler, T. M.; Whitlock, P. W.; Smith, T. L.; Van Dyke, M. W.; and Poehling, G. G.: A Tissue-Engineered Scaffold Derived from Human Allograft Tissue for Use in ACL Reconstruction. Podium Presentation, OREF/ORS Atlantic Coast Region Resident Research Symposium, Durham, NC, 2010.

107. Mannava, S.; Saul, K. R.; Plate, J. F.; Seyler, T. M.; Koman, L. A.; Smith, T. L.; and Tuohy, C. J.: Rotator Cuff Dysfunction After Acute and Chronic Tears: Translating Animal Studies to Clinical Recommendations Using Simulation Analysis. Podium Presentation, OREF/ORS Atlantic Coast Region Resident Research Symposium, Durham, NC, 2010.

108. Mannava, S.; Saul, K. R.; Plate, J. F.; Seyler, T. M.; Koman, L. A.; Tuohy, C. J.; and Smith, T. L.: Rotator Cuff Dysfunction After Acute and Chronic Tears: Translating Animal Studies to Clinical Recommendations Using Simulation Analysis. Gold Medal Winner of Basic Science Award, Division of Surgical Sciences Research Day, Wake Forest University, School of Medicine, Winston-Salem, NC, 2010.

109. Seyler, T. M., and Jinnah, R. H.: Outcomes of Lateral UKA Using an All-Polyethylene Design. Podium Presentation, 41st Annual Meeting Eastern Orthopaedic Association, The Ritz Carlton Naples, Naples, FL, 2010.

208

110. Smith, T. L.; Whitlock, P. W.; Seyler, T. M.; Northam, C. N.; Van Dyke, M. W.; Poehling, G. G.; and Koman, L. A.: Effect of Strain on the Tensile Properties and mRNA Expression of a Tissue-Engineered Flexor Tendon. Podium Presentation, 2010 Annual Meeting Biomedical Engineering Society, Austin Convention Center, Austin, TX, 2010.

111. Mannava, S.; Tuohy, C. J.; Seyler, T. M.; Stitzel, J. D.; Hayon, S.; Whitlock, P. W.; Smith, T. L.; and Saul, K. R.: The Science of Rotator Cuff Repairs: Translating Basic Science into Clinical Recommendations. Scientific Exhibit Presentation, 78th Annual Meeting American Academy of Orthopaedic Surgeons, San Diego, CA, 2011.

112. Mannava, S.; Seyler, T. M.; Pace, L. A.; Whitlock, P. W.; Smith, B. P.; Koman, L. A.; Smith, T. L.; and Tuohy, C. J.: Age-Related Changes Affect Supraspinatus Muscle Function: An Experimental Study in an Animal Model. Poster Presentation, 57th Annual Meeting Orthopaedic Research Society, Long Beach, CA, 2011.

113. Whitlock, P. W.; Seyler, T. M.; Smith, T. L.; Parks, G. D.; Van Dyke, M. W.; and Poehling, G. G.: A Tissue-Engineered Scaffold Derived From Human Allograft Tissue for Use in Anterior Cruciate Ligament Reconstruction. Poster Presentation, 57th Annual Meeting Orthopaedic Research Society, Long Beach, CA, 2011.

114. Sikes, C. V.; Brown, P. J.; Danelson, K. A.; Seyler, T. M.; Mannava, S.; Jinnah, R. H.; Stitzel, J. D.; and Lang, J. E.: Biomechanical Analysis of Hip Core Decompression Techniques and Fracture Risk. Poster Presentation, 57th Annual Meeting Orthopaedic Research Society, Long Beach, CA, 2011.

115. Mannava, S.; Koman, L. A.; Seyler, T. M.; Plate, J. F.; Tuohy, C. J.; Saul, K. R.; and Smith, T. L.: Translating Animal Studies to Clinical Recommendations with Computational Analysis for the Study of Rotator Cuff Dysfunction After Acute and Chronic Tears. Silver Medal Winner Integrative Science Category, Post-doctoral Research Day, Wake Forest University, Graduate School of Arts and Sciences, Winston-Salem, NC, 2011.

116. Whitlock, P. W.; Seyler, T. M.; Parks, G. D.; Smith, T. L.; Van Dyke, M. W.; and Poehling, G. G.: Development and Characterization of a Tissue-Engineered Scaffold For Anterior Cruciate Ligament Reconstruction Derived From Human Achilles Tendon Allograft. Podium Presentation, 8th Biennial Congress International Society of Arthroscopy, Knee Surgery & Orthopaedic Sports Medicine, Riocentro Convention Center, Rio de Janeiro, Brazil, 2011.

117. Schweppe, M. L.; Goddard, M. S.; Seyler, T. M.; Plate, J. F.; Mannava, S.; and Lang, J. E.: A clinical comparative study of the direct anterior with the posterior approach in primary THA. Podium Presentation, Annual Meeting North Carolina Orthopaedic Association, Kiawah Island Golf Resort, Kiawah Island, SC, 2011.

118. Plate, J. F.; Mannava, S.; Seyler, T. M.; Saul, K. R.; Pace, L. A.; Smith, T. L.; and Tuohy, C. J.: Age influences Healing and Function of Rotator Cuff Muscles. Podium Presentation, Annual Meeting North Carolina Orthopaedic Association, Kiawah Island Golf Resort, Kiawah Island, SC, 2011.

119. Seyler, T. M.; Pulley, B.; and Lang, J. E.: A Multivariant Retrospective Analysis of Preoperative Nutritional Parameters as Predictors of Outcomes in Joint Arthroplasty. Podium Presentation, 31st Annual Oscar Miller Day Symposium, The Blake Hotel, Charlotte, NC, 2011.

