238
Tissue Engineering Bone - Reconstruction of Critical Sized Segmental Bone Defects in a Large Animal Model Johannes Christian Reichert, MD Faculty of Built Environment and Engineering, School of Engineering Systems, Queensland University of Technology Thesis submitted for: Doctor of Philosophy (PhD) 2010

Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

  • Upload
    others

  • View
    8

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

Tissue Engineering Bone - Reconstruction of

Critical Sized Segmental Bone Defects in a

Large Animal Model

Johannes Christian Reichert, MD

Faculty of Built Environment and Engineering, School of Engineering

Systems, Queensland University of Technology

Thesis submitted for:

Doctor of Philosophy (PhD)

2010

Page 2: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized
Page 3: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

I

Keywords

Bone, segmental defect, tibia, tissue engineering, scaffold, tricalcium-

phosphate, polycaprolactone, bone morphogenetic protein, osteoblasts,

mesenchymal stem cells

Page 4: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

II

Page 5: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

III

Abstract

Currently, well established clinical therapeutic approaches for bone

reconstruction are restricted to the transplantation of autografts and

allografts, and the implantation of metal devices or ceramic-based implants

to assist bone regeneration. Bone grafts possess osteoconductive and

osteoinductive properties, their application, however, is associated with

disadvantages. These include limited access and availability, donor site

morbidity and haemorrhage, increased risk of infection, and insufficient

transplant integration. As a result, recent research focuses on the

development of complementary therapeutic concepts. The field of tissue

engineering has emerged as an important alternative approach to bone

regeneration. Tissue engineering unites aspects of cellular biology,

biomechanical engineering, biomaterial sciences and trauma and

orthopaedic surgery. To obtain approval by regulatory bodies for these novel

therapeutic concepts the level of therapeutic benefit must be demonstrated

rigorously in well characterized, clinically relevant animal models.

Therefore, in this PhD project, a reproducible and clinically relevant, ovine,

critically sized, high load bearing, tibial defect model was established and

characterized as a prerequisite to assess the regenerative potential of a

novel treatment concept in vivo involving a medical grade polycaprolactone

and tricalciumphosphate based composite scaffold and recombinant human

bone morphogenetic proteins.

Page 6: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

IV

Page 7: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

V

Table of contents

Keywords I

Abstract III

Table of contents V

List of illustrations and diagrams XI

List of abbreviations XVII

Statement of original authorship XIX

Acknowledgments XXI

Introduction 1

Chapter I – Preclinical models for segmental bone defect research

5

- Clinical background 7

- Introduction 9

- Definition of critical-sized defect 9

- Large animal models in bone defect research 11

- Tibial fracture models 15

- Tibial segmental defect models 19

- Summary 33

Chapter II – Ovine bone and marrow derived progenitor cells: Isolation, Characterization, and Osteogenic Potential

37

- Introduction 39

- Materials and Methods 41

- Isolation of ovine MPC and OB 41

- Flow cytometric analysis 42

- Cell proliferation assay 43

- CFU-F clonogenic assay 43

Page 8: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

VI

- 2D differentiation in vitro 44

- Dynamic cell culture 44

- Alkaline phosphatase activity 45

- Aliyarin red staining 45

- Wako HR II calcium assay 46

- X-ray photoelectron spectroscopy 47

- Immunohistochemistry 47

- Total RNA isolation, primer design and qRT-PCR 48

- 3D cultures 49

- Scanning electron microscopy 50

- Confocal laser microscopy 50

- In vivo transplantation studies 51

- !CT analysis 52

- Histology 52

- Image analysis 53

- Statistical analysis 53

- Results 54

- MPC show a higher proliferation rate than OB in vitro 54

- MPC and OB exhibit a similar phenotype 55

- Clonogenic efficiency of MPC and OB 56

- 2D differentiation potential of ovine MPC and OB in vitro 56

- Static vs. dynamic culture 60

- 3D differentiation potential of MPC and OB in vitro 65

- Differentiation potential of MPC and OB in vivo 67

- Discussion 70

- Conclusion 76

Page 9: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

VII

Chapter III – Ovine bone and marrow derived progenitor cells and their

Potential for Scaffold based Tissue Engineering

Applications in vivo

77

- Introduction 79

- Materials and Methods 81

- Isolation of ovine MPC and OB 81

- Scaffold fabrication and cell seeding 81

- Cell sheet fabrication 82

- In vivo transplantation studies 83

- BrdU labelling of cells 84

- Biomechanical testing 85

- !CT analysis 86

- Immunohistochemistry 86

- Histochemistry 87

- Tartrate resistant acid phosphatase (TRAP) staining 88

- Detection of BrdU-labelled cells 88

- SEM and EDX 89

- Image analysis 89

- Statistical analysis 90

- Results 91

- Biomechanical testing 91

- !CT analysis 91

- Histology 93

- SEM and EDX 107

- Discussion 108

- Conclusion 114

Page 10: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

VIII

Chapter IV – Establishment of a Preclinical Ovine Model for Tibial

Segmental Bone Defect Repair by Bone Tissue

Engineering Methods

115

- Introduction 117

- Regulatory framework 119

- Intramedullary nail versus plate fixation versus external

fixator

124

- Road map to establish a preclinical model for segmental bone

defect research

128

- Pilot study limited contact locking compression plate

(LC-LCP)

129

- Finite element modelling

131

- Implant testing 132

- Pilot study dynamic compression plate 134

- Summary 137

Chapter V – Reconstructing large segmental bone defects in an ovine

model by tissue engineering methods

139

- Introduction 141

- Materials and Methods 145

- Scaffold fabrication and preparation 145

- Anaesthesia and pre-operative treatment 147

- Defect model 146

- Harvest of autologous cancellous bone graft 150

- Experimental groups 151

- Euthanasia 151

- Radiographic analysis 152

- Computed tomography 152

- Biomechanical testing 154

Page 11: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

IX

- !CT analysis 155

- Histology 157

- Statistical analysis 157

- Results 158

- Animal model 158

- X-ray analysis 158

- Computed tomography 160

- Biomechanical testing 165

- !CT analysis 168

- Histology 173

- Discussion 176

- Summary 185

Conclusions and Recommendations 187

Bibliography 191

Page 12: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

X

Page 13: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

XI

List of illustrations and diagrams

Chapter I

Figure 1: PMMA sections of ovine tibial bone demonstrating secondary

osteon formation Table 1: Factors influencing the size of a critical sized defect Table 2: Bone biomechanical properties of different species Table 3: Overview of segmental bone defect studies Table 4: Comparison of animal models for fracture and segmental bone

defect research Table 5: Summary of human and large animal bone properties

Chapter II

Figure 1: Mesenchymal progenitor cell and osteoblast isolation and

expansion Figure 2: Uncoated and collagen I coated polycaprolactone tricalcium-

phosphate scaffold Figure 3: Ovine mesenchymal progenitor cell and osteoblast proliferation

over seven days in monolayer Figure 4: Surface antigen expression of ovine mesenchymal progenitor

cells and osteoblasts Figure 5: Clonogenic efficiency of ovine mesenchymal progenitor cells

and osteoblasts Figure 6: Alizarin red, osteocalcin, and type I collagen staining of

mesenchymal progenitor cell and osteoblast monolayers after 28 days of osteogenic induction

Figure 7: Quantification of alizarin red incorporated in mesenchymal

progenitor cell and osteoblast monolayer cultures after 28 days of osteogenic culture

Figure 8: Quantification of alkaline phosphatase activity at day 14 and 28

of osteogenic culture in monolayer

Page 14: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

XII

Figure 9: Quantitative RT-PCR for osteogenic markers after 28 days of culture under osteogenic conditions in monolayer

Figure 10: Alkaline phosphatase activity under static and dynamic

monolayer culture conditions Figure 11: Alizarin red quantification at day 14 of static and dynamic

osteogenic monolayer cultures Figure 12: Calcium amounts in dynamic cultures after 14 days Figure 13: XPS analysis of ovine bone and Thermanox coverslips Figure 14: XPS analysis of extracellular matrix synthesized by ovine

mesenchymal progenitor cells and osteoblasts Figure 15: Proliferation of mesenchymal progenitor cells and osteoblasts

under static and dynamic conditions Figure 16: Scanning electron microscopy, FDA-PI and Phalloidin-DAPI

staining of mesenchymal progenitor cells and osteoblasts cultured on mPCL-TCP scaffolds for 28 days

Figure 17: MicroCT analysis of mesenchymal progenitor cell and

osteoblast 3D cultures on collagen I coated polycaprolactone tricalcium-phosphate scaffolds

Figure 18: Histology, histomorphometry and microCT analysis of

transplanted in vivo specimens after eight weeks

Chapter III

Figure 1: Schematic illustrating the in vitro generation of a transplantable

tissue engineered construct Figure 2: Immunohistochemical staining for BrdU of an ovine

mesenchymal progenitor cell monolayer culture Figure 3: Compressive stiffness values of subcutaneously transplanted

tissue engineered constructs after eight weeks Figure 4: 3D microCT reconstructions of subcutaneously transplanted

tissue engineered constructs after eight weeks Figure 5: Bone volume fractions and bone mineral density of

transplanted tissue engineered constructs after eight weeks as determined by microCT analysis

Page 15: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

XIII

Figure 6: H&E staining on paraffin sections of cell free mPCL-TCP scaffolds that were transplanted alone, in combination with fibrin glue or with fibrin glue containing rhBMP-7

Figure 7: H&E staining on paraffin sections of mesenchymal progenitor

cell or osteoblast seeded mPCL-TCP scaffolds that were transplanted in combination with or without rhBMP-7

Figure 8: Von Kossa/van Gieson staining on PMMA sections of cell free

mPCL-TCP scaffolds that were transplanted alone, in combination with fibrin glue or with fibrin glue containing rhBMP-7

Figure 9: Histomorphometric analysis of mineralized tissue within

transplanted tissue engineered constructs Figure 10: Histomorphometric analysis of neovascularisation within

transplanted tissue engineered constructs Figure 11: Von Kossa/van Gieson staining on PMMA sections of

mesenchymal progenitor cell or osteoblast seeded mPCL-TCP

scaffolds that were transplanted in combination with or without

rhBMP-7

Figure 12: High magnification of PMMA sections stained for von

Kossa/van Gieson showing differences in morphology of bone lining cells

Figure 13: Immunohistochemistry for osteocalcin on paraffin sections Figure 14: Alcian blue staining on paraffin sections demonstrating

glycosaminoglycan deposits Figure 15: Immunohistochemistry for type II collagen on paraffin sections Figure 16: Histochemical staining for tartrate resistant acid phosphatase

on paraffin sections illustrating osteoclast activity Figure 17: Immunohistochemical staining and histomorphometry for BrdU

labelled cells on paraffin sections Figure 18: Backscattered scanning electron microscopy and Energy

dispersive X-ray spectroscopy

Page 16: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

XIV

Chapter IV

Figure 1: Flow chart describing a road map to establish a critically sized defect model

Figure 2: Schematic of commonly applied methods for segmental defect

fixation Figure 3: Image showing a segmental tibial defect of 2 cm length

stabilized with a LC-LCP Figure 4: Image of different implants chosen for biomechanical testing

and set up of the four point bending test Figure 5: Equivalent bending stiffnesses of tested implants Figure 6: Image illustrating a segmental tibial defect of 2 cm length

stabilized with a DCP, postoperative radiograph and x-ray after 12 week

Table 1: Advantages and disadvantages of different fixation devices

Chapter V

Figure 1: 3D microCT reconstruction of a cylindrical mPCL-TCP scaffold

of 3 height and 2cm diameter Figure 2: Image series demonstrating the application of and OP-implant

to a cylindrical mPCL-TCP scaffold Figure 3: Intraoperative images of the critical size defect creation in an

ovine tibia Figure 4: Image series illustrating the harvesting procedure of autografts

from the iliac crest Figure 5: 3D CT reconstruction of a 3 cm tibial defect overlayed with a

developed CT scoring system Figure 6: DICOM image of an intact ovine tibia (axial view)

Figure 7: Embedding procedure for torsional testing Figure 8: Schematic illustrating the method of calculation for the polar

moment of inertia Figure 9: Illustration of the cutting planes for histological sectioning

Page 17: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

XV

Figure 10: Radiographs demonstrating bone formation within defects of

the different experimental groups after 12 weeks

Figure 11: Bar graphs demonstrating union rates and determined CT

scores for the different experimental groups

Figure 12: 3D CT reconstructions of representative specimens of each

experimental group

Figure 13: Box plots demonstrating the total bone volumes and newly

formed bone in the cortical region determined by quantitative

analysis of CT scans

Figure 14: Bone volumes determined for the marrow and external callus

regions determined by quantitative analysis of CT scans

Figure 15: Box plot illustrating torsional moment values measured for the

different experimental groups

Figure 16: Box plot showing torsional stiffness values calculated for the

different experimental groups

Figure 17: MicroCT sections and 3D microCT reconstructions of

representative samples of the different experimental groups

Figure 18: Box plots demonstrating the volume of newly formed bone

within the defects determined by quantitative microCT analysis

Figure 19: Bone volume distribution along the z axis of the 3 cm defects

Figure 20: Box plots showing the tissue mineral density of the newly

formed bone within the 3 cm defects

Figure 21: Box plots illustrating the trabecular thicknesses for proximal,

medial and distal defect portions and thickness distribution

along the z axis.

Figure 22: Box plots demonstrating the polar moment of inertia calculated

for the 3 cm defect regions of the different experimental groups

Figure 23: Histological sections of the entire 3 cm defects of each group

stained for Safranin O/von Kossa and Movat’s pentachrome

Page 18: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

XVI

Figure 24: Higher magnification images of histological defect sections

stained for Movat’s pentachrome showing the different

composition of tissue formed within the 3 cm defects

Figure 25: High magnification images of PMMA sections stained for

Movat’s pentachrome illustrating the different degrees of bone

maturation within the different experimental groups

Page 19: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

XVII

List of abbreviations

2D Two dimensional 3D Three dimensional ABG Autologous bone graft ALP Alkaline phosphatase AO Arbeitsgemeinschaft osteosynthese ARC Australian Research Council bFGF Basic fibroblast growth factor BMP Bone morphogenetic protein BrdU 5-bromo-2-deoxyuridine BSA Bovine serum albumin BV Bone volume CAD Computer aided design CD Cluster of differentation CFU Colony forming unit CO2 Carbon dioxide CSD Critical sized defect DAPI 4',6-diamidino-2-phenylindole DCP Dynamic compression plate DMEM Dulbecco’s modified eagle media DNA Deoxyribonucleic acid ECM Extracellular matrix EDTA Ethylene-di-amine-tetra-acetic acid EDX Energy dispersive X-ray

spectroscopy EU European union f frequency FACS Fluorescence activated cell sorting FBS Foetal bovine serum FDA Fluorescein diacetate FDA Food and drug administration FDM Fused deposition modelling FEM Finite element modelling FGF Fibroblast growth factor FITC Fluorescein isothiocyanate GAGs Glycosaminoglycans GDF Growth differentiation factor H2O water HCl Hydrogen chloride HE Haematoxylin-eosin HGF Hepatocyte growth factor IGF-1 Insulin like growth factor 1 ISO International organization for

standardization LC-DCP Limited contact dynamic

compression plate LC-LCP Limited contact locking compression

plate LISS Less invasive stabilization system

Page 20: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

XVIII

MPC Mesenchymal progenitor cell mPCL-TCP Medical grade poly-caprolactone

tricalcium-phosphate MSC Mesenchymal stem cell MVF Mineralized volume fraction (!)CT (Micro) computed tomography NaOH Sodium hydroxide NOD/SCID Nonobese diabetes/severe combined

immunodeficiency OB Osteoblast OC Osteocalcin OP-1 Osteogenic protein 1 PBS Phosphate buffered saline PC-Fix Point contact fixator PCL Poly-caprolactone PDGF Platelet derived growth factor PDLA Poly-D-lactic acid PEG Polyethylene glycol PGA Poly-glycolic acid PI Propidium iodide PLA Poly-lactic acid PLLA Poly-L-lactic acid PMMA Poly-methyl-methacrylate pMOI Polar moment of inertia PRP Platelet rich plasma rh Recombinant human RNA Ribonucleic acid RT-PCR Real time polymerase chain reaction SD Standard deviation SEM Scanning electron microscopy TCP Tricalcium-phosphate TEC Tissue engineered construct TGF Transforming growth factor TRAP Tartrate resistant acid phosphatase UHN Universal humeral nail UTN Universal tibial nail VEGF Vascular endothelial growth factor XPS X-ray photoelectron spectroscopy

Page 21: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

XIX

Statement of original authorship

“The work contained in this thesis has not been previously submitted to meet

requirements for an award at this or any other higher education institution. To

the best of my knowledge and belief, the thesis contains no material

previously published or written by another person except where due

reference is made.”

Friday, 18 June 2010

Page 22: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

XX

Page 23: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

XXI

Acknowledgements

I would like to gratefully acknowledge Prof. Dietmar W. Hutmacher for his

friendship and enthusiastic supervision of my work. I thank Prof. Michael

Schütz and Prof. Georg Duda for stimulating, critical discussions, their

support and intellectual input, and Dr. Martin Wullschleger, Dr. Siamak

Saifzadeh and the team of the QUT Medical Engineering Research Facility

for their assistance with the animal surgeries. I’m grateful to Dr. Maria Ann

Woodruff and Dr. Devakara Epari for their friendship, advice and support in

matters of histology, finite element modelling, biomechanics, image analysis,

manuscript writing and proof reading.

I’d further like to thank the members of the IHBI Regenerative Medicine

Group and my fellow research students for companionship, inspiration and

advice.

Finally, I’d like to express gratitude to my wife Verena for her love and

support, sacrifice, patience and proofreading and to my parents and siblings

for support, understanding, patience and encouragement over the years.

Page 24: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

XXII

Page 25: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

1

Introduction

Bone as such displays a high intrinsic regenerative potential. Consequently,

the majority of fractures and bone defects heal spontaneously. This healing

process recapitulates pathways of embryonic development following complex

yet well orchestrated biological patterns.

Improved surgical techniques and biologically favourable implant designs as

well as novel peri-operative management strategies have significantly

decreased undesirable treatment complication rates. However, various

factors such as a compromised wound environment and biomechanical

instability can result in large defects with impaired healing capacity. In many

aspects, these cases pose a major challenge and dramatically influence

patients’ quality of life. The treatment options are then mainly restricted to

bone graft transplantations (autograft, allograft) or segmental bone transport.

These grafts, however, are limited in availability, face integration related

problems, carry the risk of infection, and are associated with donor site

morbidity. Bone transport on the other hand is a long-lasting, inconvenient

procedure with recurrent pin track infections as a frequent complication.

As a result, recent research efforts have focused on the development and

application of bone graft substitutes and the concept of tissue engineering

has emerged as it unites aspects of cellular biology, biomechanical

engineering, biomaterial sciences, and trauma and orthopaedic surgery.

Tissue engineering generally involves the association of growth factors

and/or cells with a naturally derived or synthetically manufactured,

mechanically supporting scaffold to produce a three-dimensional,

Page 26: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

2

implantable construct. Complying with these principles, it was the overall aim

of the present study to develop a bone graft substitute consisting of a

suitable scaffold and a bone growth stimulating agent that results in

comparable or even better bone healing when compared to the standard

cancellous autograft transplantation.

The performance of such bone graft substitutes is usually evaluated in large

animal models as these models simulate human in vivo conditions as closely

as possible. However, most of the preclinical animal models are not well

described, defined or standardized as summarized in Chapter I, which

provides comprehensive information on the advantages and disadvantages

of the different animal models described in the literature. Over the last ten

years, sheep have become increasingly popular as a model species to

investigate bone remodelling and turnover and it was therefore decided to

assess the performance of the developed bone graft substitute in an ovine

critically sized tibial defect. Surprisingly, the molecular and cellular events

surrounding osteoneogenesis in these models have only been scarcely

investigated. Knowledge of cellular behaviours and molecular processes,

and comparison with data available on human conditions, however, provides

essential information pertaining to the validity of an animal model.

It has been demonstrated that - amongst others - mesenchymal progenitor

cells and osteoblasts exhibit distinct intrinsic osteogenic characteristics, are

capable of synthesizing bone-like extracellular matrix, and therefore play a

major role in bone formation in humans.

Hypothesizing that ovine marrow derived cells of seven to eight year old

sheep are equivalent to those previously described for adult humans,

Page 27: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

3

populations of marrow cells derived from the iliac crest and cells from

compact bone were isolated, characterized, and compared regarding

phenotype, genotype, and osteogenic potential in vitro using techniques

already established for human mesenchymal progenitor cells as described in

Chapter II of this study.

The processes of de novo bone formation in vivo are partly attributed to

members of the transforming growth factor-" superfamily, specifically the

bone morphogenetic proteins (BMPs). In particular BMP-7 was shown to

have not only osteoinductive but also angiogenic effects. However, the

molecular and cellular events surrounding osteoneogenesis remain poorly

understood with respect to neovascularisation, and the recruitment and

differentiation of osteogenic precursor cell populations. The potential of ex

vivo expanded ovine marrow and bone derived cells to produce tissues with

properties consistent with those of mature bone and the influence of

recombinant human BMP-7 on bone formation was therefore assessed in

vivo in a small animal model of ectopic bone formation. It was hypothesized

that bone cell origin, ossification type, and degree of vascularisation and

bone neoformation is dependent on the nature and commitment of

transplanted cells as well as supplemented growth factors such as BMP-7.

The results obtained from these experiments are summarized in Chapter III

of this study.

Translations from bench to bedside in the field of bone tissue engineering

are still infrequent which might be related to difficulties in integrating

individual technical discoveries in model tissue engineering systems, in

manufacturing scale up, in funding, and in regulatory approval. Translating a

Page 28: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

4

tissue engineering concept to a clinical setting requires a rigorous

demonstration of the level of therapeutic benefit in clinically relevant animal

models. As preclinical models published are difficult to reproduce, effort was

subsequently concentrated on the establishment of a well characterized,

standardized preclinical animal model. The approach of how to translate

tissue engineering of bone from bench to bedside is illustrated in Chapter IV.

In this chapter a road map is presented which describes the validation of the

functionality of a highly load-bearing large animal model to study the

regeneration of critically sized segmental bone defects.

The last chapter of the present study finally summarizes the large animal

study designed to investigate the effects of recombinant human BMP-7 in

combination with a composite scaffolds built by computer aided design using

rapid prototyping technologies on bone healing in an ovine, tibial, critically

sized, segmental bone defect.

Page 29: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

5

Chapter I

Preclinical animal models for segmental bone defect research

Page 30: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

6

Page 31: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

7

Clinical Background

The vast majority of fractures and bone defects heal spontaneously. This

process is stimulated by well balanced biological and micro-environmental

conditions. Newly introduced, improved surgical techniques, implant designs

and peri-operative management strategies have procured better treatment

outcomes of complex fractures and other skeletal defects caused by high

energy trauma, disease, and tumours [1-6]. However, a compromised wound

environment, biomechanical instability and other factors can result in large

defects with limited intrinsic regeneration potential [7]. Such defects pose a

major surgical, socio-economical and research challenge, and highly

influence patients’ quality of life [8, 9].

Cancellous bone fractures of the proximal humerus, distal radius, or the tibial

plateau can lead to bone impaction and consequently defect formation after

reduction [4]. The tibial diaphysis, however, represents the most common

anatomic site for segmental bone defects since it is devoid of muscle

coverage on its anteromedial surface [8]. This poor soft tissue coverage both

increases the risk of bone loss and complicates treatment [8].

Over the years, bone autografts have advanced as the “gold standard”

treatment to augment and accelerate bone regeneration [1, 2, 10-16]. The

application of autografts, however, is associated with considerable negative

side effects. Graft harvest leads to prolonged anaesthetic periods and

requires personnel [12, 14, 17]. Often, insufficient amounts of graft can be

obtained while the access to donor sites is limited [12, 13, 18, 19]. Donor site

morbidity (persistent pain and haemorrhage) is common, the risk of infection

is increased, and the transplanted bone is predispositioned to failure [4, 12,

Page 32: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

8

13, 20]. Graft failures usually result from incomplete transplant integration,

particularly in large defects [14]. In addition, graft devitalisation due to

insufficient graft vascularisation and subsequent resorption processes can

lead to decreased mechanical stability [21].

The transplantation of vascularised autografts is time consuming and

technically demanding. Allografts and xenografts carry the risk of immune-

mediated rejection, graft sequestration, and transmission of infectious

disease [9, 22-28]. The dense nature of cortical bone allografts impedes

revascularization and cellular invasion from host sites following implantation

[18]. This limited ability to revascularize and remodel is believed to account

for an allograft associated failure rate of 25% and a complication rate of 30-

60% [18, 29]. In addition, the maintenance of bone banks is associated with

considerable operating expenses.

A technique introduced to avoid graft integration-related difficulties known as

the “Ilizarov technique”, involves the osteotomy of bone combined with

distraction to stimulate bone formation. This procedure has been applied

successfully to treat large bone defects, infected non-unions, and limb length

discrepancy [30]. However, the Ilizarov technique is a long-lasting procedure,

inconvenient for the patient [31, 32], and recurrent pin track infections are a

frequent complication [24, 33].

In order to avoid the limitations associated with the current standard

treatment modalities for segmental bone deficiencies, research efforts have

focused on the use of naturally derived and synthetic bone graft substitutes

during the past decades.

Page 33: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

9

More recently, the concept of tissue engineering has emerged as an

important approach to bone regeneration related research. Tissue

engineering unites aspects of cellular biology, biomechanical engineering,

biomaterial sciences, and trauma and orthopaedic surgery. Its general

principle involves the association of cells with a natural or synthetic

supporting scaffold to produce a three-dimensional, implantable construct.

Introduction

To biomechanically simulate human in vivo conditions as closely as possible,

and to assess the effects of implanted bone grafts and tissue engineered

constructs on segmental long bone defect regeneration, a number of large

animal models have been developed. However, most of the preclinical

models published are not well described, defined or standardized. In 2008,

the Journal of Bone and Joint Surgery published a number of review papers

on preclinical models in fracture healing and non-unions [34]. However,

these articles provide only rudimentary information on how to establish

relevant segmental bone defects in a preclinical large animal model. Hence,

the following chapter provides detailed, comprehensive information on the

advantages and disadvantages of the different published animal models.

Definition of a Critical-Sized Bone Defect

It has been postulated that an experimental osseous injury inflicted to study

bone repair mechanisms needs to be of dimensions to preclude spontaneous

healing [35]. Therefore, the non-regenerative threshold of bone was

determined in different research animal models inducing so-called “critical

Page 34: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

10

sized” defects. These critical sized defects are defined as “the smallest size

intraosseous wound in a particular bone and species of animal that will not

heal spontaneously during the lifetime of the animal” [29, 36, 37] or as a

defect which shows less than ten percent bony regeneration during the

lifetime of an animal [37].

Table 1: Factors influencing the size of a critical defect

The minimum size that renders a defect ‘‘critical’’ is not well understood. For

practical reasons, it has been defined as a segmental bone deficiency of a

length exceeding 2-2.5 times the diameter of the affected bone [24, 33].

Results of various animal studies suggest that critical sized defects in sheep

however, could even be approximately three times the diameter of the

corresponding diaphysis [33]. Nevertheless, a critical defect in long bones

cannot simply be defined by its size, but also depends on the species

phylogenetic scale, anatomic defect location, associated soft tissue, and

biomechanical conditions in the affected limb as well as age, metabolic and

systemic conditions, and related co-morbidities affecting defect healing

(Table 1)[24, 36].

Factors determining a CSD [24] [29, 36]

o Age o Species phylogeny o Defect size o Anatomic location o Bone structure and vascularisation o Presence of periosteum o Adjacent soft tissue o Mechanical loads and stresses on the limb o Metabolic and systemic conditions o Fixation method/stiffness o Nutrition

Page 35: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

11

Large animal models in bone defect research

Animal models in bone repair research include representations of normal

fracture-healing, segmental bone defects, and fracture non-unions, in which

regular healing processes are compromised in the absence of a critical-sized

defect site [38]. In critical-sized segmental defect models, bridging of the

respective defect does not occur despite a sufficient biological

microenvironment as critical amounts of bone substance are removed. In

contrast, in true non-unions, deficient signalling mechanisms, biomechanical

stimuli or cellular responses may prevent defect healing rather than the

defect size.

When selecting a specific animal species as a model system, a number of

factors need to be considered. With comparison to humans, the chosen

animal model should clearly demonstrate both significant physiological and

pathophysiological analogies in respect to the scientific question under

investigation. Moreover, it must be manageable to operate and observe a

multiplicity of study objects over a relatively short period of time [39-41].

Further selection criteria include costs for acquisition and care, animal

availability, acceptability to society, tolerance to captivity, and ease of

housing [42].

Over the last decades, several publications have described dogs as a

suitable model for research related to human orthopaedic conditions [43]. It

was found that dogs closely resemble humans with regards to bone weight,

density and bone material constituents such as hydroxyproline, extractable

proteins, IGF-1, organic, inorganic and water fraction although clear

differences in bone microstructure and remodelling have been described [44,

Page 36: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

12

45]. While the secondary structure of human bone is predominantly

organized into osteons, osteonal bone structure in dogs is limited to the core

of cortical bone, whereas in areas adjoining the periosteum and endosteum

mainly laminar bone is found as characteristic for large, fast-growing animals

[46]. It has been reported that generally, higher rates of trabecular and

cortical bone turnover can be observed in dogs compared to humans [47]

and differences in loads acting on the bone, as a result of the dog’s

quadrupedal gait must also be taken into consideration as well. Various

biomechanical properties as described in table 2.

Bone biomechanical properties

Cortical bone

Trabecular bone

Dog Humerus (bending) E: 2.66 (GPa) UStress 193.23 (MPa) [48]

Femur E: 209 (MPa) UStress: 7.1 (MPa) Tibia E: 106-426 (MPa) UStress: 2-24 (MPa) [40]

Sheep Femur (compression) E:19.3 (GPa) UStrain: 0.019 [49] [50]

Tibia E: 1192 (MPa) Ustress: 21.4 (MPa) [51]

Goat Tibia (bending)

E: 278.08 (MPa) Bending strength: 46.24 (MPa) [12]

Femur

E: 399-429 (MPa) UStress: 14.1-23.5 (MPa) Tibia E: 532-566 (MPa) UStress: 24.7-26.1 (MPa)

Pig

Femur E: 14.6 (GPa) (plexiform bone) 8.3 (GPa) (Haversian bone) [52]

Femur E: 5900 (MPa) [40]

Human

Femur (compression) E: 14.7-19.7 (GPa) UStrain: 167-215 (MPa) Tibia (compression) E: 24.5-34.3 (GPa) UStrain: 183-213 (MPa) [40]

Femur E: 298 (MPa) UStrain: 5.6 (MPa) Tibia E: 445 (MPa) UStrain: 5.3 (MPa) [40]

Table 2: Bone biomechanical properties of different animal species and humans.

Page 37: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

13

A review article by Neyt states that between 1991 and 1995 11% of

musculoskeletal research was undertaken in dogs, results that are confirmed

by Martini et al. who found that, between 1970 and 2001, 9% of orthopaedic

and trauma related research used dogs as animal models for orthopaedic

and trauma related research [43, 53]. Recently, the use of dogs as

experimental models has significantly decreased mainly due to ethical

issues, although between 1998 and 2008, still approximately 9% of articles

published in prominent orthopaedic and musculoskeletal journals described

dogs as animal models for fracture healing research [34].

Mature sheep and goats possess a bodyweight comparable to adult humans

and long bone dimensions enabling the use of human implants [54]. The

mechanical loading environment occurring in sheep is well understood [55,

56]. The loading of the hind limb bones, forces and moments, is roughly half

of that determined for humans during walking. Since no major differences in

mineral composition [57] are evident and both metabolic and bone

remodelling rates are akin to humans [58], sheep are considered a valuable

model for human bone turnover and remodelling activity [59]. Bone histology

however reveals some differences in bone structure between sheep and

humans. In sheep, bone consists principally of primary bone structure [60] in

comparison with the largely secondary, haversian bone composition of

humans [61]. Furthermore, the secondary, osteonal remodelling in sheep

does not take place until an average age of 7-9 years (Fig. 1)[54]. Although a

significantly higher trabecular bone density and greater bone strength are

described for mature sheep when compared to humans, the trabecular bone

Page 38: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

14

in immature sheep is weaker, has a lower stiffness and density, a higher

flexibility due to higher collagen content [51], and shows comparable bone

healing potential and tibial blood supply [62].

Fig. 1: Ground and polished bone sections from the tibia of a 7 year old sheep

embedded in poly-methyl-methacrylate (PMMA) and stained with Toluidine blue (A)

and Movat pentachrome (B). The images show secondary bone with clearly

distinguishable osteones (arrows). Secondary osteone formation can only be

observed in sheep older than 7 years and makes the ovine secondary bone

structure comparable to human findings. Bar = 0.05 mm.

In a variety of study designs, pigs are considered the animal of choice and

were - despite their denser trabecular network [63] - described as a highly

representative model of human bone regeneration processes in respect to

anatomical and morphological features, healing capacity and remodelling,

Page 39: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

15

bone mineral density and concentration [44, 64]. However, pigs are often

neglected in favour of sheep and goats given that the handling of pigs has

been described as rather intricate [54]. Furthermore, the short length of the

tibiae and femora in the pig might bring about the need for special implants,

as one cannot use implants designed for human use.

Tibial fracture models

Animal fracture models have been widely investigated to identify and further

characterize physiological and pathophysiological processes surrounding

fracture healing of long bones. One of the most important elements in the

study of fracture healing or fixation is the establishment of standardized

methods to create reproducible fractures. Although a substantial number of

articles on fracture models in animals and treatment options have been

published over the last decades, only few publications describe the actual

infliction of a fracture by trauma rather than the creation of a bony defect < 3

mm size by osteotomy, which is generally accepted as an alternative since it

is problematic to standardize. In 1988, Macdonald et al. [65, 66] reported a

device for the reproducible creation of transverse fractures in canine tibiae

utilizing a three-point bending technique.

Similarly, to compare the effects of reamed versus unreamed locked

intramedullary nailing on cortical bone blood flow, Schemitsch et al. created

a standardized spiral fracture by three-point bending with torsion in a

fractured sheep tibia model [67, 68], a method also described by Tepic [69]

to establish a standardized oblique fracture in sheep tibiae in order to

compare healing in fractures stabilized with either a conventional dynamic

Page 40: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

16

compression plate (DCP) or an internal point contact fixator (PC-Fix). A

minimally invasive approach to create a multifragmental fracture in the sheep

femur (classification by the Association for the Study of Internal Fixation, AO

type 32-C), in which the bone was weakened by two short, transverse

anterior osteotomies and bi-cortical drill holes created through small

incisions, has recently been described by Wullschleger et al. (unpublished

data). The insertion of two chisels and one blade bar were then used to

initiate cracks connecting both the osteotomies and the drill holes, thereby

creating a standardized multifragmental fracture. This technique could easily

be adopted when establishing standardized tibial fractures as well.

Fracture models of osteotomized long bones have been well characterized

over the years in different large animal species. A number of publications

have described fracture models in dogs since the dog, beside pigs, is

considered the most closely related model for research of human

orthopaedic conditions. The effect of bending stiffness of external fixators on

the early healing of transverse tibial osteotomies was described in a canine

model by Gilbert [70]. Tiedemann et al. assessed densitometric approaches

to measure fracture healing in 6 mm tibial segmental defects and single-cut

osteotomy defects in adult mongrel dogs [71]. Bilateral tibial transverse

osteotomies were performed with a 2 mm gap by Markel et al. to quantify

local material properties of fracture callus during gap healing [72]. To

compare the dosage-dependent efficacy of recombinant human bone

morphogenetic protein-2 (rhBMP-2) on tibial osteotomy healing, adult female

dogs underwent right midshaft tibial osteotomies with a 1 mm gap. The

Page 41: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

17

operated bones were stabilized using type I external fixators [73]. In a similar

study by Edwards, bilateral tibial osteotomies were performed to evaluate the

capacity of a single percutaneous injection of rhBMP-2 delivered in a rapidly

resorbable calcium phosphate paste (alpha-BSM) to accelerate bone-healing

[74]. The effect of shock wave therapy on acute tibial fractures was studied

by Wang et al. in adult dogs after creation of bilateral tibial osteotomies with

a 3 mm defined fracture gap [75]. Similar models were also described by

Hupel to compare the effects of unreamed and reamed nail insertion [76].

Jain et al. [77] investigated whether or not the limited contact design of the

low contact dynamic compression plate (LC-DCP) provided advantages over

the dynamic compression plates (DCP) regarding their influence on cortical

bone blood flow, biomechanical properties, and remodelling of bone in

segmental tibial fractures. Nakamura [78] also evaluate the effects of

recombinant human basic fibroblast growth factor (bFGF) on fracture healing

in beagle dogs.

As previously mentioned, mature sheep and goats possess a bodyweight

similar to adult humans, show no major differences in bone mineral

composition with similar metabolic and bone remodelling rates, and therefore

are considered a valuable model for human bone turnover and remodelling

activity often used in fracture research. In the period between 1990 and

2001, sheep as an animal model were used in 9-12% of orthopaedic

research, compared to only 5% between 1980 and 1989 [43]. Over the last

ten years numbers of studies utilizing sheep and goats as animal models

have increased to 11-15% [34].

Page 42: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

18

The significance of postoperative mechanical stability for bony repair of a

comminuted fracture was investigated in a sheep study comparing four

commonly applied operative methods of fracture stabilization. In this study, a

triple-wedge osteotomy of the right sheep tibia was performed [79]. Using a

standard osteotomy of the ovine tibia stabilised by an external skeletal

fixator, Goodship et al. elucidated the influence of fixator frame stiffness on

bone healing rates [80]. Wallace et al. [81] used a similar model to

investigate serum angiogenic factor levels after tibial fracture. Likewise,

transverse mid-diaphyseal osteotomies with an interfragmentary gap of 3

mm, as an experimental fracture model in sheep, were used to assess

fracture repair processes [82-85]. To validate the principle of external fixation

dynamization to accelerate mineralized callus formation by in vivo

measurements of callus stiffness, transverse fractures with an

interfragmentary gap of 3 mm width were created in the mid third of the tibial

diaphysis [86]. Hantes et al. investigated the effect of transosseous

application of low-intensity ultrasound on fracture-healing in a midshaft

osteotomy sheep model [87]. Epari et al. were the first authors to report on

the pressure, oxygen tension and temperature in the early phase of callus

tissue formation of six Merino-mix sheep that underwent a tibial osteotomy to

model fracture conditions [88]. In this study, the tibia was stabilized with a

standard monolateral external fixator. It was found that the maximum

pressure during gait increased from three to seven days. During the same

interval, there was no change in the peak ground reaction force or in the

interfragmentary movement. Oxygen tension in the haematoma was initially

high post surgery and decreased steadily over the first five days. The

Page 43: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

19

temperature increased over the first four days before reaching a plateau on

day four.

Mechanical strain during callus distraction is known to stimulate

osteogenesis. It is, however, unclear whether this stimulus can enhance the

healing of a fracture without affecting bone length. Just recently, Claes et al.,

reported the acceleration of fracture healing by a slow temporary distraction

and compression of a diaphyseal osteotomy [89] in an ovine, mid-diaphyseal

osteotomy fracture model of the right tibia, stabilized by external fixation.

Tibial segmental defect models

In order to ascertain whether newly developed bone graft substitutes or

tissue engineered constructs (TEC) comply with the requirements of

biocompatibility, mechanical stability and safety, the materials must be

subject to rigorous testing, both in vitro and in vivo. To extrapolate results

from in vitro studies to in vivo patient situations, however, is often difficult.

Therefore, the application and systematic evaluation of new concepts in

animal models is often an essential step in the process of assessing newly

developed bone grafts prior to clinical use. To simulate human in vivo

conditions as closely as possible, a variety of large critical sized tibial defect

models - mainly in sheep - have been developed over the past decade in

order to investigate the influence of different types of bone grafts on bone

repair and regeneration. Critical sized segmental defects in long bones are

usually defined by multiplying the diaphyseal diameter by 2.0-2.5 [24, 33].

Interestingly, the method of ostectomy may influence the study outcome.

Page 44: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

20

Kuttenberger et al. could show that CO2-laser osteotomy impaired the

adjacent bone less than oscillating saw osteotomy [90].

To evaluate the effects of different bioceramics on bone regeneration during

repair of segmental bone defects Gao et al. [91] implanted biocoral and

tricalcium phosphate cylinders (TCP) in sheep tibial defects of 16 mm length.

The defects were stabilized medially using two overlapping contoured auto-

compression plates of 4 mm thickness (8 and 6 holes) and cortical screws.

When compared to TCP, a significant increase in external callus formation

and callus density was seen with the biocoral implants after three weeks and

an increase of torque capacity, maximal angle of deformation, and energy

absorption could be measured after 12 weeks while microscopically

osseointegration appeared better. However, in his study, Gao used both

male and female animals with a relatively large variation in body weight. Both

factors, gender and body weight are known to have an influence on bone

regeneration due to effects on both the biomechanical environment and

hormonal feed-back control mechanisms. Hence, variations in sex and body

weight should be avoided. The defect fixation method used in this study can

most likely be interpreted as a means to countervail bending forces on the

implant after earlier failures. However, defect fixation by overlapping plates is

not necessarily lege artis and has never been introduced and applied

clinically. Therefore, a thicker and hence stiffer plate should have been

chosen instead.

Den Boer et al. reported a new segmental bone defect model where a 30

mm segmental defect was inflicted on sheep tibiae and stabilized by an

interlocking intramedullary nail (custom made AO unreamed humeral nail).

Page 45: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

21

X-ray absorptiometry was applied to quantify healing [59]. Groups of this pilot

study included untreated controls and autografts. After 12 weeks, despite

higher bone mineral density in the autograft group, no significant difference

in torsional strength and stiffness could be revealed. Since 33% of the

control animals showed sufficient bridging of the defect, it needs to be

questioned if the authors succeeded in establishing a reliable non-union

model. Removal of the periosteum or a larger defect site might have been

beneficial. In a subsequent study, the authors described the fabrication of

biosynthetic bone grafts and their application in the very same animal model

[4]. The five treatment groups included empty controls, autografts,

hydroxyapatite alone, hydroxyapatite combined with recombinant human

osteogenic protein I (rhOP-1), and hydroxyapatite with autologous bone

marrow. At 12 weeks, healing of the defect was evaluated radiographically,

biomechanically and histologically and revealed a two-fold higher torsional

strength and stiffness for animals treated with autograft and hydroxyapatite

plus rhOP-1 or bone marrow. Since healing was only evaluated after 12

weeks, no conclusions could be drawn regarding the process of healing or

bone remodelling. The mean values of both combination groups were

comparable to those of autografts. A higher number of defect unions was

described when hydroxyapatite plus rhOP-1 was applied rather than

hydroxyapatite alone. Analysing this study, it has to be taken into account

that animals treated with hydroxyapatite and bone marrow were of a different

breed with a higher average body weight. Animals were held at a different

holding facility and accustomed to unequal forage all of which possibly could

have influenced study outcomes.

Page 46: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

22

Bone healing in critical sized segmental diaphyseal defects in sheep tibiae

was also investigated by Gugala et al. [3, 37]. Defects were bridged with a

single porous tubular membrane or with anatomically shaped porous double

tube-in-tube membranes. Membranes with different pore structures were

applied alone and/or in combination with autologous bone graft. The

diaphyseal defects were 40 mm in length and stabilized with a bilateral AO

external fixator. Operated animals were six to seven years of age. Of the six

treatment groups defect healing could only be observed in groups where the

defect was filled with autologous cancellous bone graft and covered with a

single perforated membrane or where the bone graft was administered in a

space between a perforated internal and external membrane. The authors

partly contributed the healing effect to their membrane system; however a

control group, where autologous bone graft is administered without any

membrane was not described. It could also be criticized that post surgery

animals were suspended in slings over the entire experimental period

preventing the animals from sitting and therefore getting up, thus not

reflecting normal, physiological, load bearing conditions.

Wefer et al. [92] conducted a study to develop and test a scoring system

based on real-time ultrasonography to predict the healing of a bone defect.

Defects were filled with a porous hydroxyapatite bone graft substitute or

cancellous bone graft from the iliac crest and stabilized by anterolateral plate

osteosynthesis. After sacrifice, tibiae were tested for torsion to failure. The

results were then correlated with radiographic and ultrasound scores. Sheep

with ceramic implants that developed non-unions showed a significantly

lower score than sheep with sufficient implant integration. A significant

Page 47: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

23

correlation between these scores and the biomechanical results was found.

However, although the authors describe their 20 mm defect as a critical sized

model, no control group was included for proof of principle. Hence, the

critical nature of the defect in this study can be questioned.

The effects of new resorbable calcium phosphate particles and paste forms,

which harden in situ after application, on bone healing were investigated by

Bloemers et al. [93]. A 30 mm segmental tibial defect was established and

fixed by a custom made AO unreamed interlocking titanium tibial nail. Twelve

weeks after defect reconstruction, radiological, biomechanical, and

histological examinations were performed. Radiographically, the resorbable

paste group performed better than all other groups. Biomechanical tests

revealed a significantly higher torsional stiffness for the resorbable calcium-

phosphate paste group in comparison with autologous bone. The study

indicated that new calcium phosphate based materials might be a potential

alternative for autologous bone grafts in humans. As with several other

studies, animals of a minimum age of two years with a significant variation in

body weight were used in this study. As mentioned before, it must be

considered that secondary osteonal bone remodelling in sheep does not

occur until an age of seven to nine years. Therefore, it might be difficult to

extrapolate results from this study for applications in adult human patients as

human bone primarily undergoes secondary osteonal bone remodelling.

Insulin-like growth factor I (IGF-1) exerts an important role during skeletal

growth and bone formation. Therefore, its localized delivery appears

attractive for the treatment of bone defects. To prolong IGF -1 delivery,

Meinel et al. entrapped the protein into biodegradable poly(lactide-co-

Page 48: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

24

glycolide) microspheres and evaluated the potential of this delivery system

for new bone formation in a non-critical 10 mm segmental tibia defect [94].

The defect was stabilized using a 3.5 mm 11 hole DCP. Administration of

100 !g of IGF-1 in the microspheres resulted in bridging of the segmental

defect within 8 weeks. To avoid excessive load on the operated limbs and

fracturing of the freshly operated tibial defects, the animals were

accommodated in a suspension system for a period of 4 weeks

postoperatively thus preventing physiologic-like biomechanical conditions.

When interpreting data published in this study, it must be taken into account

that the close position of the screws to the defect proximally and distally, and

the obvious fact that the screws at the defect site had not been inserted at a

defined angle might have influenced and biased the outcomes.

In a 48 mm tibial defect model in sheep, ceramic implants of 100% synthetic

calcium phosphate multiphase biomaterial were evaluated [95]. The defect

was stabilized with a 4.5 mm neutralizing plate. Although not reported by the

authors, one can observe bent plates and axial deviations in presented x-ray

and CT images, hence, from a clinical point of view, it must be concluded

that the chosen fixation in that model was insufficient. The presented x-ray

series of the two year animal suggests that the internal fixation device had

been explanted 12-14 weeks post surgery, a fact not described and

explained by the authors. Assuming recovery and bone regeneration without

any complications, in human patients, internal fixation devices would usually

not be removed until 12-18 months post implantation. Good integration

between the ceramic implants and the adjoining proximal and distal bone

ends was observed. A progressive increase in new bone formation was seen

Page 49: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

25

over time, along with progressive resorption of the ceramic scaffold. Based

on x-ray analysis, at the one-year time-point, approximately 10% to 20% of

the initial scaffold substance was still present, and after two years it was

almost completely resorbed. The authors state that approximately 10-20% of

the periosteum were deliberately left in situ as a source of osteogenic cells.

However, one might conclude that this procedure appears to be rather

difficult to standardize in order to develop a reproducible model.

Another study using an ovine segmental defect model investigated the

influence of recombinant human transforming growth factor 3 (rhTGF"-3) on

mechanical and radiological parameters of a healing bone defect [96]. In four

to five year old sheep, an 18 mm long osteoperiosteal defect in the tibia fixed

with a unilateral external fixator was treated by rhTGF"-3 delivered by a

poly(L/DL-lactide) carrier, with the carrier only, with autologous cancellous

bone graft, or remained untreated. Weekly in vivo stiffness measurements

and radiological assessments were undertaken as well as quantitative

computed tomographic assessments of bone mineral density in four week

intervals. The follow up of the experiment was twelve weeks under partial

weight bearing since animals were kept in a support system to prevent

critical loads on the fixator and its interface to bone thus not reflecting

physiological loading conditions. The 18 mm defect size described as

spontaneously non-healing, might not have been sufficient to establish a

non-union model in a fully weight bearing biomechanical environment. In the

bone graft group, a significantly higher increase in stiffness was observed

than in the PLA/rhTGF"-3 group and a significantly higher increase than in

the PLA-only group. The radiographic as well as the computer tomographic

Page 50: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

26

evaluation yielded significant differences between the groups, indicating the

bone graft treatment performed better than the PLA/rhTGF"-3 and the PLA-

only treatment.

Sarkar et al. assessed the effect of platelet rich plasma (PRP) on new bone

formation in a 25 mm diaphyseal tibial defect in sheep [97]. The defect was

stabilized with a custom-made intramedullary nail (stainless steel, diameter

proximal 12 mm, distal 10 mm) with two locking screws each proximal and

distal. To reduce stress at the screw/bone interface, a custom made

stainless steel plate was additionally applied medially therefore choosing an

unconventional fixation method not applied clinically. However, no reasoning

for the additional medial plating was provided by the authors. Defects were

treated with autologous PRP in a collagen carrier or with collagen alone. A

control group to demonstrate the critical nature of the defect was also not

included. After 12 weeks, the explanted bone specimens were quantitatively

assessed by X-ray, computed tomography (CT), biomechanical testing and

histological evaluation. Bone volume, mineral density, mechanical rigidity

and histology of the newly formed bone in the defect did not differ

significantly between the PRP treated and the control group, and no effect of

PRP upon bone formation was observed. The aforementioned studies are

summarized in table 3.

Page 51: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

27

Author Animal age (years)

Defect size (mm)

Follow-up (months)

Fixation Animal housing

Support

Gao et al., 1997 a 16 4

Overlapping autocompression plates, 8 and 6 holes, 4 mm thickness

a a

DenBoer et al.,

1999/03 a 30 3

Custom-made AO unreamed nail (Synthes)

a a

Gugala et al.

1999/02 6-7 40 4

AO bilateral external fixator

Single boxes

Suspension slings

Wefer et al., 2000

# 2 20 12 Anterolat. plate osteosynthesis (not specified)

a a

Bloemers et al., 2003

# 2 30 3

AO unreamed tibial nail (Synthes)

6-8 animals in a 60 m

2 cage

a

Meinel et al.,

2003 a 10 5

3.5 mmm DCP, 11 holes a a

Mastrogiacomo

et al, 2006 2 48 12

4.5 mm plate (not specified), 10-12 holes

Single boxes

Fibre glass cast

Maissen et al., 2006

4-5 18 3 Unilateral external fixator Single

boxes Custom-made support system

Sarkar et al., 2006

5.5-7 25 3

Custom-made intramedullary nail plus medial stainless steel plate

Single boxes

a

Tyllianakis et

al., 2007 1-2 10,20,30 4

Universal Humeral Nail (UHN, Synthes)

Single boxes for 3 days post surgery

a

Liu et al., 2007 a 26 8 Circular external fixator Single

boxes a

ainformation not provided by the authors

Table 3: The table lists a selection of publications on segmental bone defect studies

in sheep tibiae and summarizes animal age, selected defect size, defect fixation,

animal housing as well as supportive devices. The majority must be considered

short term studies where no complete bone remodelling can be expected during the

experimental period. In many cases, authors fail to report important information

concerning animal age, housing and supportive devices.

In 2007 Tyllianakis [98] determined the size of a bone defect that can be

restored with one-stage lengthening over a reamed intramedullary nail in

sheep tibiae. Sixteen adult female sheep were divided into four main groups:

a simple osteotomy group (group I) and three segmental defect groups (10,

20, and 30 mm gaps, groups II-IV). One intact left tibia from each group was

also used as the non-osteotomized intact control group (group V). In all

cases, the osteotomy was fixed with an interlocked Universal Humeral Nail

Page 52: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

28

(UHN-Protek-Synthes). Healing of the osteotomies was evaluated after 16

weeks by biomechanical testing. The examined parameters included

torsional stiffness, shear stress, and angle of torsion at the time of fracture.

The regenerate bone in the groups with 10 and 20 mm gaps was of

considerable mechanical properties. Torsional stiffness in these two groups

was nearly equal and values represented about 60% of the stiffness

observed in the simple osteotomy group. Gradually decreasing stiffness was

observed as the osteotomy gap increased. No significant differences were

found among the angles of torsion at fracture for the various osteotomies or

the intact bone.

Teixeira et al. treated tibial segmental defects of 35 mm size in both male

and female sheep aged four to five months. Considering the age of the

animals and the preservation of the periosteum, the critical size of this defect

can be questioned and results cannot necessarily be extrapolated to adult

humans, as described correctly by the authors. An empty control group was

not included in the experiment. The bone defects in the diaphysis of the right

hind limb were stabilized with a titanium bone plate (103 mm in length, 2 mm

thickness, and 10 mm width) combined with a titanium cage. As reported by

the authors, plate bending occurred in 42% of the animals and was partly

attributed to the connection of the titanium cage to the plate. However, it

appears that the bending of the plate was rather a result of insufficient

thickness of the fixation device. The titanium cages were either filled with

autologous cortical bone graft or with a composite biomaterial consistent of

inorganic bovine bone, demineralised bovine bone, a pool of bovine bone

morphogenetic proteins bound to absorbable ultra-thin powdered

Page 53: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

29

hydroxyapatite and bone-derived denaturized collagen. Bone defect healing

was assessed clinically, radiographically and histologically. Titanium cages

might keep implanted scaffolds and biomaterials in place initially and

biomechanically support defect fixation, however, it must be taken into

consideration that – since titanium is not resorbable – the cages might hinder

complete bone remodelling in the long run.

Radiographic examination showed initial formation of periosteal callus in both

groups at osteotomy sites, over the plate or cage 15 days postoperatively. At

60 and 90 days callus remodelling occurred. Histological and morphometric

analysis 90 days post surgery showed that the quantity of implanted

materials still present were similar for both groups while the quantity of newly

formed bone was less (p=0.0048) in the cortical bone graft group occupying

51 +/- 3.46% and 62 +/- 6.26% of the cage space, respectively [99].

Recently, Liu et al. reported on the use of highly porous beta-TCP scaffolds

to repair goat tibial defects [12]. In this study, fifteen goats were randomly

assigned to one of three groups, and a 26 mm-long defect at the middle part

of the right tibia in each goat was created and stabilized using a circular

external fixator. In Group A, a porous beta-TCP ceramic cylinder seeded with

osteogenically induced autologous bone marrow stromal cells was implanted

in the defect of each animal. In Group B, the same beta-TCP ceramic

cylinder without any cells was placed in the defect. In Group C, the defect

was left untreated. In Group A, bony union could be observed by gross view,

X-ray and micro-computed tomography (!CT) detection, and histological

observation at 32 weeks post-implantation. The implanted beta-TCP

scaffolds were almost completely replaced by host bone. Bone mineral

Page 54: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

30

density in the repaired area of Group A was significantly higher than in Group

B, in which scant new bone was formed without complete resorption of the

beta-TCP after 32 weeks. Moreover, the tissue-engineered bone of Group A

had similar biomechanical properties as the contralateral tibia in terms of

bending strength and Young's modulus. In Group C, little or no new bone

was formed and non-union occurred, demonstrating the critical nature of the

defect.

To investigate the effect of chondroitin sulphate on bone remodelling and

regeneration, Schneiders et al. [100] created a 30 mm tibial mid-diaphyseal

defect site and reconstructed it using hydroxyapatite/collagen cement

cylinders. Defect stabilization was achieved by insertion of a universal tibial

nail (UTN, Synthes, Bochum). To insert the scaffold to the defect, the authors

had to use a second operative aditus mid-diaphyseally. The published data

suggest problems with defect fixation not only due to reported implant

failures but also to clearly evident signs of locking bolt loosening, poor

contact between bone and nail, and the proximal nail end extending into the

articular space. Moreover, it can be supposed that either the insertion of the

nail or undesired movement of the loosened nail has caused damage to the

implants. When interpreting the presented data, it also has to be taken into

account that obviously no fabrication method has been described to reliably

reproduce implants of corresponding geometrical shape.

Rozen et al. investigated whether blood-derived endothelial progenitor cells

promote bone regeneration once transplanted into an ovine, critical sized,

tibial defect [101]. Cells were isolated and expanded in vitro. 2 x 107 cells in

0.2 ml saline were transplanted two weeks after a 32 mm defect had been

Page 55: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

31

created (n=7). Defect fixation was achieved by a 4.5 mm stainless steel plate

with four screws each proximally and distally. In the control group (n=8) 0.2

ml saline were injected. Defect bridging was observed in six out of seven

animals in the experimental group. In the control group, five out of six defects

analysed via !CT showed discontinuous (two animals) or minute bridging

(three animals) as stated by the authors. No reference to the remaining three

animals of the control group was found throughout the manuscript.

Therefore, the critical nature of the defect has to be questioned. Not

resecting the periosteum and screw loosening as clearly evident in the

published x-ray images might have contributed to defect bridging in the

control group.

The regenerative capacity of xenogenic human and autologous ovine

mesenchymal progenitor cells was assessed by Niemeyer et al. in an ovine

critical-size defect model [102]. Human and ovine MSC from bone marrow,

were cultured on mineralized collagen and implanted into a 30 mm-long

sheep tibia bone defect (n=7). Unloaded mineralized collagen served as

control. The 30 mm mid-diaphyseal defects were fixed with a seven hole LC-

LCP (Synthes) and a carbon fibre reinforced poly-ether-ketone plate

(snakeplate, Isotec AG, Altstätten, Switzerland). Animals were kept in

suspending slings for eight weeks post surgery. Nevertheless, implant failure

occurred in one animal requiring immediate euthanasia. Wound healing

related problems were reported for another animal.

In the same study, bone healing was assessed up to 26 weeks. Presence of

human cells after xenogenic transplantation was analysed using human-

specific in situ hybridization. Radiology and histology demonstrated

Page 56: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

32

significantly better bone formation after transplantation of autologous ovine

MSC on mineralized collagen compared to unloaded matrices and to the

xenogenic treatment group. No local or systemic rejection reactions could be

observed after transplantation of human MSC although the presence of

human MSC could be demonstrated.

The rapid progression of bone graft research and the great number of novel

developments must be supported by systematic assessment based on

clinical practicability and experience, the knowledge of basic biological

principles, medical necessity, and commercial practicality. From the current

literature review, it can be concluded, that in the majority of the mentioned

studies, follow up periods, which in most cases don not exceed six months,

are not suitable to evaluate long-term effects of bone substitutes and

scaffolds on bone regeneration and remodelling, and to determine in vivo

resorption kinetics of the respective biomaterial. Variations in defect sizes

and methods of defect fixation as well as postoperative treatment and

management concepts make it difficult to compare studies and draw reliable

conclusions. The modifications of commercially available fixation devices and

supporting systems to prevent peak loads from acting on implants suggest

the occurrence of implant failures usually expected early after surgery. As a

result, most experimental settings do not reflect the actual clinical conditions

faced and impede the extrapolation of results.

Page 57: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

33

Summary

The reconstruction of large bone segments remains a significant clinical

problem. Large bone defects occur mainly as a result of extensive bone loss

due to pathological events such as trauma, inflammation, and surgical

treatment of tumours. Present therapeutic approaches include the application

of bone graft transplants (autografts, allografts, xenografts), as well as

implants made of different synthetic and natural biomaterials or segmental

bone transport. However, no existing therapy has been proven to be fully

satisfactory. As a result, a large number of research groups, that work on the

development of new bone grafting materials, carriers, growth factors, and

tissue engineered constructs for bone regeneration, are interested in

evaluating their concepts in reproducible large segmental defect models. The

optimization of cell-scaffold combinations and locally or systemically active

stimuli will remain a complex process characterized by a highly

interdependent set of variables with a large range of possible variations.

Consequently, these developments must be nurtured and evaluated by

clinical experience, knowledge of basic biological principles, medical

necessity, and commercial practicality. The area of bone tissue engineering,

which has its main focus on the development of bioactive materials, depends

on the use of animal models to evaluate both experimental and clinical

hypotheses. To tackle major bone tissue engineering problems, researchers

must rely on the functional assessment of biological and biomechanical

parameters of generated constructs. However, to allow comparison between

different studies and their outcomes, it is essential that animal models,

fixation devices, surgical procedures and methods of taking measurements

Page 58: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

34

are standardized to achieve the accumulation of a reliable data pool as a

base for further directions to orthopaedic and tissue engineering

developments.

Page 59: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

35

Mo

de

l a

pp

lica

tio

n

o

Stu

dy o

f n

orm

al fr

actu

re h

ea

ling

o

D

rug

de

live

ry t

o f

ractu

re s

ite

s

o

Eff

ects

of

-

Dru

gs

-

Gro

wth

ho

rmo

ne

s

-

An

gio

ge

nic

fa

cto

rs

-

LA

SE

R

on

fra

ctu

re h

ea

ling

o

E

ffe

ct

of

fixa

tio

n m

eth

od

s o

n p

eri

oste

al, c

ort

ica

l a

nd

so

ft t

issu

e b

loo

d f

low

[1

03

] o

E

ffe

ct

of

typ

e a

nd

rig

idity o

f fixa

tio

n o

n f

ractu

re

he

alin

g a

nd

ra

te o

f re

mo

de

llin

g

[10

4]

o

Cre

atio

n o

f n

on

-un

ion

s

[10

5]

o

Cre

atio

n o

f a

n in

fecte

d b

alli

stic w

ou

nd

mo

de

l o

E

va

lua

tio

n o

f va

rio

us a

sse

ssm

en

ts (

e.g

. x-r

ay)

of

fr

actu

re h

ea

ling

o

In

vitro

te

stin

g o

f sp

ina

l fr

actu

re f

ixa

tio

n s

yste

ms

o

Stu

dy o

f in

teg

ratio

n,

de

gra

da

tio

n a

nd

re

mo

de

llin

g

of

bo

ne

su

bstitu

tes

!

Bo

ne

gra

ftin

g

!

Au

tog

raft

!

Allo

gra

ft

!

Xe

no

gra

ft

!

Bio

ma

teri

als

of

na

tura

l a

nd

syn

the

tic o

rig

in

!

De

min

era

lise

d b

on

e m

atr

ix

!

Bio

ma

teri

als

[1

06

] 1

. H

yd

roxya

pa

tite

/tri

ca

lciu

m-p

ho

sp

ha

te

ce

ram

ics

2.