209

120. Plate, J. F.; Stone, A. V.; Seyler, T. M.; Marker, D. R.; Akbar, M.; and Mont, M. A.: Are Cultures and Frozen Sections Reliable in Diagnosing Periprosthetic Infections? Podium Presentation, 31st Annual Oscar Miller Day Symposium, The Blake Hotel, Charlotte, NC, 2011.

121. Plate, J. F.; Stone, A. V.; Seyler, T. M.; Marker, D. R.; Akbar, M.; and Mont, M. A.: Accuracy of diagnosing periprosthetic infections by both cultures and frozen sections. Podium Presentation, 99th Annual Meeting The Clinical Orthopaedic Society, Francis Marion Hotel, Charleston, SC, 2011.

122. Plate, J. F.; Stone, A. V.; Seyler, T. M.; Akbar, M.; and Mont, M. A.: Comparison of Dislocation Rates in Patients with large Diameter Metal-on-Metal and Small Diameter Metal-on-Polyethylene Bearings in Total Hip Arthroplasty. Podium Presentation, 99th Annual Meeting The Clinical Orthopaedic Society, Francis Marion Hotel, Charleston, SC, 2011.

123. Plate, J. F.; Seyler, T. M.; Halvorson, J. J.; Santago, A. C.; and Lang, J. E.: Non-Anatomic Closure of a Standard Parapatellar Knee Arthrotomy Leads to Patella Mal-Tracking and Decreased Range of Motion. Podium Presentation, Annual Meeting North Carolina Orthopaedic Association, Pinehurst Resort, Village of Pinehurst, NC, 2012.

124. Plate, J. F.; Seyler, T. M.; Halvorson, J. J.; Santago, A. C.; and Lang, J. E.: Non-Anatomic Closure of a Standard Parapatellar Knee Arthrotomy Leads to Patella Mal-Tracking and Decreased Range of Motion. Podium Presentation, 100th Annual Meeting The Clinical Orthopaedic Society (COS), The Palmer House, Chicago, IL, 2012.

210

ELECTRONIC MEDIA

1. Mont, M.A.; Delanois, R.E.; Seyler, T.M.; Plate, J.F. New Treatment Approaches to Osteonecrosis of the Femoral Head. In: Marsh, J.L, ed. Instructional Course Lectures, Video Supplement. Vol. 56. Rosemont, IL: American Academy of Orthopaedic Surgeons, 2007.

2. Mont, M.A.; Marulanda, G.A.; Seyler, T.M.; Plate, J.F.; Delanois, R.E. Core Decompression and Nonvascularized Bone Grafting for the Treatment of Early-Stage Osteonecrosis of the Femoral Head. In: Marsh, J.L., ed. Instructional Course Lectures, Video Supplement. Vol. 56. Rosemont, IL: American Academy of Orthopaedic Surgeons, 2007.

3. Bonutti, P.M.; Mont, M.A.; Seyler, T.M.; Plate, J.F. Minimally Invasive Total Knee Arthroplasty: The Mini-Subvastus Approach. Orthopaedic Theaters. Vol. 8. Rosemont, IL: American Academy of Orthopaedic Surgeons, 2008.

4. Bonutti, P.M.; Mont, M.A.; Seyler, T.M.; Plate, J.F. Minimally Invasive Total Knee Arthroplasty: The Mini-Midvastus Approach. Orthopaedic Theaters. Vol. 8. Rosemont, IL: American Academy of Orthopaedic Surgeons, 2008.

211

RESEARCH GRANTS

1. Seyler, T. M.; Poehling, G. G.; Whitlock, P. W.; Smith, T. L.; and Van Dyke, M. W.: A tissue-engineered approach to tendon and ligament reconstruction using human Achilles tendon allograft-derived scaffolds. Sponsor: Musculoskeletal Transplant Foundation. Amount: Non-Transplantable Tissue Program Grant. Duration: 2007.

2. Seyler, T. M.; Poehling, G. G.; Van Dyke, M. W.; Whitlock, P. W.; and Smith, T. L.: Tissue Engineering of Tendons and Ligaments Using Adipose-Derived Stem Cells and a Novel Naturally-Derived Scaffold. Sponsor: Orthopaedic Research and Education Foundation. Amount: $19,230.00. Duration: 7/2008 - 7/2008.

3. Seyler, T. M.; Poehling, G. G.; Whitlock, P. W.; Smith, T. L.; and Van Dyke, M. W.: A tissue-engineered approach to tendon and ligament reconstruction using human Achilles tendon allograft-derived scaffolds. Sponsor: Community Blood Center, Community Tissue Services, Dayton, Ohio. Amount: Non-Transplantable Tissue Program Grant. Duration: 2008.

4. Seyler, T. M.; Whitlock, P. W.; Smith, T. L.; Van Dyke, M. W.; and Poehling, G. G.: ACL Reconstruction in a Rabbit Model using a Novel, Biocompatible Human Achilles Allograft Tissue-derived Ligament Scaffold. Sponsor: Arthroscopy Association of North America. Amount: $24,980.00. Duration: 4/2008 - 4/2010.

5. Tuohy, C. J.; Mannava, S.; Seyler, T. M.; Smith, T. L.; Whitlock, P. W.; and Van Dyke, M. W.: Optimization of bioreactor preconditioning protocols for the maturation of seeded, naturally-derived scaffold. Sponsor: Wake Forest University Intramural Research Grant. Amount: $20,000.00. Duration: 3/2010 - 2/2012.

212

PERMISSIONS TO REPRINT PUBLISHED ARTICLES IN

DOCTORAL THESIS

213

214

215


Recommended