Po

lym

ers

3

. M

eta

ls

4.

Co

mp

osite

s

o

Bo

ne

su

bstitu

tes p

lus a

uto

ge

no

us b

on

e m

arr

ow

o

E

va

lua

tio

n o

f o

ste

og

en

ic p

ote

ntia

l o

f ce

ll-

se

ed

ed

co

mp

osite

im

pla

nts

an

d a

sse

ssm

en

t

of

oste

oin

du

ctive

pro

pe

rtie

s o

f g

row

th f

acto

rs

De

fect

fixa

tio

n

Inte

rna

l o

In

tra

me

du

llary

ro

d/p

in

o

Pla

te a

nd

scre

ws

o

Scre

ws

o

Ce

rcla

ge

wir

es

Ex

tern

al

o

Exte

rna

l fixa

tors

o

C

asts

o

S

plin

ts

No

fix

ati

on

M

ice

an

d r

ats

c

Inte

rna

l o

In

tra

me

du

llary

na

il/p

in

o

Pla

te a

nd

scre

ws

Ex

tern

al

o

Exte

rna

l fixa

tors

Me

tho

ds o

f d

efe

ct

form

atio

n

o

Ma

nu

ally

cre

ate

d f

ractu

res

b

o

Th

ree

-po

int

be

nd

ing

b

o

Gu

illo

tin

e-l

ike

ap

pa

ratu

sb

o

Oscill

atin

g s

aw

, h

igh

sp

ee

d

de

nta

l b

urr

, o

r scis

so

rsb

o

Ba

llistic in

jury

o

Oscill

atin

g s

aw

o

Gig

li’s w

ire

An

ima

l sp

ecie

s a

nd

de

fect

loca

tio

n

Do

ga

o

Fe

mu

r [1

07

] [2

4]

o

Tib

ia [7

2]

[70

] [7

1]

[73

] [7

4]

[76

] [7

7]

[78

] o

R

ad

ius [

10

8]

[10

9]

[11

0]

[11

1]

S

he

ep

a

o

Tib

ia

[89

] [6

8]

[85

] [

79

, 1

12

] [8

0]

[81

] [8

8]

[69

] [1

13

] G

oa

t o

T

ibia

[1

14

] [9

8]

[11

5]

Pig

o

P

elv

is [

11

6]

o

Fe

mu

r [1

17

] o

T

ibia

[1

18

] [1

19

] o

S

pin

e [

12

0]

[12

1]

Do

ga

o

Fe

mu

r [1

07

] [2

4]

o

Tib

ia [7

2]

[70

] [7

1]

[73

] [7

4]

[76

] [7

7]

[78

] o

R

ad

ius [

10

8]

[10

9]

[11

0]

[11

1]

S

he

ep

a

o

Tib

ia

[89

] [6

8]

[85

] [

79

, 1

12

] [8

0]

[81

] [8

8]

[69

] [1

13

] G

oa

t o

T

ibia

[1

14

] [9

8]

[11

5]

Pig

o

P

elv

is [

11

6]

o

Fe

mu

r [1

17

] o

T

ibia

[1

18

] [1

19

] o

S

pin

e [

12

0]

[12

1]

a M

ost com

monly

used

b M

eth

ods to m

imic

accid

enta

l fr

actu

res m

ore

clo

sely

c In

radia

l, u

lnar

or

fibula

r fr

actu

res w

here

additio

nal bony s

upport

is p

resen

t

Ta

ble

4

An

ima

l M

od

els

of

Fra

ctu

res

(Oste

oto

my)

An

ima

l M

od

els

of

Se

gm

en

tal B

on

e

De

fects

(O

ste

cto

my)

Table 4: Comparison of animal models for fracture and segmental bone defect

research

Page 60: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

36

Ha

ve

rsia

n

ca

na

l

15

-50

18

-12

0

18

-70

27

-17

0

Dia

me

ter

(µm

) o

f [1

22

]

Ha

ve

rsia

n

sy

ste

m

12

5-1

75

75

-36

0

50

-32

5

18

0-3

25

Bo

ne

c

om

po

sit

ion

[4

2]

Bo

ne

m

ine

ral

de

nsity s

imila

r to

hu

ma

ns

Hig

he

r tr

ab

ecu

lar

bo

ne

de

nsity t

ha

n

hu

ma

ns

(0.6

1g

/cm

3)

Ve

ry s

imila

r to

h

um

an

Ve

ry s

imila

r to

h

um

an

Tra

be

cu

lar

bo

ne

d

en

sity:

0.4

3

g/c

m3

Bo

ne

re

mo

de

llin

gb

[42

]

10

0%

La

rge

r a

mo

un

t o

f b

on

e in

-g

row

th t

ha

n

hu

ma

ns

Sim

ilar

to

hu

ma

ns

Sim

ilar

to

hu

ma

ns

10

-15

%

to 4

0-5

5%

Ad

va

nta

ge

s

[42

] [4

4]

[54

]

o

Tra

ct

ab

le n

atu

re

o

Sim

ilar

bo

ne

min

era

l d

en

sity t

o

hu

ma

ns

o

Do

cile

a

nim

als

o

S

imila

r b

od

y w

eig

ht

to

hu

ma

ns

o

Dim

en

sio

n

of

lon

g b

on

es

su

ita

ble

fo

r h

um

an

im

pla

nts

Mo

re t

ole

ran

t to

am

bie

nt

co

nd

itio

ns

Bo

ne

min

era

l d

en

sity,

an

ato

my,

mo

rph

olo

gy,

an

d

he

alin

g s

imila

r to

h

um

an

s

Dis

ad

va

nta

ge

s

[40

] [5

4]

[12

3]

[42

]

o

Hig

he

r ra

te o

f so

lid

bo

ny f

usio

n w

he

n

co

mp

are

d t

o h

um

an

s

o

Lo

w n

on

-un

ion

ra

tes

o

Eth

ica

l is

su

es a

nd

n

eg

ative

pu

blic

p

erc

ep

tio

n

Sig

nific

an

t in

ter-

an

ima

l va

ria

tio

ns

du

e t

o b

ree

d

div

ers

ity

o

Ag

e-d

ep

en

da

nt

bo

ne

re

mo

de

llin

g

o

Ha

ve

rsia

n

rem

od

elli

ng

at

7-9

ye

ars

of

ag

e (

with

m

ed

ium

-siz

ed

, ir

reg

ula

r ca

na

ls)

Inq

uis

itiv

e a

nd

in

tera

ctive

na

ture

o

Hig

h g

row

th r

ate

s

an

d e

xce

ssiv

e b

od

y

we

igh

t o

D

ifficu

lt h

an

dlin

g

Ap

pli

ca

tio

n

[42

]

Mu

scu

loske

leta

l a

nd

de

nta

l re

se

arc

h

Ort

ho

pa

ed

ic

rese

arc

h

Re

se

arc

h o

n

ca

rtila

ge

, m

en

isci

an

d lig

am

en

tou

s

rep

air

Ort

ho

pa

ed

ic a

nd

d

en

tal stu

die

s

(fe

mo

ral h

ea

d

oste

on

ecro

sis

, fr

actu

res,

bo

ne

in

-g

row

th,

de

nta

l im

pla

nts

)

a N

um

be

r o

f H

ave

rsia

n s

yste

ms p

er

sq

ua

re m

illim

ete

r bA

ve

rag

e w

ho

le b

od

y t

rab

ecu

lar

bo

ne

tu

rno

ve

r p

er

ye

ar

Ma

cro

-str

uc

ture

[1

1]

[12

4]

[12

5]

Fe

mu

r:

Pro

no

un

ce

d c

urv

atu

re a

t d

ista

l th

ird

of

sh

aft

; n

arr

ow

in

th

e m

idd

le

Tib

ia:

Le

ng

th s

imila

r to

sh

ee

p;

pro

xim

ally

co

nve

x

me

dia

lly,

dis

tally

co

nve

x la

tera

lly;

pro

xim

al

$ p

rism

atic,

rem

ain

de

r cylin

dri

ca

l

Fe

mu

r:

Ro

un

de

d (

cylin

dri

ca

l) s

ha

ft;

co

nve

x

do

rsa

lly;

cu

rve

d in

dis

tal %

; re

gu

lar

in

dia

me

ter

Tib

ia:

Ma

jor

we

igh

t-b

ea

rin

g b

on

e o

f cru

s;

lon

g

an

d s

len

de

r; s

ha

ft c

urv

ed

me

dia

lly a

nd

ca

ud

ally

at

ce

ntr

e;

rou

nd

in

mid

dle

, tr

ian

gu

lar

pro

xim

ally

, fla

tte

ne

d c

ran

io-

ca

ud

ally

in

dis

tal th

ird

; m

ed

ial su

rfa

ce

is

su

bcu

tan

eo

us

Alm

ost

ide

ntica

l to

sh

ee

p

Fe

mu

r:

Re

lative

ly w

ide

& m

assiv

e d

iap

hysis

with

4

su

rfa

ce

s

Tib

ia:

Slig

htly c

urv

ed

dia

ph

ysis

, co

nve

x m

ed

ially

o

Ep

iph

ysis

- p

roxim

al a

nd

dis

tal-

sp

on

gy b

on

e

o

Me

tap

hysis

- tr

an

sitio

n z

on

e-s

po

ng

y

bo

ne

dis

tal to

ep

iph

yse

al lin

e

o

Dia

ph

ysis

- s

ha

ft o

f th

e b

on

e –

co

mp

act

bo

ne

Wo

ve

n-f

ibe

red

bo

ne

(p

lexifo

rm b

on

e)

Se

co

nd

ary

oste

on

s (

with

sm

all

ca

na

ls)

incre

ase

in

n

um

be

r w

ith

ag

e

Pri

ma

ry b

on

e s

tru

ctu

re

(ple

xifo

rm c

ort

ica

l b

on

e)

in

yo

un

g s

he

ep

(3

-4 y

ea

rs o

f a

ge

)

No

n-h

om

og

en

eo

usly

d

istr

ibu

ted

Ha

ve

rsia

n

syste

ms

Ple

xifo

rm b

on

e (

oste

on

al

ba

nd

ing

) D

en

se

Ha

ve

rsia

n b

on

e

(with

me

diu

m c

an

als

),

incre

ase

s w

ith

ag

e

Ma

inly

cir

cu

mfe

ren

tia

l la

me

llar

bo

ne

Wo

ve

n-f

ibe

red

bo

ne

(ple

xifo

rm)

form

ed

on

ly in

ra

pid

bo

ne

re

pa

ir a

nd

rem

od

elli

ng

Mo

re H

ave

rsia

n b

on

e t

ha

n

q

ua

dru

pe

ds (

12

.87

vs.

5.5

a)

Ta

ble

5

M

icro

-str

uc

ture

[4

0]

[61

] [1

22

, 1

23

]

Do

g

Sh

ee

p

Go

at

Pig

Hu

ma

n

Table 5: Summary of human and large animal bone properties

Page 61: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

37

Chapter II

Ovine bone and marrow derived progenitor cells: Isolation,

Characterization, and Osteogenic Potential

Page 62: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

38

Page 63: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

39

Introduction

In general, bone displays a high intrinsic regenerative capacity following

insult or disease. Therefore, the majority of bone defects and fractures heal

spontaneously, stimulated by well orchestrated endogenous cell populations

and micro-environmental cues. Improvements in surgical techniques, implant

design and peri-operative management have significantly improved

treatment outcomes of complex fractures and other skeletal defects resulting

from high energy trauma, disease, developmental deformity, revision

surgery, and tumour resection [1-6]. However, a compromised wound

environment, insufficient surgical technique or biomechanical instability can

lead to formation of large defects with limited regeneration potential [7]. Such

defects pose a major surgical, socio-economical and research challenge and

can significantly influence patients’ quality of life [8, 9]. Over the years, bone

grafts have advanced as the “gold standard” treatment for bone

augmentation [1, 2, 10-16]. However, the use of autologous bone is

associated with additional anaesthetic time and personnel required for graft

harvesting [12, 14, 17]. Often, insufficient amounts of graft can be obtained

while access to donor sites is limited [12, 13, 18, 19]. Donor site pain, nerve

damage or haemorrhage can occur while donor bone is predispositioned to

failure [4, 12, 13, 20]. Synthetic and naturally derived bone graft substitutes

have been investigated thoroughly during the past decades to address these

limitations. This research has brought about the rise of tissue engineering as

an important alternative approach to bone related orthopaedic and trauma

research. Orthopaedic research often necessitates the use of animal models.

Therefore, a number of large animal models has been developed to

Page 64: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

40

biomechanically simulate human in vivo conditions as closely as possible,

and to assess the effects of implanted bone grafts and tissue engineered

constructs on segmental long bone defect regeneration. In particular, mature

sheep are considered a valuable model for human bone turnover and

remodelling activity since animals of seven to nine years of age show similar

bone structure and composition, possess a bodyweight comparable to adult

humans, and long bone dimensions enabling the use of human implants [34,

40, 42, 54]. However, published fracture and segmental defect models have

often used considerably younger animals [126]. Whilst, sheep are a well

recognized animal model for bone related research, the molecular and

cellular events surrounding fracture and bone defect healing remain poorly

understood with respect to the recruitment and differentiation of osteogenic

precursor cell populations.

In the present study it was hypothesized that ovine marrow derived cells of

seven to eight year old sheep are equivalent to those previously described

for adult humans. The aims were to isolate, characterize and compare

populations of marrow cells derived from the iliac crest with cells from

compact bone. Initial characterization of cell properties, phenotype and

genotype was performed using techniques already established for human

mesenchymal progenitor cells (MPC). The potential of ex vivo expanded

ovine marrow and bone derived cells to produce tissues with properties

consistent with those of mature bone was further assessed in vitro and in

vivo. The purpose of this study was to provide a detailed characterisation of

the ovine segmental bone defect model on a cellular level.

Page 65: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

41

Materials and Methods

Isolation of ovine MPC and OB

Ovine osteoblast (OB) explants were obtained from 6-7 year old Merino

sheep undergoing experimental surgery as approved by the animal ethics

committee of the Queensland University of Technology, Brisbane, Australia

(ethics number 0700000915). Compact tibial bone samples were collected

under sterile conditions, minced, washed with PBS (Invitrogen) and vortexed

prior to incubation with 10 ml 0.25% trypsin/EDTA (Invitrogen) for 3 min at

37°C, 5% CO2. After trypsin inactivation with 10 ml low glucose Dulbecco's

Modified Eagle Media (DMEM) containing 10% foetal bovine serum (FBS)

(Invitrogen), samples were washed once with PBS and transferred to 175

cm2 tissue culture flasks (Nunc). Samples were topped-up with 12 ml of

DMEM containing 10% FBS and 1% penicillin/streptomycin. Osteoblast

outgrowth could be observed after 5-7 days (Fig. 1). Cells were expanded to

the second or third passage for subsequent experiments.

Bone marrow aspirates were obtained from the iliac crest under general

anaesthesia. Total bone marrow cells (0.5-1.5 x 107 cells/ml) were plated at

a density of 1-2 x 107 cells/cm2 in complete medium comprising low glucose

DMEM supplemented with 10% FBS, 100 U/ml penicillin and 100 !g/ml

streptomycin. Cells were subsequently plated at a density of 103 cells/cm2.

Page 66: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

42

Fig. 1: Cultures of MPC isolated from ovine bone marrow and OB derived from tibial

compact bone (&) after 3, 7 and 14 days in culture. Magnification 10x.

Flow cytometric analysis

Ex vivo expanded populations were used at passage three of culture for

immunophenotypic analysis. Ex vivo expanded MPC or OB were treated with

trypsin/EDTA and resuspended in blocking buffer for 30 min. Individual tubes

containing 1 x 105 cells were incubated with murine monoclonal IgG

antibodies reactive to either ovine CD14, CD31, and CD45 (Serotec), ovine

and human CD29, CD44 (Clone H9H11; Division of Haematology, IMVS,

Adelaide, SA, Australia), and CD166 (BD Biosciences), or isotype matched

controls, 1B5 (IgG1), and 1A6.11 (IgG2b) at a concentration of 10 !g/ml for 1

h on ice. After washing, cells were incubated with secondary detection

reagents, goat anti-mouse IgG-FITC or IgM-FITC conjugated antibodies

(1:50; Southern Biotechnology Associates, Inc., Birmingham, AL) for 45 min

on ice. Following washing, samples were analysed using a Cytomics FC 500

flow cytometry system (Beckman Coulter).

Page 67: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

43

Cell proliferation assay

Adherent passage three MPC and OB were seeded in triplicates at 3000/cm2

in flat bottomed 24-well plates (Nunc) and maintained in 1 ml standard

culture medium consisting of low glucose DMEM supplemented with 10%

FBS for 1, 3, 5 or 7 days in a humidified atmosphere (37°C, 5% CO2). At

each time point, cells were washed twice with PBS and stored at -80°C until

analysis. For analysis, samples were digested overnight with 0.5 mg/ml

proteinase K in 1 x Tris-EDTA buffer at 55°C. DNA content for 100 µl of each

sample in triplicate was measured and quantified using a Quant-iT

PicoGreen dsDNA assay kit according to the protocol supplied by the

manufacturer (Invitrogen). An equal volume of the Quant-iT PicoGreen

aqueous working solution was added to each triplicate and incubated for 3

min on a rocking plate. Fluorescence was measured with a Polar Star

Optima plate reader (BMG Labtech, Offenburg, Germany) at an excitation

wavelength of 485 nm and an emission wavelength of 520 nm.

CFU-F clonogenic assay

CFU-F assays were performed as described previously [127] [128]. Briefly,

passage two sheep osteoblasts and bone marrow derived, mesenchymal

progenitor cells were plated at a density of 0.25 x 104 in six-well plates. Cells

were maintained in low glucose DMEM supplemented with 100 U/ml

penicillin G, 100 µg/ml streptomycin, and 10% or 20% FBS respectively, at

37°C with 5% CO2 for 6 days. To enumerate colonies, cultures were washed

with PBS, fixed in ice cold methanol for 15 min, and stained with 0.05% w/v

crystal violet in dH2O for 15 min. Stained aggregates of greater than 50 cells

Page 68: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

44

were scored as CFU-colonies under a light microscope (TS100-U, Nikon,

Melville, NY).

2D differentiation in vitro

Passage three MPC and OB were seeded in triplicate into 6-well plates

(Nunc) at a density of 3000 cells/cm2 and expanded in low glucose

DMEM/10% FBS (Invitrogen) until confluent. Osteogenic induction was then

performed over the following 28 days using DMEM/10% FBS supplemented

with 50 µg/ml ascorbate-2-phosphate, 10 mM "-glycerophosphate, 0.1 µM

dexamethasone (Sigma-Aldrich). Controls were cultured in standard

expansion medium (DMEM/10% FBS).

Dynamic cell culture

For dynamic cell culture, MPC and OB were seeded into 6-well plates (Nunc,

Rochester, NY) at a density of 3000 cells/cm2 in triplicates and expanded in

low glucose DMEM/10% FBS (Invitrogen, Carlsbad, CA) until confluent.

MPCs were then differentiated osteogenically with low glucose DMEM/10%

FBS or DMEM/20% FBS each supplemented with 50 µg/ml ascorbate-2-

phosphate, 10 mM "-glycerophosphate, 0.1 µM dexamethasone both under

standard static and dynamic culture conditions on a rocking plate (Bioline

platform rocker, Edwards Instruments, Narellan, Australia; f=0.125 Hz).

Page 69: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

45

Alkaline phosphatase activity

At day 14 and 28, ALP enzyme activity was quantified using a colorimetric

assay. Triplicates were washed with PBS, incubated with 0.1% Triton X in

0.2 M Tris buffer at -20°C for 10 min. Cells were harvested and centrifuged at

10000 rpm for 10 min at 4°C and 100 µl of the cell extraction supernatant

was incubated with 125 µl p-Nitrophenylphosphate (1mg/ml) in 0.2 M Tris

buffer (Sigma-Aldrich) in 96 well plates (Nunc) and OD was measured after

30 min at 405 nm in a Polar Star Optima plate reader. ALP activity was

normalized against the sample DNA content determined using a Quant-iT

PicoGreen dsDNA assay kit (Invitrogen).

Alizarin red staining

To determine matrix mineralization, at day 14 and 28, triplicate samples were

washed twice with PBS and fixed with ice cold methanol for 10 min at room

temperature. Samples were then washed twice with ddH2O and incubated

with 1% alizarin red s (Sigma-Aldrich) in ddH2O, pH 4.1 for 10 min with

gentle shaking. After aspiration of the unincorporated dye, samples were

washed three times with ddH2O and air dried. Stained monolayers were

documented using inverted phase microscopy (TS100-U, Nikon, Melville,

NY). For quantification of staining, 800 µl 10% (v/v) acetic acid was added to

each well, and the plate incubated at room temperature for 30 min with

shaking. The monolayer was then scraped from the plate with a cell scraper

and transferred with 10% (v/v) acetic acid to a 1.5 ml microcentrifuge tube.

After vortexing for 30 s, the slurry was overlaid with 500 µl mineral oil

(Sigma-Aldrich), heated to 85°C for 10 min, and transferred to ice for 5 min.

Page 70: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

46

The slurry was then centrifuged at 20,000 x g for 15 min and 500 µl of the

supernatant was removed to a new 1.5 ml microcentrifuge tube. Then 200 µl

of 10% (v/v) ammonium hydroxide were added to neutralize the acid.

Aliquots (150 µl) of the supernatant were read in triplicate at 405 nm in 96-

well format using opaque-walled, transparent-bottomed plates (Nunc).

Obtained values were normalized against the DNA content of separate

samples since treatment with acetic acid was expected to cause

denaturation of DNA.

Wako HRII calcium assay

To further analyse the calcium contents in extracellular matrices produced by

ovine MPC or OB, a calcium assay was performed as per the manufacturer’s

instructions (Wako). Briefly, samples were washed with ddH2O and

incubated with 800 !l of 10% acetic acid at room temperature for 30 min.

Monolayers were scraped off, heated to 85˚C for 10 min, transferred to ice

for 10 min, then 200 !l of 10% ammonium hydroxide was added to 500 !l of

each sample. Triplicate 10 !l aliquots were transferred into a 96 well plate, to

which 100 !l of a monoethanolamine buffer, pH 11.0, was added for a 3 min

incubation at 37˚C, followed by 100 !l of o-cresolphthalein complexion and

incubation for 5 min at 37˚C. To generate a standard curve, a dilution series

of Multichem calibrator A (Wako) in 10% acetic acid was used.

Measurements were taken at '=570 nm in a Polar Star Optima plate reader

(BMG Labtech).

Page 71: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

47

X-ray photoelectron spectroscopy

For X-ray photoelectron spectroscopy (XPS) cells were seeded on

thermanox coverslips and cultured under osteogenic conditions as described

above. Samples were fixed with glutaraldehyde and dehydrated. XPS

spectra were acquired on a Kratos AXIS Ultra spectrophotometer operating

at a base pressure of 10-9mbar and equipped with a monochromatized Al K(

source. Acquisition was done with an analyser pass energy of 160eV on all

cell culture samples, with an energy of 20eV on native ovine bone and

thermanox coverslips. Samples were investigated with a charge

compensation gun (emission current, 0.15!A).

Immunohistochemistry

For immunohistochemistry, OB and MPC were cultured on Thermanox

coverslips to fit 24-well plates (Nunc). Media was removed; samples were

washed twice with PBS and fixed in 4% paraformaldehyde for 1 h on ice.

Cells were then permeabilized with 0.1% Triton X in PBS for 5 min and

quenched with 0.15 M glycine in PBS for 15 min (Sigma-Aldrich). Samples

were blocked with 1% BSA (Sigma-Aldrich) in PBS for 60 min and incubated

with primary mouse anti-human type I collagen (1:100) (MP Biomedicals,

Irvine, CA) and mouse anti-bovine osteocalcin (1:500) (Takara Bio Inc.,

Japan) antibodies in 1% BSA in PBS for 1 h at room temperature. Samples

were then washed three times with 0.1% BSA in PBS for 5 min each wash

and incubated with a FITC-conjugated goat anti-mouse secondary antibody

(Invitrogen) at a concentration of 1:200 for 30 min. Cover slips were then

Page 72: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

48

mounted on glass microscope slides and visualized using a fluorescent

microscope (TE2000-U, Nikon, Melville, NY).

Total RNA isolation, primer design and qRT-PCR

Total RNA was harvested from triplicate wells, from both differentiated and

control cells, on days 7, 14, 21 and 28. Cells were washed twice with PBS

and lysed in 1 ml Trizol reagent (Invitrogen) and RNA isolated following the

manufacturer’s instructions. cDNA was synthesized from 1!g of total RNA

using SuperScript III (Invitrogen) according to the manufacturer’s

instructions. Sheep specific oligonucleotides (Geneworks, TheBarton, SA,

Australia) were designed according to these parameters: 20-30 nt in length;

melting temperature 60ºC, +/- 2ºC; at least one primer spanning an exon

boundary; amplicon length 150 nt, +/- 50 nt; GC content between 40 and

60%; and at the 3’ end a C, G, CG or GC. In those cases where an Ovis

aries mRNA transcripts was not available, a blast search was perform on the

International Sheep Genome Consortium database

(https://isgcdata.agresearch.co.nz/) using the equivalent human mRNA

transcript. To date, the exon boundaries of sheep transcripts have not been

annotated; these boundaries were therefore baseded exon boundary

information for human and mouse mRNA transcripts. Quantitative RT-PCR

was performed on an Applied Biosystems 7900HT FAST Real Time PCR

system (Applied Biosystems, Scoresby, VIC, Australia) using a 384-well

plate layout; templates and reagents were aliquoted using an Eppendorf

5075 epMotion pippeting robot (Quantum Scientific, Murarrie, QLD,

Australia). The reaction volumes per well were as follows: 5 !l 2X SYBR

Page 73: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

49

Green (Roche, Castle Hill, NSW, Australia), 1 !l forward and reverse primers

at 1 !M final concentration, 1 !l water, 2 !l cDNA template diluted 1:10 from

stock. The thermo cycling conditions were as follows: 1 cycle of 10 min at

95°C for activation of the polymerase, 40 cycles of 10 sec at 95°C and 1 min

at 60°C for amplification. Dissociation curve analysis was carried out to verify

the absence of primer dimers and/or non-specific PCR products. The

expression of the genes of interest was normalized against the GAPDH

housekeeping gene.

3D cultures

Fused deposition modelling was used to fabricate circular mPCL-TCP

scaffolds of 5 mm diameter and 3 mm thickness. Type I rat tail collagen

(Vitrogen 100, Cohesion, Palo Alto, CA) was lyophilized into the pore space

forming a microporous mesh throughout the polymer (Fig. 2). For 3D

cultures, 120.000 ovine MPC or OB suspended in 60 !l of basal medium

were seeded onto each type I collagen coated mPCL-TCP scaffold and

placed in an incubator. After 1 h, 1 ml of medium was added to each 24-well.

Cell scaffold constructs were cultured in DMEM/20% FBS supplemented with

50 µg/ml ascorbate-2-phosphate, 10 mM beta-glycerophosphate, and 0.1 µM

dexamethasone on a rocking plate (f=0.125 Hz) for up to 4 weeks.

Page 74: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

50

Fig. 2: Medical grade polycaprolactone-tricalciumphosphate scaffold (left) coated

with rat tail collagen type I (right) of 5 mm in diameter and 3 mm thickness.

SEM

Cell scaffold constructs were fixed with 3% (v/v) glutaraldehyde in 0.1 M

sodium cacodylate buffer solution (pH 7.3) for 1 h at 4°C. Fixed specimens

were then dehydrated through a series of alcohols; two changes each of

50%, 70%, 90%, and 100% ethanol and were incubated for 10 min between

each change. Specimens were then critical point dried (Denton Vacuum,

Moorestown, NJ) and gold coated in a SC500, Bio-Rad sputter coater (Bio-

Rad) before examination using a FEI Quanta 200 scanning electron

microscope (FEI, Hillsboro, OR).

Confocal laser microscopy

To assess cell viability and morphology of MPC and OB seeded onto type I

collagen coated mPCL-TCP scaffolds, samples were stained with fluorescein

diacetate (FDA) and propidium iodide (PI)(Invitrogen) or rhodamine

conjugated phalloidin and 4',6-diamidino-2-phenylindole (DAPI)(Invitrogen).

For FDA-PI staining, samples were rinsed 3 times with PBS and incubated

with FDA staining solution (2 !g/ml) at 37°C for 15 min in the dark. FDA is a

cell-permeant esterase substrate which is hydrolysed by living/viable cells to

Page 75: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

51

give green fluorescence. Samples were then rinsed 3 times with PBS and

incubated with PI staining solution (20 !g/ml) at room temperature for 2 min

in the dark. PI is actively excluded by live cells thus dead cells in a

population are stained red. Samples were again rinsed 3 times with PBS and

visualised with a Leica SP5 confocal microscope (Leica Microsystems

GmbH, Wetzlar, Germany).

For phalloidin-DAPI staining, samples were fixed with 4% paraformaldehyde

for 20 min and permeabilized with 0.2% Triton X-100 in PBS for 20 min at

room temperature with gentle rocking. Samples were then washed twice for

5 min with 1 ml PBS at room temperature, and 700 !l rhodamine-conjugated

phalloidin (0.8 U/ml in 1% BSA in PBS) was added to each sample and

incubated for 1 h at room temperature with gentle rocking. Phalloidin binds to

F-actin of the cytoskeleton. Samples were then washed twice for 5 min with 1

ml PBS at room temperature and nuclei were stained with DAPI staining

solution (1.0 !g/ml in PBS) for 40-50 min at room temperature. Samples

were washed twice for 5 min with 1 ml PBS at room temperature and

visualised with a Leica SP5 confocal microscope.

In vivo transplantation studies

Ovine MPC and OB were seeded onto type I collagen coated mPCL-TCP

scaffolds at a density of 120.000 cells/scaffold and cultured for 4 weeks in

DMEM/20% FBS supplemented with 50 µg/ml ascorbate-2-phosphate, 10

mM "-glycerophosphate, and 0.1 µM dexamethasone on a rocking plate

(f=0.125 Hz). The cell scaffold constructs were then transplanted

subcutaneously into both left and right side pockets formed in the dorsal

Page 76: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

52

surface of 10-week-old immunocompromised, male NOD/SCID mice (ARC,

Perth, WA, Australia). Implants were recovered after 8 weeks and fixed in

4% paraformaldehyde.

!CT analysis

Micro CT analysis was performed on both the in vitro constructs and the in

vivo constructs. After 4 weeks of in vitro culture, constructs were carefully

removed from each well and inserted into polycarbonate sleeves for micro-

CT analysis. In vitro mineralization within the constructs was quantified using

a Micro-CT 40 scanner (Scanco Medical, Brüttisellen, Switzerland) at a voxel

size of 6 µm. Samples were evaluated at a threshold of 72, a filter width of

3.0 and filter support of 5.0. In vivo transplanted constructs were scanned at

a voxel size of 16 !m and were evaluated at a threshold of 140, a filter width

of 1.0 and filter support of 2. X-ray attenuation was correlated to sample

density using a standard curve generated by scanning hydroxyapatite

phantoms with known mineral density. Mineralized matrix volume or bone

volume fraction, and mineral density were quantified throughout the entire

construct.

Histology

For histological examination, specimens were fixed in 4% paraformaldehyde,

and dehydrated using an ethanol gradient (30 min in 70%, 1 h in 90%, 95%

and 100% ethanol). The samples were then processed through xylenes for

40 min three times, infiltrated with MMA for 3 h and embedded in MMA

containing 3% PEG. Seven micrometre sections were cut with an

Page 77: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

53

osteomicrotome (SM2500; Leica Microsystems, Wetzlar, Germany),

stretched with 70% ethanol onto a polylysine coated microscope slide (Lomb

Scientific), overlayed with a plastic film and slides were clamped together

before being dried for 12 h at 60°C. Sections were then stained using

combined von Kossa and van Gieson [129] stains to visualise the

mineralised bone and connective tissue respectively.

Image analysis

Histology sections were quantified using Image J software to quantify the

amount of mineralisation in a given area of section. Briefly, a JPEG image of

the entire tissue section was selected, converted to grayscale and a scale

bar was calibrated onto the image. The entire tissue section area was then

calculated by segmenting the entire tissue region from the background, and

then measuring the area. Next, only the mineralised (black) area was

segmented from the entire tissue area, and measured. The total mineralised

area was then calculated as a percentage of the total section area. Six

sections were analysed per sample group.

Statistical analysis

Statistical analysis was carried out using the student’s t test and p values <

0.05 were considered significant (SPSS, SPSS Inc., Chicago, Illinois, USA).

Page 78: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

54

Results

MPC show a higher proliferation rate than OB in vitro

Cells isolated from ovine bone marrow showed a significantly higher

proliferative potential after 1, 3 and 5 days in culture (p<0.05) when

compared to ovine tibial osteoblasts as represented by the higher DNA

content per 24-well for MPC (Fig. 3). After 6-7 days both MPC and OB

entered a plateau phase indicative for contact inhibition of cells reaching

confluency (p=0.0502).

Fig.3: Proliferative potential of MPC and OB determined by a PicoGreen assay.

When compared to OB, MPC displayed a significantly higher proliferative rate

before reaching confluency between day 5 and 7 (n=3). Values represent the mean

+/- standard deviation.

*

*

*

Page 79: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

55

MPC and OB exhibit a similar immunophenotype

Fluorescence-activated cell sorting (FACS) analysis was performed to

characterise the phenotype of ex vivo expanded ovine bone marrow derived

MPC and OB. The two cell populations exhibited similar expression patterns

for CD29 ("1-Integrin), CD44, CD166 (ALCAM) and CD14 (LPS-R). CD44

and CD166 have previously been identified as markers associated with

human bone marrow stromal, adipose, and dental pulp stem cells [127, 130-

132]. Importantly, both populations did not react with hematopoietic markers

CD45 (common leukocyte antigen) and CD31 (PECAM-1, endothelial) (Fig.

4).

Fig. 4: Surface antigen expression for MPC and OB as determined by FACS

analysis. The two cell populations exhibited similar expression patterns for CD29

and CD44. CD166 and CD14 was >50% MPC while only low levels were detected

on OB. Both populations did not react with the hematopoietic markers CD45 and

CD31.

Page 80: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

56

Clonogenic efficiency of MPC and OB

The mean frequency of CFU-F derived from marrow aspirates was 2.5 ± 0.5

per 0.25 x 104 mononuclear cells (MNC) in both DMEM/10% FBS and 20%

FBS, with the incidence of CFU-F forming MSC within the marrow MNC

population being approximately 0.08-0.12%. The mean frequency of CFU-F

derived from tibial bone explants was 2 ± 0.6 in DMEM/10% FBS and 1.8 ±

0.7 in DMEM/20% FCS respectively (incidence of colony forming cells 0.04-

0.12%)(Fig. 5).

Fig. 5: Clonogenic efficiency of passage two MPC and OB cultured under defined

media conditions.

2D differentiation potential of ovine MPC and OB in vitro

The potential of bone marrow derived MPC to differentiate into osteoblasts

and of bone derived osteoblasts to secrete a mineralised extracellular matrix

was investigated by culturing cells in the presence of L-ascorbic-2-

phosphate, dexamethasone, and "-glycerophosphate [132]. After 4 weeks of

induction, cultured MPC and OB had formed extensive amounts of alizarin

red-positive mineral deposits throughout the adherent layers. However, OB

consistently formed significantly fewer mineralized nodules (p<0.05)

Page 81: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

57

compared to bone marrow derived cells (Fig. 7, Fig. 6 B and H). Extracellular

matrix produced by both MPC and OB stained positive for type I collagen

and osteocalcin (Fig. 6 C-F and I-L). ALP activity measured at day 14 and

day 28 displayed a typical rise-fall pattern [133, 134] and was significantly

increased in osteogenically induced OB and MPC (p<0.05) when compared

to their respective controls (Fig. 8). Under osteogenic conditions a significant

increase in type I collagen expression could be observed over the course of

4 weeks for MPCs whilst no significant increase was found in the control

culture without osteogenic supplements (Fig. 9 A). Osteocalcin expression

was significantly up-regulated around day 14 and further increased towards

the end of week 4 (Fig. 9 C). A significant increase in osteopontin expression

could be detected at day 7 and 14. Osteopontin expression then decreased

towards day 21 and 28 (Fig. 9 E). For OB, no significant changes in type I

collagen expression were found (Fig. 9 B), only a small increase in

osteocalcin expression (Fig. 9 D). Osteopontin levels slightly increased

between day 0 and 7 to further stay on that level (Fig. 9 F).

Page 82: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

58

Fig. 6: Alizarin red staining and immunohistochemistry for osteocalcin (OC) and type

I collagen for MPC and OB cultures after 28 days on tissue culture polystyrene.

Under osteogenic conditions (ost), both MPC and OB were found to secrete a

mineralized matrix (B, H) that stained positive for the osteogenic marker protein OC

(D, J) and the extracellular matrix protein type I collagen (F, L). The control cultures

(ctrl) stained negative for both proteins. Magnification 10x.

Fig. 7: Quantification of the incorporated alizarin red s dye. After 28 days of culture

in osteogenic media, MPC cultures showed a significantly higher degree of

mineralization when compared to OB cultures or non-induced controls (ctrl)(n=3).

Error bars represent standard deviations.

Page 83: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

59

Fig. 8: Quantification of ALP activity within osteogenically induced (ost) and non-

induced (ctrl) OB and MPC cultures after 14 and 28 days. In osteogenically induced

MPC and OB cultures, ALP activity showed a typical rise and fall pattern and was

significantly higher than for the respective controls (n=3). Error bars represent

standard deviations.

Fig. 9: Quantitative RT-PCR for osteogenic markers. RT-PCR revealed significant

increases in type I collagen and osteocalcin expression over 4 weeks for MPC

under osteogenic conditions (MPC+) and an increase in osteopontin expression

between at day 7 and 14. For OB, no significant changes in type I collagen,

osteopontin and osteocalcin expression were found (n=3).

Page 84: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

60

Static vs. dynamic cultures

Culture systems that enhance mass transport and deliver controlled

mechanical stimuli have been shown to improve cell mediated extracellular

matrix synthesis [135]. To investigate the influence of mechanical stimuli on

ovine MPC and OB, cells were cultured over 14 days under static or dynamic

conditions with osteogenic media containing 10% or 20% FBS respectively. It

was found that that the combination of fluid shear forces in dynamic culture

and 20% FBS media content significantly increased ALP activity at day 7

(followed by a significant decrease towards day 14)(Fig. 10) and deposition

of mineralized matrix at day 14 (Fig. 11, 12) for both MPC and OB cultures.

Furthermore, the media content of 20% FBS significantly stimulated ALP

activity in MPC at day 14. When comparing the osteogenic OB and MPC

groups amongst each other, the increased degree of mineralization in the

group cultured with 20% FBS on the rocking plate appeared not be an effect

of increased cell number (p-values > 0.05, Fig. 15). XPS analysis for ovine

MPC and OB cultured on Thermanox coverslips for a period of 14 days

under dynamic osteogenic conditions with 20% FBS media content showed a

characteristic double peak for calcium in MPC cultures, considerably smaller

amounts of calcium were found for OB cultures (Fig. 13, 14).

Page 85: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

61

Fig. 10: Relative alkaline phosphatase (ALP) activity for ovine osteoblasts (OB) and

mesenchymal progenitor cells (MPC) under different culture conditions. The

combination of fluid shear forces in dynamic culture and 20% FBS media content

appeared to increase enzyme activity (n=3). Error bars represent standard

deviations.

Fig. 11: Quantification of the incorporated alizarin red s dye after 14 days showed a

significantly higher degree of mineralized extracellular matrix production for MPC

when compared to OP when subjected to a combination of fluid shear forces in

dynamic culture and 20% FBS media content (n=3). Error bars represent standard

deviations.

Page 86: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

62

Fig. 12: The quantification of calcium incorporated in the extracellular matrix after 14

days using a Wako HR II calcium assay confirmed a significantly higher amount of

calcium for osteogenically induced MPC under dynamic culture conditions with 20%

FBS media content. The average calcium content for MPC was 0.024 mg per 24-

well compare to 0.0037 mg for OB (p<0.05)(n=3). Error bars represent standard

deviations.

Page 87: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

63

Fig. 13: XPS analysis for native ovine bone derived from the mid diaphysis of the

tibia showing the presence of calcium with a characteristic double peak and

phosphate (A). The XPS analysis of plain Thermanox coverslips (B) as a negative

control confirmed the absence of both calcium and phosphate.

A

B

Page 88: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

64

Fig. 14: XPS analysis for ovine MPC (A) and OB (B) cultured on Thermanox

coverslips for a period of 14 days under dynamic osteogenic conditions with 20%

FBS media content. A characteristic double peak for calcium could be detected for

MPC cultures (A), considerably smaller amounts of calcium were found for OB

cultures (B).

A

B

Page 89: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

65

Fig. 15: PicoGreen assays for ovine MPC and OB on day 0, 1, 3, 5, and 7 after

osteogenic induction revealed that increased degrees of mineralization in the group

cultured with 20% FBS on a rocking plate (n=3) appeared not be an effect of

increased cell number but rather increased metabolic activity (p>0.05). Error bars

represent standard deviations.

3D differentiation potential of MPC and OB in vitro

Viability, morphology and osteogenic potential of ovine MPC and OB in a

three dimensional environment was assessed by FDA/PI staining, phalloidin-

DAPI staining, SEM, and micro computed tomography. After 28 days of

Page 90: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

66

osteogenic induction under dynamic conditions, cell viability was assessed

>90% (Fig. 16 A and D). Phalloidin-DAPI staining (Fig. 16 B and E) and SEM

analysis (Fig. 16 C and F) revealed elongated, spindle-shaped, osteoblast-

like cell morphology for both OB and MPC. However, MPC seemed to have

proliferated at a higher rate on the type I collagen coated mPCL-TCP

scaffolds forming a dense, interconnected three dimensional network. Micro

CT analysis displayed mineral deposition throughout the entire thickness of

OB and MPC-constructs, compared to control constructs (Fig. 17 A-F).

Scaffolds seeded with MPC showed a significantly higher mineral volume

fraction (MVF) compared to scaffolds seeded with OB (p=0.000197) (Fig. 17

G) while no significant difference in mineral density could be found between

MPC and OB-constructs (Fig. 17 H).

Fig. 16: SEM, live-dead and phalloidin-DAPI staining (confocal laser microscopy) of

MSC and OB on mPCL-TCP scaffolds cultured for 28 days. Cell viability was

assessed >90% for both cell types (A and D). Phalloidin-DAPI staining (B and E)

and SEM analysis (C and F) revealed elongated, spindle-shaped, osteoblast-like

cell morphology for both OB and MPC forming a dense, interconnected, three

dimensional network

Page 91: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

67

Fig. 17: !CT analysis of 3D in vitro cultures. !CT displayed mineral deposition

throughout the entire thickness of all constructs. After 28 days, medical grade PCL-

TCP scaffolds seeded with MPC (C, D) showed a significantly higher mineral

volume fraction (MVF) (G) compared to OB seeded (E, F) or cell free scaffolds (A,

B). No significant difference in mineral density was found (H)(n=5). Error bars

represent standard deviations.

Differentiation potential of MPC and OB in vivo

The developmental potential of culture-expanded, ovine MPC and OB was

assessed in vivo following transplantation into NOD/SCID mice in association

with type I collagen coated mPCL-TCP scaffolds. Transplants were

recovered after eight weeks, subjected to micro CT analysis and then

processed for histology. Micro CT analysis revealed a significantly higher

degree of ectopic bone formation for the scaffolds seeded with OB prior to

implantation (Bone volume fraction: 20.15%) when compared to MPC

(6.12%) or the respective cell free controls (0.55 %) (Fig. 18 A-C, M).

However, no significant difference could be found with regard to the mineral

density of newly formed bone matrix (Fig. 18 N).

Page 92: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

68

Histological examination of ectopic explants, using von Kossa staining

revealed no mineralisation for the control (no cells) constructs (Fig. 18 D, G,

J) whereas both MPC (Fig. 18 E, H, K) and OB (Fig. 18 F, I, L) seeded

mPCL-TCP scaffolds formed extensive ectopic bone within the implants,

over a course of 8 weeks in vivo. Mineral nodules containing calcium, stain

black with the von Kossa staining by virtue of silver ions (positive charge)

binding with the mineralised tissue (negative portion of the calcium salt)

forming a silver salt which is black in colour. The amount of ectopic bone

formed was significantly higher for OB seeded tissue engineered constructs

compared with MPC-seeded constructs (Fig. 18 O)(p<0.05). Residual mPCL-

TCP scaffold was evident within all transplants (Fig. 18 D) evidenced by

voids in the tissue from longitudinal and transverse sectioning of the scaffold

struts. The infiltration of haematopoietic cells together with associated

adipose elements was reminiscent of native bone marrow (Fig. 18). The

formation of different tissue types within the transplanted constructs included

mineralised bone (mb), fat (f) and fibrous connective tissue (c) with clear

blood vessel (bv) formation. The predominantly formed tissue type in both

MSC and OB samples, however, was bone with mature osteocytes enclosed

in characteristic lacunae surrounded by the bone extracellular matrix.

Page 93: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

69

Fig. 18: Histology and !CT of in vivo specimens 8 weeks post subcutaneous

transplantation into male NOD/SCID mice (n=4). !CT revealed significantly more

bone formation for OB compared with MPC (B, C) and control (A, M). No difference

in mineral density was observed (N). Combined von Kossa/van Gieson staining on

histological sections revealed extensive bone formation for MPC (E, H, K) and OB

seeded scaffolds (F, I, L), other tissue types included muscle (m) fat (f), blood

vessels (bv) and fibrous connective tissue (c). Error bars represent standard

deviations.

Page 94: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

70

Discussion

In translational orthopaedic research, the utilisation of large preclinical animal

models is a conditio sine qua non. Animal models utilized in the field include

representations of normal fracture-healing, segmental bone defects, and

fracture non-unions [38]. The selection of a specific animal species as a

model system involves the consideration of a number of factors.

Physiological and pathophysiological analogies are essential in respect to

the scientific question under investigation. It must be manageable to operate

and observe a multiplicity of study objects over a relatively short period of

time [39-41]. Further selection criteria include costs for acquisition and care,

animal availability, acceptability to society, tolerance to captivity and ease of

housing [42]. Mature sheep and goats possess a bodyweight comparable to

adult humans and long bone dimensions enabling the use of human implants

[54]. The mechanical loading environment occurring in sheep is well

understood [55, 56]. Since no major differences in mineral composition [57]

are evident and both metabolic and bone remodelling rates are akin to

humans [58], sheep are considered a valid model for human bone turnover

and remodelling activity [59] and show comparable bone healing potential

and bone blood supply [62]. Sheep bone, however, represents human bone

physiology and anatomy much closer as animals reach an age of six to

seven years. Only at this age bone remodels from a so called plexiform bone

into bone showing secondary osteon formation, which is a hallmark of adult

human bone. Therefore, the characterisation of cell populations derived from

animals of this age is absolutely essential.

Page 95: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

71

As described previously, bone marrow aspirates collected from adult sheep

contain a proportion of CFU-F forming cells with an incidence and

morphology highly similar to human MSC [128, 132, 136-139]. The incidence

of ovine CFU-F exceeds those described for most other animals, but lies

within the normal range reported for human bone marrow CFU-F [128, 140-

143].

To date, there is limited information available on the cell surface

characteristics of ovine bone marrow derived MPC and bone derived OB.

This can mainly be attributed to the limited availability of antibodies specific

for and cross-reacting with equivalent sheep antigens [128, 140, 144].

However, to validate sheep as a model system for research on a cellular and

molecular level, and to provide insight into fundamental processes such as

haematopoiesis, cell migration and homing, injury repair, differentiation, and

proliferation, the availability of suitable antibodies plays a key role. The cell

surface expression profile of ovine MPC and OB showed high and uniform

levels of cell surface CD29, CD44 and CD166 which have previously been

identified as markers associated with human bone marrow stromal, adipose,

and dental pulp cells [127, 130-132]. Characteristic for ovine MPC and OB

cultures was the absence of expression of the endothelial associated

adhesion marker CD31 and haematopoietic marker CD45 consistent with

expression patterns previously described for human bone marrow derived

MSC [127, 128].

When compared to ovine OB isolated from compact tibial bone, the cells

isolated from the bone marrow showed a higher proliferative potential (Fig. 3)

indicative for immature progenitor cells. Both MPC and OB followed a normal

Page 96: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

72

growth curve reported for different human cell types consisting of a lag phase

followed by a log phase of exponential cell growth, ending with a plateau

phase in which the growth rate declined [145-148]. The steeper slope of the

MPC growth curve between day 1 and 3 after seeding results in a higher

density of cells before the rate of growth begins to decline. A high

proliferation capacity is desirable when it comes to the application of cell

based tissue engineering strategies in preclinical models since large cell

numbers generated over a relatively short period of time may be required for

various clinical applications.

After 28 days of culture under osteogenic conditions both MPC and OB were

shown to produce mineralized extracellular matrix positive for alizarin red,

osteocalcin and type I collagen. It was shown that MPC and OB can be

induced to form mineral in culture by treatment with osteogenic medium in

vitro. Osteogenic medium contains a source of phosphate, ascorbic acid, and

dexamethasone in a rich medium such as DMEM containing fetal bovine

serum (FBS) [149]. In a chelation process, alizarin red S forms complexes

with calcium and therefore allows simultaneous evaluation of mineral

distribution and inspection of fine structures by phase contrast microscopy. It

is particularly versatile in that the dye can be extracted from the stained

monolayer and readily assayed with low variability and a much wider linear

detection range than traditional calcium detection methods [150, 151].To

monitor inorganic phosphate deposition may be problematic due to the high

levels of contaminating phosphate associated with other components of the

cell and the high levels of free phosphate in the cytosol.

Page 97: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

73

The extracellular matrix produced by ovine MPC and OB was shown to stain

positive for type I collagen and osteocalcin. Type I collagen comprises

approximately 95% of the entire collagen content of bone and about 80% of

the total proteins present in bone [152, 153]. The increase in type I collagen

expression on the gene level in MPC and its presence in the extracellular

matrix deposited by MPC under osteogenic conditions is therefore consistent

with osteogenic differentiation processes. Osteocalcin is synthesized and

secreted by normal maturing osteoblasts. It is one of the major non-

collagenous bone matrix proteins, with osteocalcin comprising 1% to 2% of

the total proteins in the skeleton [154]. Osteocalcin binds with high affinity to

hydroxyapatite crystals, the key mineral component of bone, and regulates

bone crystal formation [155]. In the MPC cultures, osteocalcin expression

was up-regulated around day 21 and further increased towards the end of

week 4 which is consistent with osteocalcin being a late osteogenic marker.

Osteocalcin protein was additionally detected immunohistochemically in the

ECM produced by both ovine MPC and OB further suggesting a bone like

composition. Osteopontin is biosynthesized by a variety of tissue types

including pre-osteoblasts, osteoblasts, osteocytes, and bone marrow cells

and is considered an early osteogenic marker and was shown to be up-

regulated in the ovine MPC cultures early during differentiation. It has further

been implicated as an important factor in bone remodelling [156]. For the

OB, no significant changes in type I collagen expression were found, only a

small increase in osteocalcin expression. Osteopontin levels increased

between day 0 and 7 and remained at this level. The findings suggest that

while MPC undergo a differentiation process when supplemented with

Page 98: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

74

osteogenic factors, OB – as they are mature cells already – very much

maintain their gene expression pattern. Alkaline phosphatase (ALP) is

considered a marker for osteoblastic activity in vitro. ALP activity measured

at day 14 and 28 displayed a typical rise-fall pattern [133, 134] and was

significantly increased in osteogenically induced OB and MPC (p<0.05) when

compared to their respective controls (Fig. 9).

The inorganic, crystalline phase of native bone consists mainly of

hydroxyapatite, which includes calcium phosphate, calcium carbonate,

calcium fluoride, calcium hydroxide and citrate [157]. The presence of

calcium and phosphate in the matrices produced by MPC and OB as

confirmed by XPS, WAKO HRII calcium assay and alizarin red staining

suggests the deposition of a mineralized, bone-like matrix in vitro.

Culture systems that enhance mass transport and deliver controlled

mechanical stimuli have been shown to improve cell mediated extracellular

matrix synthesis in vitro [135] and it was shown that fluid shear stress can

synergistically enhance osteoblastic differentiation [158] [159]. In the present

study, for both ovine MPC and OB, it was found that the combination of fluid

shear forces in dynamic culture and 20 % FBS media content significantly

increased ALP activity at day 7 (Fig. 11) and deposition of mineralized matrix

at day 14 (Fig. 12, 13). MPC cultures seemed to be even more susceptible to

mechanical stimuli than OB. Interestingly, it was demonstrated that the

increased degree of mineralization was not a function of proliferation rather

than a result of stimulated metabolic cell activity with the higher FBS content

providing the increased demand for nutrients.

Page 99: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

75

Since the extrapolation from results in 2D to cell behaviour in 3D is rather

difficult, 3D in vitro cultures of ovine MPC and OB were established on

medical grade PCL-TCP scaffolds produced via fused deposition modelling.

Polymer–calcium phosphate composites confer favourable mechanical and

biochemical properties for bone tissue engineering, including strength

(ceramic phase), toughness and plasticity (polymer phase), more favourable

degradation and resorption kinetics, and graded mechanical stiffness [106,

160]. Results obtained from 3D in vitro culture confirmed the findings from

2D differentiation studies with MPC demonstrating a higher proliferative and

osteogenic potential (Fig. 17, 18). Micro CT analysis revealed no significant

difference in matrix mineral density suggesting that the MPC had undergone

an osteogenic differentiation process towards osteoblast like cells actively

secreting mineralized matrix.

Preliminary analysis of the in vivo osteogenic developmental potential of

ovine MPC and OB was undertaken by subcutaneous transplantation into

immune-compromised NOD/SCID mice. Both MPC and OB demonstrated

osteogenic potential upon transplantation with type I collagen coated mPCL-

TCP composite scaffolds, as indicated by the presence of extensive deposits

of ectopic bone (Fig. 19). The observation of ectopic bone, fibrous tissue,

and haematopoiesis is analogous with studies using these scaffolds seeded

with porcine bone marrow derived progenitor cells [160].

Page 100: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

76

Conclusion

In summary the detailed characterisation of possible cell candidates for bone

tissue engineering applications is of utmost importance. As mature sheep are

considered a valuable model for human bone turnover and remodelling,

experimental data obtained from cells derived from these animals are of

special interest. In the present study, ovine MPC isolated from bone marrow

aspirates and OB from cortical bone explants exhibited morphological,

immunophenotypical and multipotential characteristics similar to those in

humans. MPC reproducibly showed a higher osteogenic potential in vitro.

When transplanted subcutaneously, OB however displayed a higher

developmental potential in respect to bone formation. This underlines the

difficulties in extrapolating results obtained in vitro to an in vivo setting and

suggests that OB isolated from compact bone might represent a suitable

alternative cell population for tissue engineering applications.

Microenvironmental conditions in ectopic bone assays, however, again may

not be representative of specific cues transplanted cells would face in an

actual bone defect. Although the study represents an essential first step

towards the detailed characterization of ovine MPC and OB in translational

studies towards the establishment of preclinical in vivo models, further

studies are required to verify these findings in orthotopic models.

Page 101: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

77

Chapter III

Ovine Bone and Marrow derived Progenitor Cells and their Potential for

Scaffold Based Bone Tissue Engineering Applications in vivo

Page 102: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

78

Page 103: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

79

Introduction

The investigation of biological processes driving tissue regeneration is a key

step in order to optimize tissue engineering approaches aiming to correct

and repair severe tissue damage necessitating complex surgical procedures

including organ and tissue transplantation. Thus, a detailed understanding of

host responses to implanted donor cells would facilitate the development of

novel clinical treatment strategies. In bone tissue engineering, some

traditional approaches rely on the transplantation of osteogenic cells seeded

onto scaffolds mimicking chemical and physical properties of native tissue

[126, 161]. Therefore, a large number of different materials has been

investigated both in vitro and in vivo to evaluate material biocompatibility as

well as their osteoinductive and osteoconductive properties [162, 163]. In a

number of bone tissue engineering studies, mesenchymal stem cells (MSC)

and osteoblasts (OB) have been used in conjunction with these materials.

Both cell types exhibit distinct intrinsic osteogenic characteristics, which

make these cells suitable candidates for bone tissue engineering

applications. Furthermore, transplantation studies have demonstrated that

MSC and OB are capable of synthesizing bone-like extracellular matrix

(ECM) [164-166]. The processes of de novo bone formation in vivo are partly

attributed to members of the transforming growth factor-" (TGF-")

superfamily, specifically the bone morphogenetic proteins (BMPs). In

particular BMP-7 was shown to have not only osteoinductive but also

angiogenic effects [167, 168]. However, the molecular and cellular events

surrounding osteoneogenesis remain poorly understood with respect to

Page 104: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

80

neovascularisation, and the recruitment and differentiation of osteogenic

precursor cell populations.

In the present study, ovine MSC and OB were transplanted subcutaneously

into NOD/SCID mice combined with and without recombinant human BMP-7

(rhBMP-7). This was to assess the influence of donor cell commitment on

bone synthesis and to evaluate the contribution of host cells to

neovascularisation as well as formation and maturation of tissue engineered

bone. It was hypothesized that bone cell origin, ossification type, and degree

of vascularisation and bone neoformation is dependent on the nature and

commitment of transplanted cells as well as supplemented growth factors

such as rhBMP-7.

Page 105: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

81

Materials and Methods

Isolation of ovine MSC and OB

Ovine osteoblast explants were isolated from six to seven year old Merino

sheep as approved by the animal ethics committee of the Queensland

University of Technology, Brisbane, Australia (ethics number 0700000915).

Mid-diaphyseal, compact tibial bone samples were collected, minced, and

washed prior to incubation with 10 ml 0.25% trypsin/EDTA (Invitrogen) for 3

min at 37°C, 5% CO2. After trypsin inactivation with 10 ml low glucose

DMEM containing 10% FBS (Invitrogen), samples were washed with PBS

and transferred to 175 cm2 tissue culture flasks (Nunc). Samples were

topped-up with 12 ml of DMEM containing 10% FBS and 1%

penicillin/streptomycin. Osteoblast outgrowth occurred after 5-7 days. Cells

were expanded to the second or third passage for subsequent experiments.

Bone marrow aspirates were obtained from the iliac crest under general

anaesthesia. Total bone marrow cells (0.5-1.5 x 107 cells/ml) were plated at

a density of 1-2 x 107 cells/cm2 in complete medium comprising low glucose

DMEM supplemented with 10% FBS, 100 U/ml penicillin and 100 !g/ml

streptomycin. Cells were subsequently plated at a density of 103 cells/cm2.

Scaffold fabrication and cell seeding

Bioresorbable scaffolds of medical grade )-poly-caprolactone incorporating

20% "-tricalcium phosphate (mPCL–TCP) were produced by fused

deposition modelling (FDM) as described previously (Osteopore

International, Singapore; www.osteopore.com.sg) [169]. The structural

parameters of the scaffolds were tailored by computer aided design (CAD)

Page 106: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

82

and included 100% pore interconnectivity within a range of 350–500 µm size,

70% scaffold porosity, and a 0/90° lay down pattern. Using biopsy punches,

cylindrical scaffolds of an outer diameter of 8 mm, an inner diameter of 4 mm

and a height of 4 mm were produced. Prior to cell seeding, scaffolds were

surface treated with 1M NaOH for 6 h, washed five times with PBS, sterilized

under UV light, and transferred to 24-well plates (Nunc). Scaffolds were then

incubated with DMEM/20% FBS for 1 h before 250.000 ovine MSC or OB

suspended in 60 !l of basal medium were seeded onto each mPCL-TCP

scaffolds and placed in an incubator. After 1 h, 1 ml of medium was added to

each well. Cell scaffold constructs were cultured in DMEM/20% FBS

supplemented with 50 µg/ml ascorbate-2-phosphate, 10 mM beta-

glycerophosphate, and 0.1 µM dexamethasone on a rocking plate (f=0.125

Hz) for 4 weeks.

Cell sheet fabrication

For cell sheet fabrication, MSC and OB were seeded at a density of

3000/cm2 into 6-well plates and cultured in low glucose DMEM

supplemented with 10% FBS and 1% penicillin/streptomycin until confluent.

Cells were then cultured in osteogenic media consisting of DMEM/20% FBS

supplemented with 50 µg/ml ascorbate-2-phosphate, 10 mM beta-

glycerophosphate, and 0.1 µM dexamethasone for 4 weeks to allow

mineralization of the cell sheets. One day prior to transplantation, cell sheets

were carefully detached using a cell scraper and two sheets of matching cell

type were wrapped around each of the cell seeded scaffolds and cultured in

osteogenic media until transplantation.

Page 107: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

83

In vivo transplantation studies

Cell scaffold constructs were transplanted subcutaneously into both left and

right side pockets formed in the dorsal surface of 21 ten week old, male,

immune compromised NOD/SCID mice (ARC, Perth, WA, Australia).

Experimental groups included

1.) mPCL-TCP scaffold

2.) scaffold + fibrin glue

3.) scaffold + fibrin glue + rhBMP-7

4.) scaffold + oMSC

5.) scaffold + oMSC + fibrin glue + rhBMP-7

6.) scaffold + oOB

7.) scaffold + oOB + fibrin glue + rhBMP-7.

An amount of 50.5 !l fibrin glue (Tisseel, Baxter) was used to administer 5

!g of rhBMP-7 (Stryker) to the inner duct of the scaffolds intraoperatively.

Each group included six implants. For experimental group 4-7 additional four

constructs with BrdU labelled cells were transplanted.

Page 108: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

84

Fig. 1: The schematic illustrates the in vitro generation of tissue engineered

constructs (TEC) from primary cells and scaffolds as well as the method of rhBMP-7

administration at the time of transplantation.

BrdU labelling of cells

Cells for Bromodeoxyuridine (5-bromo-2-deoxyuridine, BrdU) labelling were

seeded at a density of 3000/cm2 and 250.000/scaffold in DMEM/10% FBS

and allowed to attach overnight. The day after seeding, BrdU labelling was

achieved by incubating ovine MSC or OB with the BrdU labelling reagent

(Invitrogen) at a concentration of 1:100 in DMEM/10% FBS for 5 h [170] (Fig.

2). BrdU is a synthetic nucleoside that is an analogue of thymidine. It can be

incorporated into newly synthesized DNA of replicating cells substituting for

thymidine during DNA replication thus labelling the respective cells. Specific

antibodies can then be used to visualize the incorporated chemical.

Specimens for transplantation were generated as described above,

recovered after 8 weeks, and fixed in 4% paraformaldehyde.

Page 109: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

85

Fig. 2: Monolayer of ovine bone marrow derived cells labelled with BrdU.

Bromodeoxyuridine (5-bromo-2-deoxyuridine, BrdU) is a synthetic nucleoside that is

an analogue of thymidine. BrdU can be incorporated into newly synthesized DNA of

replicating cells substituting for thymidine during DNA replication. Antibodies

specific for BrdU can then be used to detect the incorporated chemical (brown

nuclei, arrows). The cultures were counterstained with Haematoxylin to visualize the

nuclei of non-labelled cells (asterix, violet nuclei). Magnification: 4x (A) and 10x (B).

Biomechanical testing

To determine differences in mechanical properties resulting from new bone

formation, compression testing was performed on all six specimens per

group. During the testing period, samples were kept in phosphate-buffered

saline (PBS) at ambient conditions. Tests were performed on an Instron

5848 testing system with a 500 N load cell. The specimens were

compressed at a rate of 1 mm/min up to a strain level of approximately 5%.

The compression stiffness was calculated from the stress-strain curve as the

slope of the initial linear portion of the curve.

Page 110: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

86

!CT analysis

In vivo transplanted constructs were scanned at a voxel size of 16 !m (!CT

40, Scanco Medical AG, Brütisellen, Switzerland) and evaluated at a

threshold of 140, a filter width of 1.0 and filter support of 2. X-ray attenuation

was correlated to sample density using a standard curve generated by

scanning hydroxyapatite phantoms with known mineral density. Bone volume

fraction and mineral density were quantified throughout the entire construct.

Immunohistochemistry

For immunohistochemistry, three samples per group were fixed with 4%

paraformaldehyde, decalcified in 15% EDTA in Tris-HCl (pH 7.4) for 10 days

and embedded in paraffin [171]. Sections of 5 !m thickness were prepared,

deparaffinised and rehydrated. Subsequently, sections were rinsed in

distilled water and placed in 0.2 M Tris-HCl buffer (pH 7.4). The sections

were incubated with 2% bovine serum albumin (BSA) (Sigma, Sydney,

Australia) in DAKO antibody diluent (DAKO, Botany, Australia) in a

humidified chamber at room temperature for 20 min to block nonspecific

binding sites. Endogenous peroxidase was quenched by incubating the

sections in 3% H2O2 in Tris-HCl for 20 min. This was followed by three

washes with Tris buffer (pH 7.4) for 2 min each. Subsequently,

immunohistochemical staining was performed using a primary mouse

monoclonal antibody specific to osteocalcin (ab13418, Abcam Ltd.) and to

type II collagen (MS-235-P0, Invitrogen). Non-immunized mouse IgG

(086599, Invitrogen) was used as an isotype control to rule out non-specific

reactions of mouse IgG with ovine tissues as well as non-specific binding of

Page 111: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

87

the secondary antibodies and/or peroxidase labelled polymer to ovine

tissues.

The sections were incubated with the specific antibody or negative control at

room temperature in humidified chambers for 30 min with the primary

antibody at a concentration of 1:200 in DAKO antibody diluent. The sections

were washed three times for 2 min with Tris buffer (pH 7.4) and incubated

with peroxidase labelled dextran polymer conjugated to goat anti-mouse and

anti-rabbit immunoglobulins (DAKO EnVision+ Dual Link System Peroxidase,

DAKO) at room temperature in humidified chambers for 30 min. Colour was

developed using a liquid 3,3-diaminobenzidine (DAB) based system (DAKO).

Mayer’s haematoxylin was used as a counterstain, and Kaiser’s glycerol

gelatin (DAKO) for coverslip mounting.

Histochemistry

For histological examination half of the specimens of each group were fixed

in 4% paraformaldehyde, and dehydrated using an ethanol gradient (30 min

in 70%, 1 h in 90%, 95% and 100% ethanol). The samples were then

processed through xylenes for 40 min three times, infiltrated with MMA for 3

h and embedded in MMA containing 3% PEG. Seven micrometre sections

were cut with an osteomicrotome (SM2500; Leica Microsystems, Wetzlar,

Germany), stretched with 70% ethanol onto a poly-lysine coated microscope

slide (Lomb Scientific), and overlayed with a plastic film. Slides were then

clamped together before being dried for 12 h at 60°C. Sections were stained

using combined von Kossa and van Gieson [129] stains to visualise the

mineralised bone and connective tissue respectively.

Page 112: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

88

In addition, paraffin sections were stained for alcian blue/nuclear fast red and

haematoxylin/eosin using standard protocols [172, 173].

Quantification of Neovascularisation

Mallory’s trichrome (Sigma) stainings according to manufacturer’s

instructions were performed to reveal the presence of blood vessels [174].

Blood vessel formation was quantified by image analysis as described below

for mineralized tissue.

Tartrate resistant acid phosphatase (TRAP) staining

To detect osteoclast activity, TRAP staining was performed as described

previously by Erlebacher and Derynck [175]. Sections of 5-8 !m thickness

were deparaffinised in two changes of xylene, rehydrated through a graded

series of ethanols and placed in 0.2 M acetate buffer (0.2 M sodium acetate

and 50 mM L(+) tartartic acid in ddH2O, pH 5.0) and incubated for 20 min at

room temperature. Sections were then incubated with 0.5 mg/ml naphtol AS-

MX phosphate and 1.1 mg/ml fast red TR salt (both Sigma) in 0.2 M acetate

buffer for 1-4 h at 37°C until osteoclasts appeared bright read. Sections were

then counterstained with haematoxylin, air-dried and mounted.

Detection of BrdU-labelled cells

BrdU labelled specimens were fixed with 4% paraformaldehyde decalcified in

EDTA for 10 days and embedded in paraffin using an automated tissue

processor (Excelsior ES, Thermo Scientific). Sections of 3-4 !m thickness

were prepared, deparaffinised and rehydrated. For detection of BrdU labelled

Page 113: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

89

cells, a Zymed® streptavidin-biotin based system for BrdU staining

(Invitrogen) was used according to the manufacturer’s protocol.

Quantification of BrdU positive cells located within newly formed bone was

achieved as described in detail by Tatapudy et al. [176].

SEM and EDX

To confirm the presence of calcium and phosphate in areas histologically

identified as bone, scanning electron microscopy (SEM) in combination with

energy dispersive X-ray spectroscopy (EDX) was performed on 7 !m

sections of the PMMA embedded samples on a JEOL JSM 6300 Scanning

Electron Microscope (JEOL, Tokyo, Japan) [177, 178].

Image analysis

Histology sections were quantified using Image J software to determine the

amount of mineralisation in a given area of a section. Briefly, a JPEG image

of the entire tissue section stained for von Kossa/van Gieson was selected,

converted to greyscale, and a scale bar was calibrated onto the image. The

entire tissue section area was then calculated by segmenting the entire

tissue region from the background, and then measuring the area. Next, only

the mineralised (black) area was segmented from the entire tissue area, and

measured. The total mineralised area was then calculated as a percentage

of the total section area. Six sections were analysed per sample group.

To quantify the level of blood vessel formation low-magnification micrographs

of paraffin sections stained for Mallory’s trichrome were captured, and the

area covered by blood vessels was calculated using Image J and expressed

Page 114: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

90

as a percentage of the total tissue area [179]. Six sections were analysed per

sample group.

Statistical analysis

TStatistical analysis was carried out using the student’s t test and p values <

0.05 were considered significant (SPSS, SPSS Inc., Chicago, Illinois, USA).

Experiments were repeated three times, results are presented as mean

values +/- standard deviation.

Page 115: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

91

Results

Biomechanical testing

When compared to the cell free control groups or constructs including MSC,

compression testing at ambient conditions showed higher stiffness values for

mPCL-TCP scaffolds seeded with OB (58 N/mm, SD=3.52) and a significant

increase in compressive stiffness when rhBMP-7 was administered

intraoperatively to OB seeded scaffolds (71 N/mm, SD=5.8)(Fig. 3).

Fig. 3: Biomechanical testing was performed and compressive stiffness values

determined. The transplantation of osteoblasts (OB) or osteoblasts with rhBMP-7

(PCL-OB-BMP) led to increased stiffness when compared to controls or groups

including MSC (n=6).

!CT analysis

MicroCT analysis was performed to determine the bone volume fraction and

bone mineral density throughout the transplanted constructs. 3D

reconstructions of scanned specimens (Fig. 4) showed no bone formation in

the scaffold only (Fig. 4 A) or scaffold-fibrin (Fig. 4 B) group and only

scattered bone formation in scaffolds seeded with MSC (Fig. 4 C). More

Page 116: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

92

deposited bone was observed when scaffolds or MSC seeded scaffolds were

combined with rhBMP-7 (Fig. 4 D, E). However, the highest amount of bone

deposition was observed in OB seeded scaffolds (Fig. 4 F) and especially

when the OB seeded scaffolds were additionally stimulated with rhBMP-7

(Fig. 4 G). The visual, macroscopic impression based on the 3D

reconstructions was confirmed by quantitative analysis of the bone volume

fraction within the transplanted specimens. In the mPCL-OB-BMP group the

proportion of ectopic bone was found to be 17% (SD=3.04) followed by 12%

(SD=2.8) in the PCL-OB group and 2-5% in all other groups (Fig. 5 A). No

significant difference in bone mineral density was found between the different

groups (Fig. 5 B).

Fig. 4: 3D !CT reconstructions 8 weeks after subcutaneous transplantation into

NOD/SCID mice. Transplantation of the mPCL scaffold only (A) and scaffold with

fibrin glue (B) did not lead to ectopic bone formation. Only little bone formation was

found when the scaffold was seeded with MSC (C) and combined with rhBMP-7 (D).

The scaffold plus rhBMP-7 group showed slightly more bone formation (E) whilst the

highest amounts were seen in the groups including OB (F) or OB with rhBMP-7 (G).

Page 117: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

93

Fig. 5: Quantitative analysis of bone volume fraction (A) and bone mineral density

(B) 8 weeks after transplantation. The highest amounts of bone formation were

observed for scaffolds seeded with OB or OB seeded scaffolds combined with

rhBMP-7 (PCL-Fibrin-OB-BMP). No significant difference in bone mineral density

between the different groups was detected (n=6). The asterix indicates statistical

significance. Error bars represent standard deviations.

Histology

Eight weeks after transplantation into NOD/SCID mice, Haematoxylin and

Eosin (H&E) staining of paraffin sections revealed that residual mPCL-TCP

scaffold was evident within all transplants (Fig. 6 A-P, asterix) evidenced by

A

B

Page 118: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

94

voids within the tissue from longitudinal and transverse sectioning of the

scaffold struts. The formation of different tissue types within the transplanted

constructs included mineralised bone (NB), adipose tissue (AT) and fibrous

connective tissue (CT) with clear blood vessel (BV) formation. The

predominant tissue type in the scaffold only group represented fibrous

connective tissue (Fig. 6 A-D) with scattered adipose islets. Similar findings

were observed in the scaffold-fibrin group (Fig. 6 E-H) although a higher

proportion of adipose tissue was evident. In the scaffold-BMP group (Fig. 6 I-

L) islets of newly formed bone, mainly located at the periphery of the

scaffold, intermingled with the surrounding fibrous connective and adipose

tissue. Both scaffold-MSC and scaffold-MSC-rhBMP-7 group (Fig. 7 A-D and

I-L) showed primarily peripheral bone formation along the outer boundaries

of the scaffold also lining the inner duct while a higher degree of bone

formation was observed in the scaffold-MSC-BMP group. The main tissue

type in central regions of the scaffold was adipose with smaller proportions of

connective tissue.

The predominantly formed tissue type in both groups including transplanted

OB, however, was bone with mature osteocytes (arrows) embedded in

characteristic lacunae surrounded by the bone extracellular matrix. The bone

tissue was evenly distributed in central and peripheral regions of the scaffold.

However, larger amounts of newly formed bone could be observed when

osteoblasts were transplanted in combination with rhBMP-7.

Qualitative assessment of histological sections from each of the groups

showed a higher degree of neovascularisation with blood vessels of larger

diameters within all transplanted constructs including rhBMP-7.

Page 119: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

95

A combined von Kossa-van Gieson stain was performed on 8 !m sections of

PMMA embedded specimens to specifically visualise and quantify the

mineralised bone within the tissue engineered constructs. The stain principle

is based on a photochemical reaction in which silver ions react with

phosphate. The staining confirmed the results obtained by !CT analysis with

the significantly highest amount of mineralisation found in the PCL-OB-BMP

group (Fig. 8, 9 and 11). Interestingly, bone lining cells in the groups

including MSC were of round, hypertrophic appearance and reminiscent of

chondrocytes. Bone synthesizing cells in the two groups including OB,

however, were elongated, spindle-shaped and of fibroblast-like morphology

(Fig. 12).

Fig. 6: H&E staining on histological paraffin sections 8 weeks after transplantation

revealed no bone formation in the scaffold only (A-D) and scaffold-fibrin group (E-H)

and only a small amount, peripheral ectopic bone formation (NB) for the scaffold-

BMP group (I-L). Other tissue types included fat (AT), blood vessels (BV) and

fibrous connective tissue (CT). Residual mPCL-TCP (&) evidenced by voids within

the tissue. Scale bars: B, F, J: 200 !m; C, G, K: 100 !m; D, H, L: 50 !m.

Page 120: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

96

Fig. 7: After 8 weeks, H&E staining on paraffin sections revealed little, bone

formation in the scaffold-MSC group with large proportions of adipose tissue (AT)

(A-D). More ectopic, evenly distributed, new bone (NB) was observed in the

scaffold-OB group (E-H). With both MSC (I-L) and OB (M-P) the addition of rhBMP-

7 lead to increased deposition of mineralised bone. However, larger amounts of

bone with mature osteocytes (arrows) enclosed in characteristic lacunae were

observed in the scaffold-OB-BMP group while lower proportions of adipose (AT) and

connective tissue (CT) were evident. Residual mPCL-TCP (&) resulted in voids

within the tissue. Scale bars: B, F, J, N: 200 !m; C, G, K, O: 100 !m; D, H, L, P: 50

!m.

The vascularisation of an implant is one of the first responses of the host to a

graft and is of utmost importance for its survival and functionality. Therefore,

the blood vessel formation was quantified on histological sections. Image

analysis showed a higher amount of neovascularisation in all groups that

included the administration of rhBMP-7 (Fig. 10) independent of transplanted

cell type.

Page 121: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

97

Fig. 8: Sections of PMMA embedded cell free control specimens stained for von

Kossa/van Gieson. No bone formation was observed in the PCL and PCL-fibrin

group, only little in the PCL-BMP group. Besides bone (NB, black), identified tissue

types included muscle (M), connective tissue (CT), adipose tissue (AT), and blood

vessels (BV). Scale bars: B, F, J: 200 !m; C, G, K: 100 !m; D, H, L: 50 !m.

Fig. 9: Bar graph demonstrating the results of the histomorphometric analysis of

PMMA embedded sections stained for von Kossa-van Gieson. No mineralized

tissue was found in the scaffold only group (PCL, 1%) and scaffold-fibrin group

(PCL-Fibrin, 1%). No significant difference in bone formation was evident between

the scaffold-BMP (PCL-BMP, 17%), scaffold-MSC (PCL-MSC, 19) and scaffold-

MSC-BMP (PCL-MSC-BMP, 26%) groups. Osteoblasts transplanted in combination

with the PCL-scaffold (PCL-OB) formed significantly more ectopic bone (47%). The

addition of BMP to scaffold-OB constructs caused an even further increase in

deposition of mineralised extracellular matrix (PCL-OB-BMP, 67%). Error bars

represent standard deviations (n=6).

Page 122: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

98

Fig. 10: The figure illustrates the amount of neovascularisation within the constructs

8 weeks after transplantation. Blood vessels are expressed as a percentage of total

tissue area. The administration of rhBMP-7 appeared to stimulate blood vessel

formation. Error bars represent standard deviations.

Fig. 11: After 8 weeks, von Kossa-van Gieson staining on 8 !m PMMA sections

revealed little bone formation in the scaffold-MSC group and large amounts of

adipose tissue (AT) (A-D). More new ectopic bone was observed in the scaffold-OB

group (E-H). With both MSC (I-L) and OB (M-P) the addition of rhBMP-7 lead to

increased deposition of mineralised bone (NB). However, larger amounts of bone

were observed in the scaffold-OB-BMP group. On the other hand lower proportions

of adipose (AT) and connective tissue (CT) were evident. Residual mPCL-TCP (&)

resulted in voids within the tissue. Scale bars: B, F, J, N: 200 !m; C, G, K, O: 100

!m; D, H, L, P: 50 !m.

Page 123: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

99

Fig. 12: High magnification images of bone lining cells. The bone forming cells in

both groups including transplanted MSC (A, C) appear of round, hypertrophic

morphology (arrows) with big nuclei. Both in the scaffold-OB (B) and scaffold-OB-

BMP group (D), the cells immediately lining the ectopically formed bone (black)

were of elongated, spindle-like shape (arrows). Scale bars: 20 !m.

Osteocalcin, an extracellular matrix protein, is partly incorporated into the

bone matrix, partly delivered to the circulatory system. It is considered a late

and specific osteogenic marker determining terminal osteoblast

differentiation regulating bone crystal formation. Osteocalcin (OC) expression

was examined in all experimental groups in which new bone formation was

evident (Fig. 13). In all groups, strong extracellular, matrix associated and

cytoplasmatic OC expression was detected in osteocytes enclosed in the

Page 124: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

100

mature bone (arrow heads). Notably, bone lining osteoblasts (arrows) also

expressed high levels of OC.

Fig. 13: Immunohistochemical staining for osteocalcin (OC) within newly formed

bone (NB) counterstained with haematoxylin demonstrating high extracellular

and cytoplasmatic osteocalcin expression in both osteocytes enclosed in

extracellular matrix (arrow heads) and bone lining osteoblasts (arrows). Fibrous

connective tissue (CT) and adipose tissue (AT) did not stain positively for OC.

Only light, haematoxylin caused background staining is evident in the isotype

controls (C, F, I, L, O). Scale bars: left and right column 100 !m, middle column

50 !m.

Page 125: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

101

In general, direct and indirect (chondral) ossification processes can be

distinguished. Chondral ossification requires preformed cartilaginous tissue

which is subsequently replaced by bone. This cartilaginous tissue contains

high amounts of glycosaminoglycans. Alcian blue is a phthalocyanine dye

which stains acid mucopolysaccharides and glycosaminoglycans (GAGs)

that appear blue to bluish-green. Therefore, alcian blue staining was

performed on all groups in which bone formation was evident to evaluate

whether direct or indirect ossification had occurred in the transplanted

specimens (Fig. 14). In all groups GAGs could be detected (arrows) within

areas of newly deposited bone. While only little GAG remnants were evident

within the bony extracellular matrix in the scaffold-OB (Fig. 14 E and F) and

scaffold-OB-BMP (Fig. 14 I and J) group, larger amounts of GAGs were

found in the scaffold-MSC (Fig. 14 C and D) and especially scaffold-MSC-

BMP group (Fig. 14 G and H).

Type II collagen is considered a cartilage specific extracellular matrix protein.

To further confirm the presence of cartilaginous tissue in the transplanted

specimens, immunohistochemical staining was performed. Positive staining

could only be detected in the scaffold-MSC and scaffold MSC-BMP group

(Fig. 14, C and G).

Page 126: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

102

Fig. 14: Alcian blue staining of paraffin sections counterstained with nuclear fast

red 8 weeks after subcutaneous transplantation. The images show evidence of

glycosaminoglycan (GAG, blue) remnants (arrows) within the newly formed

bone (NB) and adjacent connective tissue (CT) in the scaffold-BMP (A, B),

scaffold-OB (E, F), and Scaffold-OB-BMP (I, J). Larger GAG deposits were

found within the extracellular matrix in the scaffold-MSC and scaffold-MSC-BMP

group (C, D and G, H). Large proportions of adipose tissue (AT) were found in

groups including MSC. Residual mPCL-TCP was present in all groups (&) as

evidenced by voids within the tissue. Scale bars: left column 100 !m, right

column 20 !m.

Page 127: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

103

Fig. 15: Immunohistochemistry for type II collagen. The images demonstrate

positive staining results (arrows) for the Scaffold-MSC (C) and Scaffold-MSC-

BMP (G) group within the connective tissue (CT) lining the newly formed bone

(NB). No collagen II deposition was evident in all other groups. Scale bars: 100

!m.

Tartrate resistant acid phosphatase (TRAP) is a glycosylated monomeric

metalloenzyme expressed in mammals. Under physiological conditions,

TRAP is highly expressed by osteoclasts, activated macrophages, and

neurons. In osteoclasts, TRAP is commonly localized within the ruffled

border area, within lysosomes, and in Golgi cisternae and vesicles. TRAP

Page 128: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

104

positive cells can be stained using a colorimetric reaction resulting in

bright red colour deposition. TRAP staining on paraffin embedded

specimens showed high osteoclastic activity in all groups including

rhBMP-7 (Fig. 16 G-O) while no colour deposition was observed in the

other experimental groups (Fig. 16 A-F). In case of positive staining,

osteoclasts were lining the outer boundary of areas of new bone

formation in close proximity to the surrounding fibrous connective tissue

(Fig. 16 G-O, arrows).

Bromodeoxyuridine (5-bromo-2-deoxyuridine, BrdU) is a synthetic

nucleoside that is an analogue of thymidine. BrdU is commonly used in

the detection of proliferating cells in living tissues. BrdU can be

incorporated into the newly synthesized DNA of replicating cells during

the synthetic phase of the cell cycle, substituting for thymidine during

DNA replication. Antibodies specific for BrdU can then be used to detect

the incorporated chemical, thus indicating cells that were actively

replicating their DNA. In any case where labelled cells were transplanted,

immunohistochemistry showed positive staining for BrdU in cells

embedded in newly formed bone matrix as well as in bone lining cells

clearly indicating that the transplanted cells had actively contributed to

neoosteogenesis. However, cells positive for BrdU were also detected in

bone surrounding adipose and connective tissue. The amount of labelled

cells within bone was higher in groups including rhBMP-7, in tissues

other than bone it appeared to be higher in the groups including MSC

transplantation suggesting a higher degree of plasticity of the less

Page 129: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

105

committed MSC when compared to compact bone derived, differentiated

OB (Fig. 17).

Fig. 16: TRAP staining of paraffin-embedded specimen counterstained with

haematoxylin. In all groups including rh-BMP-7, positive, bright red staining was

clearly evident (arrows) in areas between newly formed bone (NB) and

connective tissue (CT). Residual mPCL-TCP (&) was evident represented by

voids within the tissue. Scale bars: left column 200 !m, middle column 100 !m,

right column 50 !m.

Page 130: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

106

Fig. 17: BrdU staining of paraffin-embedded specimen counterstained with

haematoxylin. Cells positive for BrdU were found within the newly formed bone (NB)

(arrows), as bone lining cells (arrow heads), and within fat (AT) and connective

tissue (CT). The amount of BrdU positive cells was significantly higher in groups

including rhBMP-7. Residual mPCL-TCP (&) was evident. Scale bars: 50 !m.

I

Page 131: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

107

SEM and EDX of areas identified as bone (Fig. 18 *) on histological PMMA

sections revealed an identical spectrum of composing elements for both OB

(Fig. 18 A and E) and MSC (Fig. 18 B and F) derived bone with high

amounts of calcium (Fig. 18, Ca, arrow heads) and phosphate (Fig. 18, P,

arrows). No calcium or phosphate could be detected in bone lining soft tissue

(Fig. 18, asterisks).

Fig. 18: Backscattered electron microscopy images on PMMA sections of OB (A)

and MSC derived (B) bone (*) and surrounding soft tissue (&). The EDX spectra of

OB and MSC derived bone (E and F) showed and identical elementary composition

with high amounts of calcium (arrow heads) and phosphate (arrows).

Page 132: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

108

Discussion

Mature bone consists of specialized cells secreting and remodelling the

surrounding extracellular matrix (ECM). Osteoblasts (OB), which

subsequently mature into osteocytes, synthesize and deposit the

proteinaceous ECM, which then becomes calcified, and secrete growth

factors such as hepatocyte growth factor (HGF), fibroblast growth factor

(FGF), and bone morphogenetic proteins (BMPs) stimulating proliferation

and osteogenesis. Osteoclasts are derived from the monocyte-macrophage

lineage and play a major role in bone remodelling. The ECM is composed of

collagenous proteins (predominantly collagen type I), non-collagenous

proteins (e. g. osteocalcin, osteopontin, osteonectin and bone sialoprotein)

and mineralized matrix (hydroxyapatite).

The use of autologous cells from the individuals’ tissue has shown promise in

the field of tissue engineering. This approach however necessitates the

removal of tissue from the individual, and in vitro isolation and expansion

before re-implantation to the site of intervention. Although, from an

immunological stance, the replacement of tissue with autologous cells is an

ideal situation, there are associated problems. Firstly, the harvest of tissue to

allow cell isolation and expansion requires surgical intervention which would

be on par with the grafting processes described previously. In addition, some

cell populations from primary sources have a low propensity for division thus

the expansion of such cells may prove problematic. This is sometimes

compounded with cellular senescence – a phenomenon describing the cease

of cellular division in primary cells, usually caused by a shortening of

Page 133: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

109

telomere length [180]. These limitations have made the search for cell

populations, which can be expanded in culture before implantation, of the

utmost importance.

OB can be isolated from mature cancellous or compact bone and expanded

in vitro. OB originate from mesenchymal cells of the marrow stroma. These

stromal cells provide all of the benefits of primary cells, however, have the

ability to undergo multi-lineage differentiation and a higher propensity for cell

division. These cells can easily be isolated from bone marrow aspirates and

have been shown to be able to differentiate into the osteogenic, myogenic,

chondrogenic and neurogenic lineages. Therefore they have been proposed

as the most suitable cell type for bone engineering applications, a direct

comparison between osteoblasts and MSC in regards to their regenerative

potential of bone in vivo has never been made.

In the present study, the osteogenic potential of in vitro expanded ovine OB

and MSC was therefore investigated in a murine model of ectopic bone

formation. It was found that OB displayed a higher rate of ossification when

compared to MSC as quantified by histomorphometry, !CT analysis, and

compression testing. These results are consistent with previous studies (see

chapter II)[174]. EDX analysis and immunohistochemistry for osteocalcin

confirmed that the mineralized extracellular matrix synthesized by both MSC

and OB resembled mature bone. Osteocalcin is a bone-specific protein of

46–50 residues that undergoes post-translational modification by vitamin-K-

dependent +-carboxylation of three glutamic acid residues. Osteocalcin is

Page 134: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

110

expressed by mature osteoblasts, binds strongly to hydroxyapatite, is stored

in bone matrix and released into the circulation [181, 182]. It is considered a

late and specific osteogenic marker determining terminal osteoblast

differentiation regulating bone crystal formation [155]. Histologically, the bone

synthesized by OB appeared to be of higher maturity as evidenced by early

osteoclast mediated remodelling and secondary organization with

ossification starting in close apposition to the scaffold struts.

The histological and immunohistochemical examination showed that both the

transplanted MSC and OB were viable, had mitotic activity and have

contributed to new bone formation by secreting bone ECM. BrdU labelling

seemed to have no negative effect on the amount of bone formation

observed.

In all specimens including MSC, a significantly higher proportion of adipose

tissue and less bone was observed. This underlines the difficulties in

extrapolating results obtained in vitro, where MSC reproducibly exhibited a

higher osteogenic potential (see chapter II), to an in vivo setting. The findings

suggest that OB isolated from compact bone might represent a suitable

alternative cell population for tissue engineering applications. It must,

however, be taken into account that microenvironmental conditions in ectopic

models of bone formation may again not be representative of specific cues

transplanted cells experience in orthotopic sites. Less committed cells of

high plasticity such as MSC might - once transplanted subcutaneously -

respond to a higher degree to growth and differentiation/dedifferentiation

initiating factors released by the surrounding adipose, fibrous and muscular

tissue. This, in conjunction with missing mechanical stimuli experienced in an

Page 135: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

111

orthotopic bony environment, might prevent MSC from differentiating into

bone synthesizing osteoblasts.

As mentioned before, bone morphogenetic proteins (BMPs) are members of

the TGF-" superfamily. Of the BMPs that have displayed clinical utility for de

novo bone formation in vivo, BMP-2 and BMP-7 (osteogenic protein 1, OP-1)

are commercially available through the use of recombinant DNA technology.

In particular BMP-7 was shown to have osteoinductive effects in a variety of

studies [167, 183-187]. When transplanted in combination with ovine OB,

rhBMP-7 lead to a significant increase in bone formation while only little

effect was observed with MSC. One reason might be the dose dependent

response of MSC to BMPs both in vitro and in vivo [188-190]. The

administered dose of 5 !g might not have been sufficient to outplay the

continuous release of tissue specific growth factors from the surrounding

muscle or fat and to stimulate osteogenic MSC differentiation in an ectopic

model while on the other hand the BMP mediated signal was adequate to

excite a response in the more committed OB.

Tartrate-resistant acid phosphatase (TRAP) is a glycosylated monomeric

metalloenzyme expressed in mammals. It has a molecular weight of

approximately 35 kDa, a basic isoelectric point (7.6-9.5), and optimal activity

in acidic conditions. TRAP is synthesized as latent proenzyme and activated

by proteolytic cleavage and reduction. Under physiological conditions, TRAP

is highly expressed by osteoclasts, activated macrophages, and neurons

[191-193]. The exact physiological role(s) of TRAP is unknown, but many

functions have been attributed to this protein. In knockout mice studies,

those with a phenotype of TRAP-/- showed mild osteopetrosis, with greatly

Page 136: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

112

reduced osteoclast activity, resulting in thickening and shortening of the

cortices, the formation of club-like deformities in the distal femur, and

widened epiphyseal growth plates with delayed mineralization of cartilage, all

of which increased with age. Likewise in TRAP overexpressing transgenic

mice, mild osteoporosis occurred along with increased osteoblast activity and

bone synthesis. Proposed functions of TRAP include osteopontin/bone

sialoprotein dephosphorylation, the generation of reactive oxygen species,

iron transport, and as a cell growth and differentiation factor [194, 195].

Amongst others, osteoclast and TRAP activity have been shown to be

stimulated by rhBMP-7 [187, 196, 197] which might have resulted in positive

staining in all groups including the administration of 5 !g rhBMP-7 while no

positive staining for TRAP was observed in all other experimental groups.

If BMPs come in contact with osteoblasts they stimulate the production of

angiogenic factors, such as vascular endothelial growth factor and fibroblast

growth factor [198]. This then leads to the recruitment and activation of

endothelial cells necessary for new blood vessel formation. It was also

demonstrated that BMPs have direct effects on endothelial cells stimulating

migration, proliferation and tube formation. Mice with knockout phenotypes of

different BMPs often have cardiovascular problems, indicating an important

role for BMPs in angiogenesis [199, 200]. These angiogenetic effects of

BMPs in the literature might have contributed to an increased amount of

neovascularisation in transplanted specimens including rhBMP-7 as

observed in the present study.

Development and formation of the skeleton (ossification) occurs by two

distinct processes - intramembraneous and endochondral ossification. Both

Page 137: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

113

intramembraneous and endochondral ossification occur in close proximity to

vascular ingrowth. Intramembraneous ossification is characterized by

invasion of capillaries into the mesenchymal zone and the emergence and

differentiation of mesenchymal cells into mature osteoblasts. These

osteoblasts constitutively deposit bone matrix, leading to the formation of

bone spicules. These spicules grow and develop, eventually fusing with other

spicules to form trabeculae. As the trabeculae increase in size and number

they become interconnected, forming woven bone, a disorganized weak

structure with a high proportion of osteocytes, which eventually is replaced

by more organized, stronger lamellar bone. Intramembraneous ossification

occurs during embryonic development and is involved in the development of

flat bones in the cranium, various facial bones, parts of the mandible and

clavicle and the addition of new bone to the shafts of most other bones. In

contrast, bones of load-bearing joints form by endochondral formation.

Chondral ossification requires preformed cartilaginous tissue which is

subsequently replaced by bone.

In the present study, bone and especially its surrounding tissue formed after

transplantation of MSC stained positive for type II collagen and alcian blue

confirming the presence of high amounts of glycosaminoglycans

characteristic of cartilage. The findings suggest that MSC mediated bone

formation occurred by endochondral ossification through hypertrophic,

chondrocyte-like cells confirming results by other research groups [174, 201].

Furthermore, bone synthesized by OB stained only weakly for GAG and

negative for type II collagen while the adjacent connective tissue was entirely

negative for both GAG and collagen. This could indicate that the

Page 138: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

114

transplantation of OB leads to intramembraneous ossification by elongated,

spindle-shaped osteoblasts [174]. The small amounts of GAG found within

the bone synthesized by OB might be attributed to chondroitine-4-sulfate and

hyaluronic acid both of which are components of bony extracellular matrix

[202]. Nevertheless, the low GAG amounts could also indicate that bone

formation by transplanted OB follows endochondral mechanisms and may

have progressed to an advanced stage of bone maturation.

Summary

In the present study, ovine MSC isolated from bone marrow aspirates and

OB from cortical bone explants were compared regarding their osteogenic

potential after transplantation in vivo. It was found that transplanted cells

show a high degree of survival and actively contributed to enchondral

osteogenesis. When compared to MSC, OB showed a higher degree of bone

deposition while OB derived bone was of higher maturation. Additional

stimulation with rhBMP-7 increased the rate of bone synthesis for both MSC

and OB but also increased neovascularisation and osteoclast activity. These

results suggest that origin and commitment of transplanted cells highly

influence the type and degree of ossification; furthermore that rhBMP-7

represents a powerful adjuvant for bone tissue engineering applications and

that mature bone might be an adequate alternative cell source in the context

of bone regeneration.

Page 139: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

115

Chapter IV

Establishment of a Preclinical Ovine Model for Tibial Segmental Bone

Defect Repair by Bone Tissue Engineering Methods

Page 140: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

116

Page 141: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

117

Introduction

The concepts of tissue engineering have matured over the last two decades

and, when reviewing the field, it can be concluded, that the translation of

tissue engineering strategies from bench to bedside is a difficult, expensive

and time-consuming process. Hollister recently addressed the different

issues pertaining to the translation and commercialization of tissue-

engineered constructs (TEC) [203]. He argues that the so-called “Valley of

Death”, which describes the lag between research and commercialisation, is

highly prevalent in the area of tissue engineering/regenerative medicine and

originates from the high costs associated with technology development and

the need for funding preclinical animal models and clinical trials for

regulatory approval.

A set of harmonized standards (ISO 10993) that address requirements for

the biological evaluation of medical devices has been developed by the

International Organization for Standardization (ISO). Compliance with ISO

standards is required throughout Europe. The US food and drug

administration (FDA) issued the blue book memorandum #G95-1 that

adopted ISO nomenclature for device categories and included an FDA-

modified flowchart designating the type of testing needed for each device

category, and made several modifications to the testing requirements

outlined in ISO 10993-1, adding various requirements in several device

categories. Briefly, device manufacturers were asked to determine which

kinds of biological effects are of concern for the materials in a particular

device, based on the nature and duration of the product's end use. Such

effects can include sensitization, irritation, haemocompatibility, various other

Page 142: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

118

types of toxicity, and reproductive or developmental changes. In all, 12

categories of potential biological concerns are identified in both the FDA

memorandum and ISO 10993 to conclusively confirm the safety and efficacy

of the TEC under investigation.

The lack of translation in the field of bone engineering might be related to

difficulties in integrating individual technical discoveries into model tissue

engineering systems, in manufacturing scale up, in funding, and in

regulatory approval. To establish a tissue engineering concept in a clinical

setting, a rigorous demonstration of the level of therapeutic benefit in

clinically relevant animal models is a conditio sine qua non. To closely

simulate human in vivo conditions, and to assess the effects of implanted

bone grafts and tissue engineered constructs on segmental long bone

defect regeneration, a number of large animal models have been

developed. In a recent literature review [126], however, it was concluded

that many of the preclinical models published are not well described, defined

and standardized and therefore difficult to reproduce. Only a small segment

of the research community works on a paradigm shift to translate tissue

engineering of bone into the clinical orthopaedic and reconstructive surgery

area. Therefore, in this thesis, effort was concentrated on the establishment

of a well characterized, standardized preclinical animal model (Fig. 1). In the

present chapter, the approach is illustrated of how to translate tissue

engineering of bone from bench to bedside by presenting a road map which

describes the validation of the functionality of a model to study the

regeneration of critical-sized segmental bone defects in a highly load-

bearing large animal.

Page 143: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

119

Fig. 1: Road map to establish a critical sized bone defect model in a large animal.

Regulatory framework

Within the European Union (EU), the task of harmonizing and regulating

medical devices is handled by the European Commission in close

cooperation with member state health authorities. The purpose of the EU

harmonization effort is to merge the differing national requirements into one

law, which can be applied throughout the European Union. Legislation

adopted through this process covers implantable, non-implantable, and in

vitro diagnostic medical devices to provide manufacturers with the basis to

certify their compliance with EU-wide safety requirements. The task of

complying with essential requirements can be simplified by voluntarily using

EU harmonized standards.

Page 144: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

120

The Commission has listed over a hundred EU wide harmonized medical

device standards addressing various essential requirements. These

standards have been developed and/or identified by the European

standards organizations (CEN, CENELEC and ETSI) and published in the

Official Journal (the EU equivalent of the U.S. Federal Register), thereby

giving them special recognition as so-called harmonized standards linked to

EU legislation. All other existing standards not published in the Official

Journal are either national or industry standards. The European harmonized

standards for medical devices are often based on international standards

(ISO or IEC). For example, the European committee for standardization

(CEN) lists a number of mandated standardization projects for the biological

evaluation of medical devices (http://www.cen.eu/cenorm/homepage.htm).

In the EU, a company that intends to commercialize a medical product may

submit a single application to the European Medicines Agency (EMEA) for a

'marketing authorisation' that is valid simultaneously in all EU Member

States, plus Iceland, Liechtenstein and Norway. This procedure is referred

to as the 'centralised authorisation procedure'. The decisions about the

authorisation of medical products are based on an objective, scientific

assessment of their quality, safety and efficacy. Conducting these

assessments within the EU is the primary role of the EMEA. Through its

scientific committees, the EMEA assesses every medicine for which a

marketing-authorisation application has been submitted, and prepares a

recommendation that is then relayed to the European Commission, which

has the ultimate responsibility for taking decisions on granting, refusing,

Page 145: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

121

revoking or suspending marketing authorisations. The Food and Drug

Administration (FDA) is a scientific, regulatory, and public health agency

whose authority includes overseeing the marketing of products relevant to

medical practice in the United States. Devices are classified based on the

extent of oversight needed to ensure public safety. Divisions within the FDA

provide specific expertise regarding drugs, devices, biologic products and

combinations thereof. Various pathways exist to apply for marketing through

the FDA, depending on the nature of the product and its intended use.

Expert panels advise the agency on issues related to product safety and

efficacy. The development of a medical device from concept to product-

launch typically takes between 4-10 years and costs between 5 and 300

million dollars depending on the complexity of the device, its classification,

and required regulatory process [204]. The FDA recommends provision of

detailed information on the chemical composition and physical properties of

the device under review. Device performance tests include bench testing

(pH testing, dissolution and stability), biocompatibility testing and evaluation

in an animal model. According to the “Class II Special Controls Guidance

Document: “Resorbable Calcium Salt Bone Void Filler Device” issued in

2003 (http://www.fda.gov/cdrh/ode/guidance/855), the respective animal

model should

- Be representative of the indications for use

- Cover the full range of anatomical sites proposed for use, and

specifically how the device is to be used

- Include the use of skeletally mature animals and a critical size defect

- Include use of the predicate device and/or autogenous bone graft as

Page 146: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

122

the positive control group(s) and use of empty defect as a negative

control

- Involve the use of radiography, histology, and histomorphometry to

assess bone formation and device resorption at various, relevant time

points over the course of healing, in addition to supportive

biomechanical testing of the new bone formed

- Aim at adequate study duration to demonstrate bone healing and the

effects of any residual device material.

In order to translate tissue engineering based therapies from bench to

bedside, it is mandatory to rigorously demonstrate both the level of

therapeutic benefit in clinically relevant animal models and sufficient

understanding of the mechanisms by which scaffold/cell or scaffold/growth

factor constructs integrate into host sites, form new bone and restore

function. To achieve this aim in this thesis, it was decided to validate the

functionality of engineered bone constructs of clinically relevant size in a

suitably highly load-bearing large animal model such as sheep. The use of

large animals lies in the high value of the data obtained from these models

pertaining to

(i) scaffold biomechanical properties

(ii) scaffold degradation & resorbability; and

(iii) survival of transplanted cells or release kinetics of growth factors

(which are all critical parameters to the ultimate efficiency of bone

constructs).

Page 147: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

123

The high value of the data is attributed to great similarities to human

conditions in regards to bone biology, mineral composition, turnover and

remodelling and the mechanical loading environment.

Bone defect fixation can generally be achieved by plate fixation,

intramedullary nailing, or the application of an external fixator (Table 1). In

humans, intramedullary nailing is a commonly chosen treatment modality for

diaphyseal fractures of the lower extremities. Since these central load

carriers are less susceptible for tilting in the frontal plane, custom made

intramedullary nailing systems have been applied for fixation of large

segmental bone defects in animal models [4, 93, 97, 98]. In the context of

tissue engineering, however, intramedullary stabilization can impede the

placement of a solid, one-piece load-bearing scaffold and necessitates two

surgical approaches. Alternatively, external fixators have been widely used

as they offer versatility and ease of application [3, 12, 96]. However, with

external fixators, healing periods are reported to be significantly longer

[205]. External fixators represent a burden exceeding the physiological

circumference of the animal limb and are prone to infection and pin

loosening, especially in long-term studies. Therefore, defect fixation with

plates or internal fixators offers considerable advantages.

Page 148: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

124

Advantages Disadvantages

External fixator - Versatility - Ease of application - Marginal effect on

surrounding soft tissue - Minimal intraoperative

trauma - Open space for

implantation of biomedical construct

- Clinically only applied as temporary fixation device

- Schanz screw loosening - Pin track infections

Intramedullary nail - Standard treatment for diaphyseal fractures of lower extremity

- Availability of UTN to avoid reaming related problems

- Central load carrier - High tolerance of

maximum applied forces - Low axial deviations - Reaming debris as

possible source of multipotent stem cells

- Impairment of bone blood circulation by reaming

- Thermal necrosis after reaming

- Air or fat embolism - Failure of locking bolts - Limited application of

load-bearing scaffolds - Prolonged healing period

Plate fixation - Standard treatment for metaphyseal fractures

- Optimal reduction - Minimal influence on

defect (LC-LCP) - Open space for

implantation of biomedical construct

- Eccentrical load carrier - Impairment of periosteal

blood flow (non LCP) - Bone loss through stress

protection (non LCP) - Unclear role of plate

fixation in tibial shaft fractures

- Prone to axial deviations and implant failure

Table 1: Advantages and disadvantages of different fixation devices.

Intramedullary nail versus plate fixation versus external fixation

Diaphyseal tibial fractures are most commonly stabilized by intramedullary

nailing. Despite its wide-spread application, it is associated with both

positive and negative side aspects. Reaming can impair the bone’s blood

circulation system considerably [206, 207]. Reaming and nail insertion

significantly increase the intramedullary pressure. This can lead to air or fat

embolisms causing pulmonary microvascular damage [208, 209] and a rise

in temperature in the medullary canal. Rise in temperature of 50°C and

more have been described [210, 211] which may result in thermal necrosis

of bone tissue also altering the endosteal architecture. This may induce

biological failure compromising fracture or defect healing [212]. The damage

Page 149: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

125

of local tissue, however, is mostly reversible and compensated for within 6-8

weeks. With unreamed nailing systems the interlocking screws are prone to

failure while small dimension solid tibial nails cannot always provide

adequate stability. The introduction of angle stable locking bolts could

reduce failure rates. In the context of tissue engineering an intramedullary

nail may impede the placement of a solid, one-piece load-bearing scaffold or

tissue engineered construct. Central load carriers, however, are less

susceptible for tilting in the frontal plane. Therefore, custom-made

intramedullary nailing systems have been applied in a number of

publications for the fixation of segmental bone defects in large animal

models [59, 93, 97, 98] (Fig. 2).

Fig. 2: Schematic representation of commonly applied methods for the fixation of

segmental defects in large animal models.

Metaphyseal fractures are preferably treated with angle-stable plates [213].

Sufficient reduction and stability can be achieved by plate osteosynthesis

even in complicated fractures [214, 215]. Conventional plate osteosynthesis,

Page 150: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

126

however, can result in histologically and radiologically verifiable bone loss,

which has been attributed to stress protection (Wolff’s law). Studies have

revealed an increase in cortical bone porosity proximate to the plate soon

after plate osteosynthesis. This porosity was explained by impaired vascular

perfusion underneath the plate [216] due to high compression forces

between plate, periosteum and bone [217]. Contact pressure between plate

and bone and resulting circulatory disturbances may prolong fracture

healing and increase the risk of infection and re-fracture after implant

removal [218]. Therefore, plate systems were developed where load and

torque transmission act through the screws only as achieved by angle-

stable, interlocking screws. Thus, the stabilisation system acts rather like an

external fixator making plate-bone contact unnecessary to achieve stable

fixation. The application of monocortical screws can further minimize screw-

related intramedullary circulatory disturbances [219]. The role of plate

fixation in the treatment of human tibial shaft fractures varies between

different clinics and colleges of surgeons, as the literature lacks randomised

trials comparing plate fixation with other established treatment concepts. In

the area of tissue engineering and related animal studies, defect fixation

with internal fixators offers the great advantage of a minimal influence of the

fixation device on the created defect site both concerning space for scaffold

implantation and biological factors. When compared to external fixators or

intramedullary nails, rates of infections (pin-track infection), infection related

complications and non-union rates (6-25%) are lower. However, higher

numbers of mal-alignment may be observed [220].

Clinically, external fixation is commonly applied as a temporary fixation

Page 151: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

127

device in case of severe open or contaminated fractures. In animal models,

external fixators have been widely used offering versatility and ease of

application [3, 12, 96]. Due to minimal intraoperative trauma, the

surrounding soft tissue is only affected marginally with external fixators.

However, Schanz screw loosening is the most common complication with

external fixator often resulting in pin track infections. When compared to

intramedullary nails, external fixators do not affect the defect site as

extensively leaving open space for the implantation of bone grafts and

tissue engineered constructs. However, with external fixators healing

periods are reported to be significantly longer when compared to other

fixation devices [205].

In conclusion, the establishment of a critical size segmental defect model in

a large animal is a challenging task. Hence, in the following section the

knowledge and experience of how to establish such a defect model in an

ovine model (aged 6-8) is described in detail.

Page 152: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

128

Roadmap to establish a preclinical model for segmental

bone defect research

To translate scaffold-based bone engineering concepts from bench to

bedside, the level of therapeutic benefit must be conclusively demonstrated

and the essential underlying biological mechanisms elucidated. The

functionality of engineered bone constructs must be validated in a high load-

bearing large animal model and characterized with respect to scaffold

biomechanical competence, integration, degradation & resorption, cell

survival and release kinetics of growth factors (if applicable).

Among the different indications of bone grafting in orthopaedic surgery, in

the current study, focus was placed on critical-sized, diaphyseal, tibial

defects for several reasons:

(i) The tibia is the most commonly fractured long bone, with an

incidence of 17 per 100,000 person-years [221]. Compromised

healing and therefore delayed healing or non-unions in tibial shaft

fractures are common. Up to 60% of segmental defects occur in

the tibia [222]. The overall rate of delayed unions in tibial fractures

ranges from 5% to 61% and the overall rate of non-union might be

as high as 21% [223-226];

(ii) Impaired functionality is significant as the tibia is a highly weight-

bearing bone and tibial shaft fractures often occur in young and

active adults. With prolonged treatments, as in the case of

delayed or non-union, the resultant missed days of work and loss

of wages increase both the direct costs of care and the indirect

costs associated with lost productivity;

Page 153: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

129

(iii) Scaffold/cell [12] and scaffold/BMP [115] based concepts have

been used with interesting preliminary results in tibial defects but

there are no randomized studies comparing the results of different

scaffold based treatment concepts to the performance of

autologous bone graft in an ovine segmental defect model.

Pilot study limited contact locking compression plate (LC-LCP)

In order to establish a critical-sized segmental bone defect model in the

sheep tibia, a pilot series of 3 animals (merino sheep, weight 39±1 kg, age

6-7 years) was conducted. All animals were in good health but of small

statue (related to the geographic and climatic conditions in central

Queensland, Australia). Under general anaesthesia, a 20 mm mid-

diaphyseal defect was created using an oscillating saw. A previous literature

search had shown that a 20 mm defect was the smallest critical-sized defect

described in sheep tibiae [126]. The defect was stabilized using a 9-hole 4.5

mm narrow LC-LCP (limited contact locking compression plate, Synthes)

anteromedially with three bicortical screws proximal and distal and left

untreated. The implant, a modern internal fixation system, was chosen since

it had previously been used with success to stabilize an experimental

fracture in sheep femora (Wullschleger et al., unpublished data). After

surgery, the operated leg was bandaged with hard plaster (Vet lite, Runlite

SA, Micheroux, Belgium). The animals were held in a suspension trolley for

24 h to allow recovery from anaesthesia prior to release into a paddock. The

animals were allowed unrestricted weight-bearing. In all three animals, 7-10

days after surgery, plate bending occurred as suspected by clinical

Page 154: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

130

assessment and confirmed by x-ray analysis (Fig. 3). It was concluded that

these failures resulted from critical loads (valgus stress) occurring during the

act of the animal standing up from a lying position. It should be noted

however that while the implant was proven not to be strong enough in an

immediately fully weight bearing animal model, this should not be

interpreted as having implications for the treatment of similar conditions in

human patients, since compliant patients can be advised to weight bear only

partially and to avoid critical loads that may cause implant failure.

Fig. 3: 2 cm segmental bone defect in a sheep tibia stabilized with a 9-hole narrow

4.5 mm limited contact locking compression plate (LC-LCP plate) (A, E, F); implant

bending at day 7 due to critical loads and valgus stress (B). Starting defect

regeneration was observed at day 14 (C) and the uncontrolled biomechanical

stimulation led to defect bridging on the postero-lateral side at day 28 (D).

Page 155: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

131

Finite element modelling

In response to the in vivo failures of the internal fixation plate seen in the

first pilot study, finite element analysis was conducted to aid the selection of

a fixation device with mechanical properties sufficient to resist failure of the

plate via bending. In addition to the material properties and the bulk size of

the plates, the type of screw hole, single or combination, and screw hole

configuration are determinants of implant stiffness. Four plates with different

sizes and screw hole type were investigated, 3.5 mm broad LCP (stainless

steel, 220 GPa), 4.5 narrow LCP (stainless steel, 220 GPa), 4.5 mm broad

LCP (titanium, 105 GPa), and the LISS plate (less invasive stabilization

system, titanium alloy, 110 GPa).

Finite element models (tetrahedral elements) were constructed for each of

the four plates from technical drawings supplied by the manufacturer

(ABAQUS v6.5). Four-point bending tests were then simulated to determine

the equivalent bending stiffness (ISO 9585: Implants for surgery –

Determination of bending strength and stiffness of bone plates) of each of

the plates alone using a linear elastic solver. The 4.5 narrow LCP used in

the first pilot study was found to have the lowest equivalent bending

stiffness of all the plates tested. The 4.5 mm broad LCP was determined to

be the stiffest plate (15.32 Nm2) closely followed by the LISS plate (14.81

Nm2).

Page 156: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

132

Implant testing

The most rigid plates determined by finite element modelling were selected

for in vitro biomechanical testing. In addition to the locking plates, a dynamic

compression plate was also included. In vitro biomechanical testing was

performed to determine the stiffness of the implant-bone constructs. A four-

point bending test was carried out to determine the equivalent bending

stiffness. Briefly, a defect of 2 cm was created in ovine cadaver tibiae and

fixed with either (n=3) a 4.5 mm narrow LC-LCP (stainless steel), a LISS

(titanium) and a 4.5 mm broad DCP (dynamic compression plate, stainless

steel)(all Synthes, Fig. 4). N.B. The broad screw hole arrangement of the

4.5 mm broad LCP was found to be unsuitable for the narrow sheep tibia

and was thus substituted with the LISS plate, which showed similar bending

stiffness to the 4.5 mm broad LCP in finite element analyses. Both ends of

the tibiae were potted in cups for load transfer using bone cement (poly-

methyl-methacrylate (PMMA)) in a standardized manner. To ensure that the

same moment arm acted on the respective constructs care was taken to

ensure that the free length between the cups remained constant for all

bones. Constructs were mounted in four-point bending configuration and

loads applied using a biaxial universal testing machine (Instron 8874,

Instron, Norwood, USA). The test was repeated three times for each

bone/implant construct. During the entire procedure, the tibiae were

wrapped in saline soaked gauze to avoid the samples drying out. The

equivalent bending stiffness was calculated according to ISO norm 9585

“Implants for surgery – Determination of bending strength and stiffness of

bone plates”, which specifies the test configuration based on the plate screw

Page 157: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

133

hole spacing. The highest stiffness was determined for the 4.5 mm broad

DCP with E=0.058 Nm2 (SD=0.0075). The modified LISS plate provided only

marginally higher stiffness (E = 0.028, SD = 0.0049) than the 4.5 narrow LC-

LCP used in the pilot study (E=0.023 Nm2, SD=0.0024)(Fig. 5).

Fig. 4: The figure illustrates the implants used to biomechanically determine the

stiffness of implant-ovine bone constructs in vitro: A 4.5 mm narrow Limited

Contact Locking Compression Plate (LC-LCP, A, D), a tibial Less Invasive

Stabilization System (LISS, B, E) plate, and a 4.5 mm broad Dynamic Compression

Plate (DCP, C, F)(all Synthes). The length of the LISS plate was tailored to fit an

ovine tibia as indicated by the red bar. G and H demonstrate the setup of the four

point bending test.

Page 158: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

134

Fig. 5: The figure illustrates the equivalent bending stiffnesses as per ISO 9585:

Implants for surgery – determination of bending strength and stiffness of bone

plates. The highest stiffness was determined for the 4.5 mm broad DCP with

E=0.058 Nm2 (SD=0.0075). Error bars represent standard deviations.

Pilot study dynamic compression plate

For further experimental surgeries, the 4.5 mm broad DCP as the stiffest

implant was chosen. Another series of three animals (42±1 kg, age 6-7

years) was then operated on to evaluate if the new implant would show

sufficient biomechanical strength in vivo and to evaluate if a defect size of 2

cm was critical. The 10-hole DCP plate was fixed with 4 screws proximal

and 3 screws distal of the defect. Care was taken to completely remove the

periosteum within the defect area as well as 1.5 cm proximal and distal of

the defect [185]. Animals were subjected to the same postoperative

treatment as described previously. After 12 weeks, two out of three animals

showed islets of bridging of the defect as assessed by conventional x-ray,

!CT analysis (microCT 40, Scanco, Switzerland) and histology. For !CT

analysis, samples were scanned with a voxel size of 16 !m. Samples were

Page 159: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

135

evaluated at a threshold of 220, a filter width of 0.8 and filter support of 1.0.

No further implant failures in terms of plate bending had occurred. In one of

the animals where the defect was bridged, the most proximal screw had

loosened (Fig. 6). This may have changed the rigidity of fixation and hence

the biomechanical conditions allowing micro movements at the defect site

which in turn could have stimulated callus formation. Other factors such as

surgical technique and inter animal variability in terms of weight etc. might

have caused the second of three defects to bridge.

Fig. 6: 2 cm ovine tibial defect stabilized with a broad 4.5 mm dynamic

compression plate (DCP) (A) fixed with 4 bicortical screws proximally and 3 screws

distally. X-ray image after surgery (B) and after 12 weeks (C) shows loosening of

two screws proximal of the defect (arrows) and consecutive bone formation.

To ensure a non-union rate of 100%, the defect size was then increased to

3 cm, again the periosteum was removed. This resulted in non-unions after

12 weeks in all of eight animals in the negative control group (weight 45±2

Page 160: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

136

kg, age 6-7 years). Hence, a defect size of 3 cm in the sheep may be

considered of critical size (Fig. 7). As a positive control eight 3 cm defects

were reconstructed with autologous cancellous bone graft from the iliac

crest representing the gold standard treatment. No implant failures were

observed and after 12 weeks all defects had healed and radiographic

evaluation revealed defect consolidation. However, it must be pointed out

that radiographs cannot provide any information on bone quality and

functionality, thus it was necessary to undertake biomechanical testing. This

baseline data was later on used to assess the performance of medical grade

polycaprolactone-tricalcium phosphate (mPCL-TCP) scaffolds in

combination with and without human recombinant bone morphogenetic 7

(rhBMP-7) in respect to their bone regeneration capacity.

Fig. 7: 3 cm mid-diaphyseal tibial defect in a sheep tibia (A) stabilized with a 4.5

mm dynamic compression plate (DCP) (B). Radiographic images immediately (C)

and 12 weeks after surgery (D) show the critical nature of the defect size.

Page 161: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

137

Summary

To replace or restore the function of traumatized, damaged or lost bone is a

major clinical and socio-economic challenge. Bone tissue engineering has

been suggested as an alternative strategy to regenerate bone. In principal,

the discipline of tissue engineering aims at combining cells with scaffolds, or

appropriate growth factors such as FGF-2 (fibroblast growth factor 2), VEGF

(vascular endothelial growth factor) or BMPs (bone morphogenetic

proteins), to initiate and stimulate tissue repair and regeneration. Despite

initial success, it must be recognized that bone engineering has not yet

been able to deliver significant progress in terms of translated clinical

applications and commercialized products. To tackle major bone tissue

engineering problems, researchers must perform functional assessment of

the biological and biomechanical parameters of generated constructs.

Furthermore, to allow comparison between different studies, animal models,

fixation devices, surgical procedures and methods of taking measurements

need to be standardized to achieve an efficient accumulation of reliable data

as a foundation for future orthopaedic and tissue engineering developments

and their translation to the clinic.

Conventional DCPs may not meet all the requirements postulated for a

modern fracture fixation device and higher rates of screw loosening have

been described (full load carried by the screws) when compared with LCPs

where loads are equally carried by both screws and plate which, from a

mechanical point of view, can be seen as one unit as a result of the locking

mechanism. It was reported that DCPs impede periosteal circulation through

compression other studies, however, have vitiated these findings.

Page 162: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

138

Nevertheless, DCPs are still used in clinical settings. To minimize effects on

periosteal blood flow, limited contact (LC) plates were developed and

introduced to the orthopaedic/trauma market [227] [228]. To further preserve

biomechanical integrity, the concept of locked internal fixators was

introduced offering improved long-term biomechanical stability and

decreased susceptibility to infection. However, narrow LCPs do not provide

the required stiffness in an immediate fully load-bearing animal model and

will only be of sufficient strength if the animals are immobilized for a longer

period of time (6-12 weeks) until the regenerating bone has gained some

intrinsic biomechanical stability [229]. Broad LCPs on the other hand do not

fit ovine tibiae as a result of their dimensions and therefore do not represent

an alternative.

In conclusion, the most important issues related to the establishment of a

large preclinical model for segmental bone defect research were addressed

and discussed, and it was demonstrated how to develop such a model

relying on a DCP plate fixation system. With respect to the “Valley of Death”,

an important milestone was achieved in establishing a highly reproducible

large animal model, which is an essential requirement to systematically

assess different bone grafting materials, scaffolds, tissue engineered

constructs and growth factors on the path towards the translation into a

routine clinical application.

Page 163: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

139

Chapter V

Reconstructing large segmental bone defects in an ovine model by

tissue engineering methods

Page 164: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

140

Page 165: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

141

Introduction

In orthopaedic and trauma surgery, extensive bone loss is associated with

major technical and biological problems. Fractures in cancellous bone areas

- such as the proximal humerus, the distal radius or the tibial plateau - often

lead to impaction of bone and subsequent defect formation after reduction.

Other causes of traumatic bone loss include highly comminuted fractures

and atrophic diaphyseal non unions of long bones. Bone grafts as the gold

standard treatment possess osteoconductive and osteoinductive properties,

however, they still face significant disadvantages. These include limited

access and availability, donor site morbidity and haemorrhage, increased risk

of infection, and insufficient transplant integration with following graft

devitalisation and subsequent resorption resulting in decreased mechanical

stability. As a result, recent research focuses on the development of

alternative therapeutic concepts.

Engineering bone by combining a suitable scaffold with osteoblastic or

osteoprogenitor cells and/or bone formation stimulating growth factors has

advanced as a promising new approach to bone repair and regeneration.

However, in order to establish a tissue-engineering concept in a clinical

setting, a rigorous demonstration of the level of therapeutic benefit in

clinically relevant animal models is absolutely essential. Difficulties in

integrating individual technical discoveries in model tissue engineering

systems and in manufacturing scale up combined with shortages in funding,

and in regulatory approval that are associated with the translation of tissue

Page 166: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

142

engineering based concepts make clinical applications a difficult target to

reach [230].

To circumvent the disadvantages associated with bone grafts, a variety of

different materials for bone replacement have been developed [231].

Calcium phosphates (CaP) form a large group of materials that are

considered suitable substitute biomaterials for bone augmentation. Calcium

phosphate based materials have been shown to be biocompatible evoking

no adverse inflammatory reactions after implantation [6, 232-234]. Calcium

phosphate has also demonstrated osteoinductive effects in numerous

publications [6, 234].

Aliphatic polyesters such as PLLA/PDLA, PLLLA/PGA, PCL and others have

demonstrated excellent safety profiles in multiple in vitro, animal, and clinical

studies and have approval for clinical applications [235, 236]. They do not,

however, meet the structural and mechanical requirements for use in

orthopaedic applications of the lower extremities. Therefore, a second

generation of scaffolds has been developed. These scaffolds are based on a

medical grade PCL-CaP composite [160, 237], and combine the favourable

mechanical properties of the polyester with the osteoinductive characteristics

of the ceramic component. So far, these mPCL-TCP scaffolds have been

tested in rat and pig skull defect as well as in a spinal fusion model with

promising outcomes [238].

Healing of osseous tissue is regulated by growth factors and other cytokines

in a sequence of overlapping events similar to cutaneous wound repair. In

ideal circumstances, this process mimics embryonic bone development,

Page 167: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

143

allowing the replacement of damaged bone with new bone, rather than with

fibrous scar tissue. This process is driven by cellular and molecular

mechanisms controlled by the TGF-" superfamily of genes, which encodes a

large number of extracellular signalling growth factors [239]. Bone

morphogenetic proteins (BMPs) are a well-studied group of these growth

factors involved in the processes of bone healing; and the human genome

encodes at least 20 of these multifunctional polypeptides [240]. Among

several of its functions, BMPs induce the formation of both bone and

cartilage by stimulating the cellular events of mesenchymal progenitor cells.

However, only a subset of BMPs, most notably BMP-2, -4, -7, and -9, have

osteoinductive activity, a property of inducing de novo bone formation [241].

Studies involving mutations of BMP ligands, receptors and signalling proteins

have shown important roles of BMPs in embryonic and postnatal

development. Severe skeletal deformation, development of osteoporosis,

reduction in bone mineral density and bone volume are all aberrations

associated with disrupted and dysregulated BMP signalling [242, 243].

Several other growth factors produced by osteogenic cells, platelets and

inflammatory cells participate in bone healing, including IGF-1 and -2, TGF-

"1, PDGF and FGF-2 [244]. The bone matrix serves as a reservoir for these

growth factors and BMPs, which are activated during matrix resorption by

matrix metalloproteases [245]. Additionally, the acidic environment that

develops during the inflammatory process leads to activation of latent growth

factors [246], which assist in the chemo-attraction, migration, proliferation,

and differentiation of mesenchymal cells into osteoblasts or chondroblasts

[246]. All of these functions are driven by a complex mechanism of

Page 168: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

144

interaction among growth factors and other cytokines, which are influenced

by several regulatory factors.

The majority of studies investigating the effects of BMP-7 on new bone

formation were conducted in small animal models and experimental studies

in critical-size defect models of the weight-bearing lower extremity show non-

uniform results. Therefore, in this present study, an animal study was

designed to investigate the effects of rhBMP-7 in combination with mPCL-

TCP composite scaffolds built by computer aided design using rapid

prototyping technologies on bone healing. It was hypothesized that such a

tissue engineered bone graft may enhance the treatment of large segmental

bone defects without the need of vascularised autografts, non-vascularised

autografts, and/or allografts, and could therefore represent a clinical

alternative to bone autografts for the reconstruction of large tibial and femoral

defects.

Page 169: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

145

Material and Methods

Scaffold fabrication and preparation

Bioresorbable cylindrical scaffolds of medical grade )-poly-caprolactone

incorporating 20% "-tricalcium phosphate (mPCL–TCP) (outer diameter: 20

mm, height: 30 mm, inner diameter: 8 mm) (Fig. 1) were produced by fused

deposition modelling (FDM) as described previously (Osteopore

International, Singapore; www.osteopore.com.sg) [169]. The structural

parameters of the scaffolds were tailored by computer aided design (CAD)

and included 100% pore interconnectivity within a range of 350–500 µm size,

70% scaffold porosity, and a 0/90° lay down pattern [169] [247]. This

architectural layout is particularly suitable for load bearing tissue engineering

applications since the fully interconnected network of scaffold fibres can

withstand early physiological and mechanical stress in a manner similar to

cancellous bone [169]. Moreover, the architectural pattern allows retaining of

coagulating blood during the early phase of healing, and bone in-growth at

later stages. Prior to surgery, all scaffolds were surface treated for six hours

with 1M NaOH and washed five times with PBS to render the scaffold more

hydrophillic. Scaffold sterilization was achieved by incubation in 70% ethanol

for 5 min and UV irradiation for 30 min.

Page 170: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

146

Fig. 1: 3D !CT reconstructions of a cylindrical mPCL-TCP scaffold produced via

fused deposition modelling with an outer diameter of 2 cm and a height of 3 cm.

Scaffold parameters include a porosity of 70%, fully interconnected pores of a

diameter of 350-500 !m, and a 0/90˚ lay down pattern. Top view (A) and lateral view

(B).

To load the scaffolds with OP-1 implant 3.5 ml of sterile saline were added to

one vial containing 3.5 mg recombinant human BMP-7 and 1 g of bovine

type I collagen and thoroughly mixed. Then, the resulting putty was

transferred to the inner duct of the scaffold and the contact interfaces

between bone and scaffold (Fig. 2).

Fig. 2: To load the scaffolds with OP-1, the lyophilized protein was mixed with 3.5 ml

of sterile saline (A, B) and transferred to the inner duct of the scaffold and onto the

contact interfaces between bone and scaffold (C, D).

Page 171: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

147

Animal surgery

Anaesthesia and pre-operative treatment

A jugular venous line was installed under aseptic conditions, and 15-20 ml of

1% propofol were injected to induce general anesthesia. The respective

animal was then intubated with a 9-10 mm silicon endotracheal tube and

connected to an automatic respirator (Campbell anaesthetic ventilator) for

assisted ventilation with 2l O2/min. The general anesthesia was maintained

with propofol at a rate of 120-140 ml/min. For analgesia, Buprenorphine (0.1

mg per 10 kg body weight) was administered, for antibiotic prophylaxis

gentamycine (5 mg/kg) and cephalothin (25 mg/kg). The animal's ECG, heart

rate, oxygen saturation and end-tidal carbon dioxide levels were monitored

and recorded continuously.

Defect model

Animals (Merino sheep, average weight: 42.5 kg, age: 6-7 years) were

placed in right lateral recumbency. The right hindlimb was carefully shaved

and thoroughly desinfected with 0.5% chlorhexidine solution red in 70 %

ethanol. The animal torso and surroundings were then covered with sterile

sheets, the surgical area additionally with Opsite (Smith and Nephew). The

right tibia was exposed by a longitudinal incision of approximately 12 cm

length on the medial aspect of the limb. A bone fixation plate (4.5 mm broad

DCP, 10 holes, Synthes) was adjusted to the morphology of the bone by

bending (plate-bending press, Synthes) and applied to the medial tibia. The

distal end of all plates was placed exactly 2.5 cm proximal of the medial

Page 172: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

148

malleolus. The screw holes were drilled and the plate was fixed temporarily

with 2 screws adjcacent to the anticipated defect (Fig. 3 A).

The middle of the defect site was measured and marked with a raspatory;

afterwards, the plate was removed. A distance of 1.5 cm each proxiamally

and distally of the defect middle was measured and marked to define the

osteotomy lines (Fig. 3 B, C). Next, the soft tissue inserting to the bone in the

designated defect area was detached and a wet compress was placed

between bone and posterolateral soft tissue to avoid damage to proximate

nerve and blood vessels during osteotomy (Fig. 3 D). Parallel oteotomies

perpendicular to the bone’s longitudinal axis were performed with an

oscilating saw (Stryker) under constant irrigation with saline solution to

prevent heat induced osteonecrosis whilst the bone segment of 3 cm length

was excised (Fig. 3 E). Care was taken to completely remove the periosteum

within the defect area and 1.5 cm proximally and distally of the osteotomy

lines (Fig. 3 F). The bone fragments were realligned and fixed applying the

dynamic compression plate (DCP) with 4 screws proximally and 3 screws

distally to leave a defect gap of exactly 3 cm size (Fig. 3 E). The wound was

closed in layers with a 2-0 Monocryl (Ethicon) and a 3-0 Novafil (Syneture)

suture for the skin. The closed wound was sprayed with Opsite (Smith and

Nephew), covered with pads and bandaged (Vetrap, 3M). After recovery

from anaesthesia, animals were allowed unrestricted weight bearing.

Page 173: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

149

Fig. 3: To create a 3 cm segmental defect, the plate was temporarily fixed with two

screws (A), the screw holes were drilled, the defect middle and osteotomy lines

were marked (B, C), and the bone segment was removed after osteotomy (D, E).

Care was taken to remove the periosteum (F) before the bone fragments were

realigned and fixed with plate and screws (G).

Page 174: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

150

Harvest of autologous cancellous bone graft

Autologous, cancellous bone graft was harvested from the left iliac crest. The

surgical area was shaved and desinfected with 0.5% chlorhexidine red in

70% ethanol. A 5 cm incision was made following the iliac crest (Fig. 4 A),

the inserting musculature was carefully detached and the cortical bone of the

lateral os ileum was fenestrated (2 x 2 cm) using a hammer and osteotome

(Fig. 4 B). Care was taken not to fracture the ala ossis ilii. The resulting lid

was carefully removed with a raspatory (Fig. 4 C) and the cancellous bone

harvested utilizing a bone curette (Fig. 4 D). The lid was then reinserted (Fig.

4 E), and the musculature reattached with 2-0 Vicryl sutures (Ethicon), and

the wound closed in layers (Fig. 4 F). The closed wound was sprayed with

Opsite (Smith and Nephew).

Fig. 4: The figure illustrates the harvesting procedure for autologous bone grafts

with the incision (A), the fenestration of the os ilium (B), removal of the lid (C),

harvested graft (D), lid reinsertion (E) and wound closure (F).

Page 175: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

151

Experimental groups

Experimental groups of the ovine segmental bone defect study included

empty control defects as a negative control, defects reconstructed with

autologous cancellous bone graft from the iliac crest as a positive control,

defects reconstructed with mPCL-TCP scaffolds and defects augmented with

mPCL-TCP scaffolds and 3.5 mg of OP-1 (OP-1 Implant, Stryker)(Table 1).

OP-1 or recombinant human bone morphogenetic protein 7 (rhBMP-7) was

introduced as an additional, biologically active, osteoinductive element.

Table 1: Experimental groups included in the ovine segmental bone defect study.

Euthanasia

The animals were euthanized by intravenous injection of 60 mg/kg

pentobarbital sodium (Lethabarb, Virbac, Australia). After euthanasia, both

hind limbs were exarticulated at the knee and the tibia was dissected and the

implanted osteosynthetic material was removed. The samples were then

processed and analysed.

Page 176: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

152

Radiographic analysis

Immediately after surgery, after 6 and 12 weeks, conventional x-ray analysis

(3.2 mAs; 65 kV) in two standard planes (anterior-posterior and medial-

lateral) was performed to assess bone formation.

Computed tomography

After sacrifice, a clinical CT scanner (Philips Brilliant CT 64 channels) was

used to scan the experimental limbs. A dipotassium phosphate phantom was

used to calibrate measurements of mineral density.

3D reconstructions from the CT data were generated with AMIRA® 5.2.2

(Visage Imaging GmbH, Berlin, Germany) (Visage Imaging) with a threshold

of 300 and qualitative analysis was performed to assess mineralization within

the defect and bridging. A scoring system (Fig. 5) was developed to describe

callus formation within the defect zone (1 point for bone formation in the

proximal defect half, 1 point for bone formation in the distal defect half),

external callus formation (1 point for external callus in the proximal, 1 for

external callus in the distal defect half; additional 2 points for external callus

in more than one location), and defect bridging (2 points).

Page 177: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

153

Fig. 5: 3D reconstruction of a 3 cm defect after 12 weeks overlayed with the scoring

system

For quantitative analysis, the CT datasets of operated and contralateral intact

tibia of each animal were first cropped to image stacks with equal bounding

box dimensions using AMIRA®. Next, cortical bone and callus tissue were

segmented by choosing appropriate threshold values (lower threshold: 300)

for the measured grey levels. A 3D surface was generated and saved as a

binary file (STL binary Little Endian format). These .stl files were loaded into

Rapidform2006 (Inus Technology, Seoul, Korea) and a minimum of 4

corresponding reference points was selected on each intact and defect tibia

and bound to the respective shell. Intact and defect tibia were then registered

to align their shells utilizing the previously defined common geometries

between them. The reference point coordinates of the defect tibia were

recorded prior to and after registration. The coordinates of the initial and final

points were entered in an in-house MATLAB program (MATLAB 7.6.0,

Page 178: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

154

MathWorks, Inc., Natick, MA, USA) to determine the matrix for the required

transformation. This transformation matrix was then used to align the image

data stacks in AMIRA®. This alignment resulted in intact and defect tibia to

have the same orientation. Next, the amount of newly formed bone in three

defined regions/volumes of interest within the 3 cm defect area was

calculated utilizing an in-house MATLAB program (Fig. 6).

Fig. 6: CT DICOM image of an intact ovine tibia (axial view) defining the three

regions of interest.

Biomechanical testing

Both ends of the tibiae were embedded in 80 ml Paladur (Heraeus-Kulzer

GmbH) and mounted in an Instron 8874 biaxial testing machine (Fig. 7). By

leaving as much soft tissue as possible attached, bone samples were

prevented from drying out.

A torsion test was conducted at an angular velocity of 0.5 deg/s and a

compressive load of 0.05 kN until the fracture point was reached (right tibiae

counter clockwise, left tibiae clockwise). The contralateral tibia was used as

a paired reference. The torsional moment (TM) and torsional stiffness (TS)

Page 179: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

155

values were calculated from the slope of the torque-angular displacement

curves and normalized against the values of the intact contralateral tibiae.

Fig. 7: Potting of a sample tibia for biomechanical testing. First, the proximal part

was embedded in Paladur (A) with the tibial axis vertically aligned. The tibia was

then rotated (B) and the distal part was embedded (C).

!CT analysis

For !CT analysis, a region of interest including the 30 mm defect gap and 5

mm of adjacent bone each proximally and distally was selected. Samples

were scanned (vivaCT 40, Scanco medical) with a voxel size of 19.5 !m.

Samples were evaluated at a threshold of 220, a filter width of 0.8 and filter

support of 1.0 and analysed for bone volume, bone mineral density, and

trabecular thickness within the defect using the software supplied by the

manufacturer of the !CT. In addition, the polar moment of inertia (pMOI) in

m4 was calculated according to the following formula: Jz = ! "! where Jz

represents the polar moment of intertia about an axis z, dA and elemental

area, and # the radial distance to the element dA from the axis z [248] (Fig.

Page 180: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

156

8). The pMOI represents a quantity used to predict an object’s ability to resist

torsion.

Fig. 8: A schematic showing how the polar moment of inertia is calculated for an

arbitrary shape about an axis o. # is the radial distance to the element dA.

Page 181: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

157

Histology

After biomechanical testing, specimens were trimmed to 15 cm length and

fixed in 10% neutral buffered formalin. For histological analysis, the mid-

defect regions were sectioned in the transversal and sagittal plane (Fig. 9).

Callus tissue composition was evaluated on 6 µm-thick methylmethacrylate

(Technovit 9100 NEU, Heraeus Kulzer, Germany) embedded sections

(embedding procedure according to the manufacturer’s protocol), stained

with Safranin Orange/von Kossa (mineralized tissue, black), and Movat’s

pentachrome [249] to demonstrate bone (yellow), cartilage (deep green) and

fibrous tissue (light green-blue).

Fig. 9: The schematic illustrates the frontal and sagittal cutting planes used for

histological sectioning and their spatial arrangement in respect to the defect area.

Statistical analysis

Statistical analysis was carried out using a two-tailed Mann-Whitney-U-test

(SPSS 16.0, SPSS Inc.) and p-values were adjusted according to Bonferroni-

Holm. Sample size for the study was determined with a power analysis

performed on the torsional strength data reported for a comparable critical

size defect study in sheep tibiae [93].

Page 182: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

158

Results

Animal model

A surgical technique was successfully developed to excise a 3 cm mid-

diaphyseal tibial bone segment without affecting surrounding soft tissue,

blood vessels and nerves. In addition, a single sided ABG harvesting

procedure was followed that ensured sufficient amounts of graft for defect

reconstruction. No postoperative infections or other complications were

observed. The chosen 4.5 mm broad DCP was proven to be biomechanically

sufficient to prevent implant failure. All animals were in good health and

survived the experimental period gaining weight in the months following

surgery. By ensuring complete removal of the periosteum within the defect

area, a defect model of critical nature could be established showing no

defect bridging in all animals included in the empty defect control groups.

X-ray analysis

Conventional X-ray analysis in two planes after 12 weeks confirmed the

critical nature of the defect. None of the empty control defects (n=8) showed

signs of external callus formation and only marginal bone formation within

the defect area. Only minor external callus and bone formation within the

defect was observed in the scaffold group. Callus was located mainly

postero-laterally. However, full defect bridging had occurred in all defects

reconstructed with ABG or mPCL-TCP with rhBMP-7. ABG treatment did not

result in substantial external callus formation, but development of new bone

within the defect. RhBMP-7 in combination with a mPCL-TCP scaffold had

stimulated both external callus formation (mainly postero-laterally) and bone

Page 183: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

159

formation within the defect gap (Fig. 10). In all groups, bone formation was

observed along the stainless steel plate.

Fig. 10: Representative x-ray images after 12 weeks of an empty control defect (A),

a defect reconstructed with cancellous bone graft from the iliac crest (B), a defect

augmented with mPCL-TCP + rhBMP-7 (C), and a defect treated with a mPCL-

TCP scaffold (D). The images show clear radiographic signs of defect bridging for

the autograft and rhBMP-7 group, with external callus formation in the rhBMP-7

group. No bone formation was observed within the empty control defect and only

little bone formation for the scaffold only group.

Page 184: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

160

Computed tomography

Qualitative CT analysis after 12 weeks confirmed the critical nature of the

defect showing non unions in all eight empty control defects, which were

filled with soft tissue only. Only minor bone formation was observed in the

mPCL-TPC scaffold group, mainly on the posterolateral side. However, full

defect bridging had occurred in all defects reconstructed with ABG or mPCL-

TCP + rhBMP-7 (Fig. 11). No radiographic signs of inflammation were found.

Scaffolds showed good osseointegration without any signs of resorption. The

qualitative CT score performed on 3D reconstructions (AMIRA 5.2.2,

threshold of 300) (Fig. 12) of all samples assessing bone and callus

formation showed significant differences between ABG (mean=6.625 out of 8

possible points) or mPCL-TCP + rhBMP-7 (mean=7.125) when compared

with the empty control (mean=3.25) or scaffold only group. Moreover, a

significant difference in bone formation between empty defect and scaffold

only group was found. No statistically significant difference was found

between ABG and mPCL-TCP + rhBMP-7 group (Fig. 13, 14).

Page 185: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

161

Fig. 11: Union rates (A) and qualitative CT score (B, box plot) assessing bone

formation within the defect, external callus formation and defect bridging (n=8).

Asterisks indicate statistical significance.

A

B

Page 186: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

162

Fig. 12: Representative 3D CT data reconstructions (AMIRA 5.2.2) of critical

segmental bone defects, which were left untreated (A), reconstructed with ABG (B),

an mPCL-TCP scaffold (C) or an mPCL-TCP scaffold combined with rhBMP-7 (D).

Median values of total bone volume (BV) in the defect area were significantly

higher in the rhBMP-7 group (8.6 cm3) than in all other groups (Fig. 10 A). In

the cortical region (region 1) no significant difference between rhBMP-7 (2.96

cm3) and ABG (2.09 cm3) group was found (Fig. 10 B). Bone formation in

region 2, the marrow region, was calculated to be significantly higher in the

ABG group (1.66 cm3) when compare to all other groups (Fig. 11 A). In

contrast to the total bone volume, bone formation in the periosteal region

(region 3) was significantly higher in both the scaffold (1.65 cm3) and rhBMP-

7 group (4.55 cm3) than in the ABG group (0.77 cm3)(Fig. 11 B).

A B C D

Page 187: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

163

Fig. 13: Box plot demonstrating the median ± 1st and 3rd quartile (n=8). The figure

illustrates the total bone volume (BV)(A) and bone volume formed in the cortical

region (region 1)(B) after 12 weeks. Asterisks indicate statistical significance. Error

bars represent minimum and maximum values.

A

B

Page 188: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

164

Fig. 14: Box plot (the median ± 1st and 3rd quartile) demonstrating the bone volume

formed within the marrow region (A) and periosteal region of the bone (B) after 12

weeks (n=8). Error bars represent minimum and maximum values. Asterisks

indicate statistical significance. Error bars represent minimum and maximum values.

A

B

Page 189: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

165

Biomechanical testing

None of the empty control defects showed signs of bridging and were filled

with soft tissue only. Therefore no biomechanical testing could be carried out

on these specimens. Biomechanical testing (Fig. 15 and 16) revealed a

significant higher torsional moment (TM) for the ABG (median=11.19 %) and

rhBMP-7 (14.41 %) groups when compared to the mPCL-TCP group (4.96

%). No significant difference was found between the ABG and rhBMP-7

group. Similar results were observed for torsional stiffness (TS) values with

significantly higher TS values for the ABG (19.317 %) and rhBMP-7 group

(25.043 %) when compared to the scaffold only group (2.535 %). Again no

statistically significant difference was found between the ABG and rhBMP-7

treated group. However, the rhBMP-7 treatment tended to result in higher

values for both torsional moment and stiffness.

Page 190: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

166

Fig. 15: Box plot demonstrating median values of torsional moment (A) ± 1st and 3rd

quartile and these values relative to the contralateral tibia (B)(n=8). Error bars

represent maximum and minimum values. Asterisks indicate statistical significance.

A

B

Page 191: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

167

Fig. 16: Box plot demonstrating median values of torsional stiffness (A) ± 1st and 3rd

quartile and these values relative to the contralateral tibia (B)(n=8). Error bars

represent maximum and minimum values. Asterisks indicate statistical significance.

A

B

Page 192: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

168

µCT analysis

MicroCT analysis confirmed results from the clinical CT scans regarding

union rates and the amount of new bone formation. All of eight defects in the

autograft and scaffold-rhBMP-7 group showed solid bony union after twelve

weeks (Fig. 17). Highest median values of newly formed bone were found for

the scaffold-rhBMP-7 group (median=2254.04 mm3). They did, however, not

differ significantly from values obtained for the positive control, the autografts

(median=2158.77 mm3), as a considerable variation in bone neoformation

was observed (minimum=1126.56 mm3, maximum=4863.87 mm3)(Fig. 18).

Fig. 17: MicroCT sections (A-D) of central defect portions and 3D reconstructions of

3 cm tibial defects (E-H) 12 weeks after surgery. Defects were left untreated (A, E),

reconstructed with autologous cancellous bone from the iliac crest (B, F), a mPCL-

TCP scaffold (C, G) or scaffold plus 3.5 mg rhBMP-7 (D, H). Defect bridging was

found in all specimens of each the autograft and rhBMP-7 group. Scaffolds showed

good osseointegration at the host bone interface.

Page 193: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

169

When compared to autograft or rhBMP-7 treated lesions, significantly lower

bone formation was observed in the defects treated with scaffolds only

(median=681.90 mm3) or untreated control defects (median=171.91)

mm3)(Fig. 18). In any case, newly formed bone was still less compared to the

amounts determined for the same anatomic level of the contralateral hind

limbs. The amount of new bone was evenly distributed throughout the

proximal, middle and distal third of the defect although a tendency towards

higher amounts in the proximal defect third was observed (Fig. 19).

The mineral density of the newly formed, woven bone or tissue was found to

be homogenous in the different experimental groups (Fig. 20). Notably,

bone/tissue mineral density values were significantly lower than those

determined for the compact bone of the contralateral healthy tibiae with

median values ranging from 71.37 to 76.34 %.

Analysis of trabecular thickness within the newly formed bone and the

distribution along the z axis showed significantly thicker bone trabeculae in

the proximal third of the ABG group (median=339.65 !m) when compared to

the defect middle (median=257.3 !m) and distal regions (median=246.7 !m)

or corresponding proximal defect segments of the groups involving mPCL-

TCP scaffolds (Fig. 21).

Values for the polar moment of inertia, which describes an object’s ability to

resist torsion, reflected the tendencies of the biomechanical testing results

and determined bone volumes (Fig. 22) and correlated well with these

findings (correlation coefficients: 0.911 and 0.896).

Page 194: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

170

Fig. 18: Box plot demonstrating median amounts of newly formed bone in mm3 ± 1st

and 3rd quartile within the 3 cm defects 12 weeks after surgery. Significantly more

bone formation was seen in defects reconstructed with either autograft (ABG) or

scaffold combined with rhBMP-7 (mPCL-TCP+rhBMP-7) when compared to scaffold

alone (mPCL-TCP) or untreated defects. Error bars represent maximum and

minimum values. Bars with asterisks indicate statistical significance.

Fig. 19: Box plot demonstrating the distribution of newly formed bone in mm3 along

the z axis of regenerated defects. A tendency towards higher amounts of bone in

the proximal defect third was observed throughout the different experimental

groups. Error bars represent maximum and minimum values.

Page 195: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

171

Fig. 20: Box plot demonstrating median values for tissue mineral density within the

defects ± 1st and 3rd quartile 12 weeks after surgery. Tissue density did not differ

significantly between the experimental groups. Tissue density in the defect zones

was however only about 70% of that determined for the compact bone of the

contralateral limb. Error bars represent maximum and minimum values.

Fig. 21: Box plot illustrating the distribution of trabecular thickness along the z axis

of regenerated defects. Significantly thicker bone trabeculae were observed in the

proximal third of the autograft (ABG) group when compared to the defect middle and

distal regions or proximal defect segments of the groups involving scaffolds.

Page 196: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

172

Fig. 22: Box plot demonstrating median values for the polar moment of inertia within

the defect region ± 1st and 3rd quartile 12 weeks after surgery (A) and these values

relative to the contralateral tibia (B). Error bars represent maximum and minimum

values.

A

B

Page 197: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

173

Histology

Von Kossa staining on 6 µm-thick methylmethacrylate embedded sections

showed extensive amounts of mineralized tissue (black) within defects of the

autograft and rhBMP-7 group. In these groups solid defect bridging had

occurred. Notably, bone formation originated predominately from endosteal

areas as the periosteum - which is an additional source of mesenchymal

progenitor cells capable of differentiating into bone synthesizing osteoblasts -

had been removed during surgery. Untreated defects showed hardly any

new bone, defects treated with mPCL-TCP scaffolds considerably less bone

formation (generally at the scaffold-compact bone interface) and mainly soft

tissue was identified within the defect areas. Movat’s pentachrome staining

revealed that the soft tissue in the empty defects consisted mainly of fibrous

connective tissue. In contrast, the soft tissue in the scaffold only group

mainly resembled cartilage like tissue.

Bone formation in both the autograft and rhBMP-7 group occurred via

endochondral ossification, in which a cartilage template is gradually replaced

by a bone matrix. Deposited bone represented unorganized

cancellous/trabecular bone with mature osteocytes embedded in

characteristic lacunae. Bone in the autograft bone appeared to be less

mature when compared to bone in the rhBMP-7 group as higher amounts of

osteoid (red) and mineralized cartilage (green) were evident. Osteoid refers

to the unmineralized, organic portion of the bone matrix that forms prior to

the maturation of bone tissue (Fig. 23-25).

Page 198: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

174

Fig. 23: Longitudinal native bone defect sections in the frontal plane (A-D)

embedded in PMMA and stained for Safranin O/von Kossa (E-H) and Movat’s

pentachrome (I-L) Von Kossa staining showed extensive amounts of mineralized

tissue (black) within defects of the autograft (F) and rhBMP-7 group (H) when

compared to untreated defects (E) or defects treated with mPCL-TCP scaffolds only

(G) where mainly soft tissue was identified (red). While the soft tissue in the empty

defects (I) consisted mainly of fibrous connective tissue (green), the soft tissue in

the scaffold only group (K) mainly resembled cartilage like tissue (light blue).

Page 199: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

175

Fig. 24: PMMA sections stained for Movat’s pentachrome showing high amounts of

fibrous tissue (FT) in the untreated defects (A). In the scaffold only group (C) large

proportions of cartilaginous tissue (CT, light blue) were present while bone

formation (BT) was only observed along the scaffold/host bone interface evidenced

by osteoid islets (red, arrows). Bone in the autograft treated defects (B) appeared

less compact and of lower maturity when compare to the rhBMP-7 group (D) as

higher amounts of unmineralized osteoid and calcified cartilage (light green) were

found.

Fig. 25: High magnification images of the fibrous connective tissue (CT) in empty

defects with characteristic, elongated fibroblasts embedded in randomly aligned

collagenous fibre bundles (A). Maturing bone tissue (BT) of the autograft group (B)

and rhBMP-7 group (D) with unmineralized osteoid (red, arrows), mineralized

cartilage (light green), mineralized bone matrix (yellow), and mature osteocytes

embedded in characteristic lacunae (arrow heads). Image C shows connective

tissue (CT) and osteoblasts embedded in unmineralized osteoid (red, arrows) as

found in the scaffold only group (interface bone/scaffold).

Page 200: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

176

Discussion

The present animal study was designed to investigate the effects of rhBMP-7

in combination with mPCL-TCP composite scaffolds built by computer aided

design using rapid prototyping technologies on bone healing. As a

prerequisite, a critical sized segmental bone defect model in ovine tibiae was

established showing a reliable non-union rate of 100%. Sheep as study

objects were chosen for their close analogies with human bone in terms of

remodelling and turnover as described in chapter I [126].

The transplantation of cancellous bone grafts represents the most frequently

chosen clinical treatment as these grafts possess osteoconductive and

osteoinductive properties. As a result, an experimental group in which

defects were treated with autologous bone graft from the iliac crest was

included as a positive control and benchmark.

A unilateral approach of the iliac crest to harvest graft was proven to be

sufficient to yield adequate amounts of graft for defect reconstruction. The

compressive strength of cancellous bone ranges between 2 and 20 MPa

depending on localization [250], however, the compressive strength of a

cancellous graft is considerably less (1-2 MPa) and therefore insufficient to

provide significant mechanical support [251]. Consequently, the required

stability to provide a loading environment facilitating bone healing has to be

provided by the osteosynthesis material. To minimize the risk of subsequent

implant failure, only partial weight bearing of the affected bone is hence

advisable until partial healing can be observed [4].

Bone graft substitutes that are capable of partially bearing loads and

disburden the osteosynthetic implants are therefore desirable and would

Page 201: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

177

allow earlier load bearing whilst additionally stimulating the bone healing

process.

Bone graft substitutes for bone defect augmentation should consist of

materials that provide structural integrity and have a space-occupying effect

in order to maintain a proper reduction and continuity between the bone

fragments during surgery and to enable an adequate osteosynthesis to be

performed. Bioresorbable aliphatic polyesters, such as polyglycolide,

polylactide, polycaprolactone (PCL), and their copolymers, are suitable

materials for the design and fabrication of biocompatible scaffolds owing to

their significant track record for regulatory approval, and minimal

inflammatory and immunological responses evoked. As such, they have

been used as components in a plethora of devices for clinical applications

[252, 253]. These materials offer favourable surface chemistries for cell

attachment, proliferation, and differentiation, while degradation by-products

are nontoxic and metabolized/eliminated via natural pathways. Most

importantly, these thermoplastic polymers can easily be processed into

three-dimensional scaffolds with desired geometry, controlled porosity and

interconnectivity by applying modern computer-based solid free-form

fabrication methods [254]. These fabrication methods may therefore allow

the design of scaffolds with biomechanical properties similar to those of

cancellous bone [106].

For the present study, bioresorbable scaffolds of medical grade )-poly-

caprolactone incorporating 20% "-tricalcium phosphate (mPCL–TCP) were

produced by fused deposition modelling (FDM) as described previously

[169]. The structural parameters of the scaffolds were tailored by computer

Page 202: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

178

aided design and included 100% pore interconnectivity within a range of

350–500 µm size, 70% scaffold porosity, and a 0/90° lay down pattern. This

architectural layout is particularly suitable for load bearing tissue engineering

applications since the fully interconnected network of scaffold fibres can

withstand early physiological and mechanical stress in a manner similar to

cancellous bone [169]. Moreover, the architectural pattern allows retaining of

coagulating blood during the early phase of healing, and bone in-growth at

later stages.

Importantly, scaffold handling during surgery was uncomplicated and

transplantation of these ready-to-use mPCL-TCP composites easily

achievable. As a result, economically, the transplantation of scaffolds

represents a time and therefore cost-effective alternative. Lohmann et al.

concluded that costs associated with the application of commercially

available bone replacement materials are comparable to those related to

bone grafts or may be even lower as high complication rates at the donor site

can significantly increase the total-costs-of-illness in the case of autografts

[255].

In animals where mPCL-TCP scaffolds were transplanted into the defects, no

signs of foreign body reactions to the transplant were observed underlining

their good biocompatibility. As evaluated by visual macroscopic assessment,

!CT analysis and histology, scaffolds were of structural integrity and had

integrated well at the host bone-scaffold interface. After 12 weeks no signs of

scaffold resorption were evident. Non-surprisingly, as biologically inactive,

cell-free materials, the scaffold type under investigation did not perform as

well as the gold standard autograft treatment as significantly lower values

Page 203: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

179

were determined for torsional moment and stiffness as well as the

parameters describing new bone formation.

To introduce a biologically active, osteoinductive element, the scaffolds were

combined with the bone growth stimulating agent recombinant human bone

morphogenetic protein 7 (rhBMP-7) [73, 185]. Previous reports had shown

that rhBMP-7 is an efficient adjuvant in the healing of segmental bone

defects [4].

Bone morphogenetic proteins are a well-studied group of growth factors

involved in the processes of bone healing; and the human genome encodes

at least 20 of these multifunctional polypeptides [240]. However, only a

subset of BMPs, most notably BMP-2, -4, -7, and -9, have osteoinductive

activity [241]. The rhBMP-7 was provided in the form of Stryker®’s OP-1

implant which consists of 3.5 mg of rhBMP-7 (or Osteogenic Protein 1, OP-

1), formulated with 1 g purified bovine type I collagen carrier. The product is

commonly reconstituted with 2-3 cc of saline to form a paste which is then

implanted. Once implanted, rhBMP-7 stimulates natural bone healing by

actively recruiting stem cells from surrounding tissue and blood supply,

initiating the bone formation cascade [256]. OP-1 Implant is approved by the

FDA under a Humanitarian Device Exemption and is indicated for use as an

alternative to autograft in recalcitrant long bone non-unions where use of

autograft is unfeasible and alternative treatments have failed. Materials such

as the OP-1 implant do not provide any structural support as they are (semi-)

liquid materials. Therefore, the combination with a biocompatible scaffold of

suitable biomechanical properties appears appealing.

Page 204: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

180

Importantly, rhBMP-7 should, however, not be used to treat patients who

have a known hypersensitivity to any of the components of the product

(about 18% of all patients develop antibodies to bovine type I collagen).

rhBMP-7 should not be applied at or near the vicinity of a resected tumour or

in patients with a history of malignancy and should also not be administered

to patients who are skeletally immature (<18 years of age or no radiographic

evidence of closure of epiphyses). As the potential effects of rhBMP-7

treatment on the human foetus have not been evaluated rhBMP-7 should not

be administered to pregnant women either. Studies in rats injected with high

doses of rhBMP-7 have shown that small amounts of rhBMP-7 will cross the

placental barrier. It has furthermore been reported that antibody formation to

rhBMP-7 occurs in up to 38% of patients the percentage of neutralizing

antibodies to rhBMP-7 was not determined separately. The clinical

significance of these antibodies, however, remains unclear [257].

Successful union requires rhBMP-7 to be retained at the surgical site long

enough to achieve osteoinduction. It is, however, unclear whether union

enhancement depends on the action of carrier-bound BMP or of freely

diffusible BMP slowly released from the carrier [258]. Nevertheless,

elucidation of retention kinetics may aid evaluation and development of BMP

delivery systems to optimize clinical outcomes. The mean residence time for

rhBMP-7 collagen putty was determined to be 10.4 ± 2.7 days in a spinal

fusion model (New Zealand White rabbits) [259]. These excretion profiles

and kinetic properties are similar to those described for rhBMP-2 (mean

residence times of 7.6 - 10.2 days) and include a biphasic release profile.

Page 205: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

181

The initial burst release correlates with protein solubility, whereas the gradual

secondary release is governed by carrier properties [260, 261].

Regardless of the exact local pharmacokinetics, in the present study, the

release pattern of rhBMP-7 from the administered collagen particles and the

mPCL-TCP scaffold appeared to be consistent with the requirements of the

healing bone, as became evident by efficient healing of the defects in this

group. Biomechanical testing, histology, CT and !CT analysis consistently

showed comparable, in tendency even higher values and volumes of newly

formed bone in rhBMP-7 treated defects when compared to autograft

controls. Significant differences between rhBMP-7 and autograft group could,

however, not always be determined as the variation of bone formation within

the rhBMP-7 group was considerable which amongst other factors could be

due to variations of the local mechanical environment, in local pH, in

composition of the defect haematoma, surgical technique, release kinetics,

and the concentration of local connective tissue progenitor cells or degree of

vascularisation [262, 263].

All experimental studies utilizing rhBMP-7 as a bone growth stimulating

adjuvant illustrate a dose dependent effect and a species dependent

response variation [262, 264]. Maintaining a critical threshold concentration

of the rhBMP at the defect site for the necessary period of time (temporal

distribution) is crucial. Such supra-physiological dosages range from

0.01 mg/ml in small animal models such as rats to 0.4 mg/ml in rabbits to

more than 1.5 mg/ml in non-human primates. Different anatomical sites

require different therapeutic doses depending on the degree of

Page 206: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

182

vascularisation, defect size and the number of resident responding cells.

Based on literature reports, in the present study, a standard application of

3.5 mg rhBMP-7 was chosen [265, 266]. The recommended dose of rhBMP-

7 for recalcitrant long bone non-unions in humans, however, is 7 mg or 2

vials (each containing 3.5 mg reconstituted with 1 g of type I bovine

collagen)[267].

Despite ample evidence of the benefit of BMPs in bone regeneration and

repair from preclinical and clinical studies, conclusive knowledge about BMP

dosage, time-course, release dynamics and carriers remains to be

determined. It is unclear why the impressive and convincing results seen in

vitro and in animal models are so far difficult to reproduce reliably in humans

[268]. Unfavourable release kinetics, insufficient mechanical scaffold stability

and porosity to allow cell and blood vessel infiltration into the carrier and

inflammatory tissue reactions might be few reasons. New delivery systems

with optimized and controlled release profiles may hence decrease or even

alleviate the need for excessive and expensive concentrations of BMPs.

As in the present study, segmental bone defects usually heal via indirect

repair mechanisms referred to as endochondral ossification. This process

involves the recruitment, proliferation, and differentiation of mesenchymal

progenitor cells into cartilage, which subsequently becomes calcified and

eventually is replaced by bone. The repair process is comprised of four

overlapping phases initiated by an immediate inflammatory response that

leads to the recruitment of mesenchymal progenitor cells and subsequent

differentiation into chondrocytes that produce cartilage and osteoblasts,

Page 207: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

183

which form bone. After cartilage matrix is produced, it mineralizes, and a

transition from mineralized cartilage to bone occurs, initiated by the

resorption of mineralized cartilage [269]. It is the eventual bridging of hard

callus areas across the central defect gap that provides the initial

stabilization and regain of biomechanical function [270]. Primary bone

formation is followed by remodelling, in which the initial bony callus is

reshaped by secondary bone formation and resorption to restore the

anatomical structure that supports mechanical loads [271].

The current study results suggested not only a tendency towards increased

bone formation in defects treated with rhBMP-7 but also stimulated callus

maturation as evidenced by fewer amounts of osteoid and mineralized

cartilage in this group compared to autograft treated defects. These findings

could be attributed to rhBMP-7 accelerating the molecular events associated

with defect healing. Bone defect repair recapitulates the molecular pathways

of normal embryonic development with the coordinated participation of

several cell types originating from the cortex, periosteum, surrounding soft

tissue, and bone marrow space. The signalling molecules regulating this

process include pro-inflammatory cytokines, the transforming growth factor-

beta (TGF-") superfamily and other growth factors, and the angiogenic

factors [271, 272]. The transforming growth factor-beta (TGF-ß) superfamily

consists of a large number of growth and differentiation factors that include

bone morphogenetic proteins (BMPs), transforming growth factor beta (TGF-

ß), growth differentiation factors (GDFs), activins, inhibins, and Mullerian

inhibiting substance. Specific members of this family - such as BMPs (2-8),

GDF (1, 5, 8 and 10), and TGF-ß 1-3 - promote various stages of

Page 208: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

184

intramembranous and endochondral bone ossification during bone healing

[273].

Recent studies demonstrate that BMP-7 specifically induces the expression

of numerous growth factors and multiple members of the BMP family

(including autoinduction), during the bone induction process [274, 275].

These factors then guide the bone formation process to completion.

Expression of BMP-7 was found to be enhanced in sites of endochondral

ossification about one week after fracture and it was therefore suggested that

BMP-7 acts predominately in the early stage of fracture healing [276]. In vitro

studies have shown that rhBMP-7 perpetuates mechanisms involved in bone

formation, including the promotion of chondrocyte maturation (extracellular

matrix component production) in normal articular chondrocytes, the

enhancement of osteoblastic characteristics (alkaline phosphate production)

of normal osteoblast cells, and the induction of the differentiation of

osteoclasts involved in the bone remodelling process. The end result of this

differentiation cascade is the production of weight-bearing bone with fully

functional bone marrow elements [277].

Considering the role of BMP-7 in mainly early stages of defect healing

together with the described release kinetics of BMP from collagen carriers,

and the favourable properties of the applied scaffolds, one could conclude

that in the present study, the requirements - based on the current knowledge

surrounding bone defect healing - have been met as closely as possible to

produce a suitable tissue engineered bone graft substitute.

Page 209: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

185

Summary

In the current study, a standardized and reproducible ovine, tibial, critical-

sized defect model was developed as reflected by the non-union rate of

100% in the empty control group. The application of autografts from the iliac

crest caused defect bridging in all cases. When compared to intact controls,

the mechanical properties of the newly formed bone however were of inferior

quality after 3 months. Medical grade PCL-TCP scaffolds did not evoke

foreign body responses. The mPCL-TCP scaffolds alone did not result in any

substantial mineralization of the defect area. However, when combined with

rhBMP-7, bone formation within the defect equalled or even exceeded

amounts observed with autografts. The study results suggest that mPCL-

TCP scaffolds combined with a biologically active stimulus such as rhBMP-7

can serve as an equivalent alternative to autologous bone grafting in the

early phase of defect regeneration. These findings however must be

confirmed by long-term studies.

Page 210: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

186

Page 211: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

187

Conclusions and Recommendations

The detailed characterisation of cells involved in bone regeneration

associated processes is of utmost importance. Since sheep represent a

valuable model for human bone turnover and remodelling, data

characterising cells derived from these animals are of special interest. Ovine

marrow derived mesenchymal progenitor cells and cortical bone osteoblasts

exhibit morphological, immunophenotypical and multipotential characteristics

similar to those in humans, which underlines the value of sheep as a model

species. Unfortunately, available methods of analysis are limited as ovine

genes are only partially sequenced and very few antibodies specific for and

cross-reacting with equivalent sheep antigens are available. If further insight

into fundamental processes such as haematopoiesis, cell migration and

homing, injury repair, differentiation, and proliferation is to be provided, these

shortcomings need to be addressed in the future.

In vitro experiments identified mesenchymal progenitor cells reproducibly

displaying a higher osteogenic potential. In vivo, however, osteoblasts

exhibited a higher potential to form new bone. These results emphasize the

difficulties in extrapolating in vitro findings to in vivo settings and suggest that

osteoblasts isolated from compact bone possibly represent a suitable

alternative cell population for cell based tissue engineering applications.

When comparing the osteogenic potential of these cells after transplantation

in vivo, subcuntaneously transplanted cells showed a high degree of survival

and actively contribute to endochondral osteogenesis. Endochondral bone

Page 212: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

188

formation, which describes a process in which a cartilage template is

gradually replaced by a bone matrix. It can also be observed in orthotopic

segmental defect sites. When compared to mesenchymal progenitor cells,

osteoblasts deposited higher amounts of new bone while osteoblast derived

bone was of higher maturation. Cell stimulation with rhBMP-7 increased the

rate of bone synthesis for both cell types and positively affected

neovascularisation and osteoclast activity. These results suggest that origin

and commitment of transplanted cells can determine type and degree of

ossification. They furthermore confirm that rhBMP-7 represents a potent

adjuvant stimulating bone formation.

It needs to be emphasized that microenvironmental conditions in ectopic

transplantation sites, again, may not be representative of specific cues cells

experience in a large segmental bone defect. However, it could also be

argued, that in such a defect cells are - similarly to ectopic sites - surrounded

by mainly soft tissue types rather than bone. Although an essential first step

was taken towards the characterization of ovine mesenchymal progenitor

cells and osteoblasts, essentially, further studies are required to verify these

findings in orthotopic models.

In the present study, the most important issues related to the establishment

of a large preclinical model for segmental bone defect research have been

addressed and discussed, and it was demonstrated how to develop such a

model. Regarding bench to bedside translations, an important milestone was

achieved in establishing a highly reproducible large animal model as an

Page 213: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

189

essential prerequisite to systematically assess different bone grafting

materials, scaffolds, tissue engineered constructs and growth factors.

The performance of a novel tissue engineered construct was finally assessed

in a large animal model and compared to the standard autograft

transplantation. The application of autografts from the iliac crest caused

defect bridging in all cases. When compared to intact contralateral controls,

the mechanical properties of the newly formed bone, however, were of

inferior quality. Transplanted scaffolds showed good biocompatibility and did

not evoke foreign body responses. They did, however, not result in any

substantial mineralization of the defect area. However, when combined with

BMP-7, bone formation within the defects equalled or even exceeded

amounts observed with autografts. Therefore, the study results suggest that

these scaffolds combined with a biologically active stimulus such as BMP-7

can serve as an equivalent alternative to autologous bone grafting in the

early phase of defect regeneration. These findings however must be

confirmed by long-term studies (12-24 months) that also assess the events

surrounding scaffold degradation and bone remodelling. Lastly, if this novel

and promising technology is to be translated into a routine clinical application

with predictable outcomes stringent treatment indications/contraindications

need to be formulated and detailed dose-effect and dose-response

relationships determined taking into account variables such as defect size

and cause or patient age.

Page 214: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

190

Page 215: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

191

Bibliography

1. Perka C, Schultz O, Spitzer RS, Lindenhayn K, Burmester GR, Sittinger M. Segmental bone repair by tissue-engineered periosteal cell transplants with bioresorbable fleece and fibrin scaffolds in rabbits. Biomaterials 2000;21(11):1145-1153. 2. Komaki H, Tanaka T, Chazono M, Kikuchi T. Repair of segmental bone defects in rabbit tibiae using a complex of beta-tricalcium phosphate, type I collagen, and fibroblast growth factor-2. Biomaterials 2006;27(29):5118-5126. 3. Gugala Z, Gogolewski S. Healing of critical-size segmental bone defects in the sheep tibiae using bioresorbable polylactide membranes. Injury 2002;33 Suppl 2:B71-76. 4. den Boer FC, Wippermann BW, Blokhuis TJ, Patka P, Bakker FC, Haarman HJ. Healing of segmental bone defects with granular porous hydroxyapatite augmented with recombinant human osteogenic protein-1 or autologous bone marrow. J Orthop Res 2003;21(3):521-528. 5. Laurencin C, Khan Y, El-Amin SF. Bone graft substitutes. Expert review of medical devices 2006;3(1):49-57. 6. Wildemann B, Kadow-Romacker A, Pruss A, Haas NP, Schmidmaier G. Quantification of growth factors in allogenic bone grafts extracted with three different methods. Cell and tissue banking 2007;8(2):107-114. 7. Perry CR. Bone repair techniques, bone graft, and bone graft substitutes. Clin Orthop Relat Res 1999(360):71-86. 8. DeCoster TA, Gehlert RJ, Mikola EA, Pirela-Cruz MA. Management of posttraumatic segmental bone defects. J Am Acad Orthop Surg 2004;12(1):28-38. 9. Clements JR, Carpenter BB, Pourciau JK. Treating segmental bone defects: a new technique. J Foot Ankle Surg 2008;47(4):350-356. 10. Lane JM, Tomin E, Bostrom MP. Biosynthetic bone grafting. Clin Orthop Relat Res 1999(367 Suppl):S107-117. 11. Einhorn TA, Lane JM, Burstein AH, Kopman CR, Vigorita VJ. The healing of segmental bone defects induced by demineralized bone matrix. A radiographic and biomechanical study. J Bone Joint Surg Am 1984;66(2):274-279. 12. Liu G, Zhao L, Zhang W, Cui L, Liu W, Cao Y. Repair of goat tibial defects with bone marrow stromal cells and beta-tricalcium phosphate. J Mater Sci Mater Med 2008;19(6):2367-2376.

Page 216: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

192

13. Blokhuis TJ, Wippermann BW, den Boer FC, van Lingen A, Patka P, Bakker FC, et al. Resorbable calcium phosphate particles as a carrier material for bone marrow in an ovine segmental defect. J Biomed Mater Res 2000;51(3):369-375. 14. Gao TJ, Lindholm TS, Kommonen B, Ragni P, Paronzini A, Lindholm TC, et al. Enhanced healing of segmental tibial defects in sheep by a composite bone substitute composed of tricalcium phosphate cylinder, bone morphogenetic protein, and type IV collagen. J Biomed Mater Res 1996;32(4):505-512. 15. Theos C, Koulouvaris P, Kottakis S, Demertzis N. Reconstruction of tibia defects by ipsilateral vascularized fibula transposition. Arch Orthop Trauma Surg 2008;128(2):179-184. 16. Sheller MR, Crowther RS, Kinney JH, Yang J, Di Jorio S, Breunig T, et al. Repair of rabbit segmental defects with the thrombin peptide, TP508. J Orthop Res 2004;22(5):1094-1099. 17. Bucholz RW, Carlton A, Holmes R. Interporous hydroxyapatite as a bone graft substitute in tibial plateau fractures. Clin Orthop Relat Res 1989(240):53-62. 18. Oest ME, Dupont KM, Kong HJ, Mooney DJ, Guldberg RE. Quantitative assessment of scaffold and growth factor-mediated repair of critically sized bone defects. J Orthop Res 2007;25(7):941-950. 19. Stevenson S. Enhancement of fracture healing with autogenous and allogeneic bone grafts. Clin Orthop Relat Res 1998(355 Suppl):S239-246. 20. Sciadini MF, Dawson JM, Johnson KD. Evaluation of bovine-derived bone protein with a natural coral carrier as a bone-graft substitute in a canine segmental defect model. J Orthop Res 1997;15(6):844-857. 21. Younger EM, Chapman MW. Morbidity at bone graft donor sites. J Orthop Trauma 1989;3(3):192-195. 22. Taylor GI, Miller GD, Ham FJ. The free vascularized bone graft. A clinical extension of microvascular techniques. Plast Reconstr Surg 1975;55(5):533-544. 23. Dell PC, Burchardt H, Glowczewskie FP, Jr. A roentgenographic, biomechanical, and histological evaluation of vascularized and non-vascularized segmental fibular canine autografts. J Bone Joint Surg Am 1985;67(1):105-112. 24. Lindsey RW, Gugala Z, Milne E, Sun M, Gannon FH, Latta LL. The efficacy of cylindrical titanium mesh cage for the reconstruction of a critical-

Page 217: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

193

size canine segmental femoral diaphyseal defect. J Orthop Res 2006;24(7):1438-1453. 25. Puelacher WC, Vacanti JP, Ferraro NF, Schloo B, Vacanti CA. Femoral shaft reconstruction using tissue-engineered growth of bone. Int J Oral Maxillofac Surg 1996;25(3):223-228. 26. Gazdag AR, Lane JM, Glaser D, Forster RA. Alternatives to Autogenous Bone Graft: Efficacy and Indications. J Am Acad Orthop Surg 1995;3(1):1-8. 27. Muscolo DL, Ayerza MA, Aponte-Tinao LA. Massive allograft use in orthopedic oncology. Orthop Clin North Am 2006;37(1):65-74. 28. Chapman MW, Bucholz R, Cornell C. Treatment of acute fractures with a collagen-calcium phosphate graft material. A randomized clinical trial. J Bone Joint Surg Am 1997;79(4):495-502. 29. Cacchioli A, Spaggiari B, Ravanetti F, Martini FM, Borghetti P, Gabbi C. The critical sized bone defect: morphological study of bone healing. Ann Fac Medic Vet di Parma 2006;26:97-110. 30. Cierny G, 3rd, Zorn KE. Segmental tibial defects. Comparing conventional and Ilizarov methodologies. Clin Orthop Relat Res 1994(301):118-123. 31. Ilizarov GA. The tension-stress effect on the genesis and growth of tissues: Part II. The influence of the rate and frequency of distraction. Clin Orthop Relat Res 1989(239):263-285. 32. Goldstrohm GL, Mears DC, Swartz WM. The results of 39 fractures complicated by major segmental bone loss and/or leg length discrepancy. The Journal of trauma 1984;24(1):50-58. 33. Gugala Z, Lindsey RW, Gogolewski S. New Approaches in the Treatment of Critical-Size Segmental Defects in Long Bones. Macromol Symp 2007;253:147–161. 34. O'Loughlin PF, Morr S, Bogunovic L, Kim AD, Park B, Lane JM. Selection and development of preclinical models in fracture-healing research. J Bone Joint Surg Am 2008;90 Suppl 1:79-84. 35. Einhorn TA. Clinically applied models of bone regeneration in tissue engineering research. Clin Orthop Relat Res 1999(367 Suppl):S59-67. 36. Rimondini L, Nicoli-Aldini N, Fini M, Guzzardella G, Tschon M, Giardino R. In vivo experimental study on bone regeneration in critical bone defects using an injectable biodegradable PLA/PGA copolymer. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2005;99(2):148-154.

Page 218: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

194

37. Gugala Z, Gogolewski S. Regeneration of segmental diaphyseal defects in sheep tibiae using resorbable polymeric membranes: a preliminary study. J Orthop Trauma 1999;13(3):187-195. 38. Tseng SS, Lee MA, Reddi AH. Nonunions and the potential of stem cells in fracture-healing. J Bone Joint Surg Am 2008;90 Suppl 1:92-98. 39. Egermann M, Goldhahn J, Schneider E. Animal models for fracture treatment in osteoporosis. Osteoporos Int 2005;16 Suppl 2:S129-138. 40. Liebschner MA. Biomechanical considerations of animal models used in tissue engineering of bone. Biomaterials 2004;25(9):1697-1714. 41. Schimandle JH, Boden SD. Spine update. The use of animal models to study spinal fusion. Spine 1994;19(17):1998-2006. 42. Pearce AI, Richards RG, Milz S, Schneider E, Pearce SG. Animal models for implant biomaterial research in bone: a review. Eur Cell Mater 2007;13:1-10. 43. Martini L, Fini M, Giavaresi G, Giardino R. Sheep model in orthopedic research: a literature review. Comparative medicine 2001;51(4):292-299. 44. Aerssens J, Boonen S, Lowet G, Dequeker J. Interspecies differences in bone composition, density, and quality: potential implications for in vivo bone research. Endocrinology 1998;139(2):663-670. 45. Gong JK, Arnold JS, Cohn SH. The Density of Organic and Volatile and Non-Volatile Inorganic Components of Bone. The Anatomical record 1964;149:319-324. 46. Wang X, Mabrey JD, Agrawal CM. An interspecies comparison of bone fracture properties. Bio-medical materials and engineering 1998;8(1):1-9. 47. Bloebaum RD, Ota DT, Skedros JG, Mantas JP. Comparison of human and canine external femoral morphologies in the context of total hip replacement. J Biomed Mater Res 1993;27(9):1149-1159. 48. Kaneps AJ, Stover SM, Lane NE. Changes in canine cortical and cancellous bone mechanical properties following immobilization and remobilization with exercise. Bone 1997;21(5):419-423. 49. Mittra EQ, Y-X. MATERIAL PROPERTIES OF SHEEP TRABECULAR BONE DETERMINED BY NANOMECHANICAL TESTING. IEEE 2003:229-230. 50. Mittra E, Rubin C, Qin YX. Interrelationship of trabecular mechanical and microstructural properties in sheep trabecular bone. J Biomech 2005;38(6):1229-1237.

Page 219: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

195

51. Nafei A, Danielsen CC, Linde F, Hvid I. Properties of growing trabecular ovine bone. Part I: mechanical and physical properties. J Bone Joint Surg Br 2000;82(6):910-920. 52. Makoto S, Kenji S, Koichi K, Jun S, Eisuke T, Toshiaki H. Measurements of Mechanical Properties of Cortical Bone Using Nanoindentation Tests. Proceedings of Annual Meeting of Japanese Society for Orthopaedic Biomechanics; 2003; 2003. p. 1-5. 53. Neyt JG, Buckwalter JA, Carroll NC. Use of animal models in musculoskeletal research. The Iowa orthopaedic journal 1998;18:118-123. 54. Newman E, Turner AS, Wark JD. The potential of sheep for the study of osteopenia: current status and comparison with other animal models. Bone 1995;16(4 Suppl):277S-284S. 55. Taylor WR, Ehrig RM, Duda GN, Schell H, Seebeck P, Heller MO. On the influence of soft tissue coverage in the determination of bone kinematics using skin markers. J Orthop Res 2005;23(4):726-734. 56. Taylor WR, Ehrig RM, Heller MO, Schell H, Seebeck P, Duda GN. Tibio-femoral joint contact forces in sheep. J Biomech 2006;39(5):791-798. 57. Ravaglioli A, Krajewski A, Celotti GC, Piancastelli A, Bacchini B, Montanari L, et al. Mineral evolution of bone. Biomaterials 1996;17(6):617-622. 58. Anderson ML, Dhert WJ, de Bruijn JD, Dalmeijer RA, Leenders H, van Blitterswijk CA, et al. Critical size defect in the goat's os ilium. A model to evaluate bone grafts and substitutes. Clin Orthop Relat Res 1999(364):231-239. 59. den Boer FC, Patka P, Bakker FC, Wippermann BW, van Lingen A, Vink GQ, et al. New segmental long bone defect model in sheep: quantitative analysis of healing with dual energy x-ray absorptiometry. J Orthop Res 1999;17(5):654-660. 60. de Kleer V. Development of bone. In: Sumner-Smith G, editor. Bone in Clinical Orthopedics. Philadelphia: W. B. Saunders Co, 2006. p. 1-80. 61. Eitel F, Klapp F, Jacobson W, Schweiberer L. Bone regeneration in animals and in man. A contribution to understanding the relative value of animal experiments to human pathophysiology. Arch Orthop Trauma Surg 1981;99(1):59-64. 62. Dai KR, Xu XL, Tang TT, Zhu ZA, Yu CF, Lou JR, et al. Repairing of goat tibial bone defects with BMP-2 gene-modified tissue-engineered bone. Calcified tissue international 2005;77(1):55-61.

Page 220: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

196

63. Mosekilde L, Weisbrode SE, Safron JA, Stills HF, Jankowsky ML, Ebert DC, et al. Calcium-restricted ovariectomized Sinclair S-1 minipigs: an animal model of osteopenia and trabecular plate perforation. Bone 1993;14(3):379-382. 64. Thorwarth M, Schultze-Mosgau S, Kessler P, Wiltfang J, Schlegel KA. Bone regeneration in osseous defects using a resorbable nanoparticular hydroxyapatite. J Oral Maxillofac Surg 2005;63(11):1626-1633. 65. Macdonald W, Skirving AP, Scull ER. A device for producing experimental fractures. Acta Orthop Scand 1988;59(5):542-544. 66. Skirving AP, Day R, Macdonald W, McLaren R. Carbon fiber reinforced plastic (CFRP) plates versus stainless steel dynamic compression plates in the treatment of fractures of the tibiae in dogs. Clin Orthop Relat Res 1987(224):117-124. 67. Schemitsch EH, Kowalski MJ, Swiontkowski MF. Soft-tissue blood flow following reamed versus unreamed locked intramedullary nailing: a fractured sheep tibia model. Annals of plastic surgery 1996;36(1):70-75. 68. Schemitsch EH, Kowalski MJ, Swiontkowski MF, Senft D. Cortical bone blood flow in reamed and unreamed locked intramedullary nailing: a fractured tibia model in sheep. J Orthop Trauma 1994;8(5):373-382. 69. Tepic S, Remiger AR, Morikawa K, Predieri M, Perren SM. Strength recovery in fractured sheep tibia treated with a plate or an internal fixator: an experimental study with a two-year follow-up. J Orthop Trauma 1997;11(1):14-23. 70. Gilbert JA, Dahners LE, Atkinson MA. The effect of external fixation stiffness on early healing of transverse osteotomies. J Orthop Res 1989;7(3):389-397. 71. Tiedeman JJ, Lippiello L, Connolly JF, Strates BS. Quantitative roentgenographic densitometry for assessing fracture healing. Clin Orthop Relat Res 1990(253):279-286. 72. Markel MD, Wikenheiser MA, Chao EY. A study of fracture callus material properties: relationship to the torsional strength of bone. J Orthop Res 1990;8(6):843-850. 73. Faria ML, Lu Y, Heaney K, Uthamanthil RK, Muir P, Markel MD. Recombinant human bone morphogenetic protein-2 in absorbable collagen sponge enhances bone healing of tibial osteotomies in dogs. Vet Surg 2007;36(2):122-131. 74. Edwards RB, 3rd, Seeherman HJ, Bogdanske JJ, Devitt J, Vanderby R, Jr., Markel MD. Percutaneous injection of recombinant human bone

Page 221: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

197

morphogenetic protein-2 in a calcium phosphate paste accelerates healing of a canine tibial osteotomy. J Bone Joint Surg Am 2004;86-A(7):1425-1438. 75. Wang CJ, Huang HY, Chen HH, Pai CH, Yang KD. Effect of shock wave therapy on acute fractures of the tibia: a study in a dog model. Clin Orthop Relat Res 2001(387):112-118. 76. Hupel TM, Weinberg JA, Aksenov SA, Schemitsch EH. Effect of unreamed, limited reamed, and standard reamed intramedullary nailing on cortical bone porosity and new bone formation. J Orthop Trauma 2001;15(1):18-27. 77. Jain R, Podworny N, Hupel TM, Weinberg J, Schemitsch EH. Influence of plate design on cortical bone perfusion and fracture healing in canine segmental tibial fractures. J Orthop Trauma 1999;13(3):178-186. 78. Nakamura T, Hara Y, Tagawa M, Tamura M, Yuge T, Fukuda H, et al. Recombinant human basic fibroblast growth factor accelerates fracture healing by enhancing callus remodeling in experimental dog tibial fracture. J Bone Miner Res 1998;13(6):942-949. 79. Heitemeyer U, Claes L, Hierholzer G. [The significance of postoperative stability for osseous repair of a multiple fragment fracture. Animal experiment studies]. Unfallchirurg 1990;93(2):49-55. 80. Goodship AE, Watkins PE, Rigby HS, Kenwright J. The role of fixator frame stiffness in the control of fracture healing. An experimental study. J Biomech 1993;26(9):1027-1035. 81. Wallace AL, Makki R, Weiss JB, Hughes SP. Measurement of serum angiogenic factor in devascularized experimental tibial fractures. J Orthop Trauma 1995;9(4):324-332. 82. Augat P, Burger J, Schorlemmer S, Henke T, Peraus M, Claes L. Shear movement at the fracture site delays healing in a diaphyseal fracture model. J Orthop Res 2003;21(6):1011-1017. 83. Augat P, Merk J, Genant HK, Claes L. Quantitative assessment of experimental fracture repair by peripheral computed tomography. Calcified tissue international 1997;60(2):194-199. 84. Epari DR, Schell H, Bail HJ, Duda GN. Instability prolongs the chondral phase during bone healing in sheep. Bone 2006;38(6):864-870. 85. Schell H, Epari DR, Kassi JP, Bragulla H, Bail HJ, Duda GN. The course of bone healing is influenced by the initial shear fixation stability. J Orthop Res 2005;23(5):1022-1028. 86. Hente R, Cordey J, Rahn BA, Maghsudi M, von Gumppenberg S, Perren SM. Fracture healing of the sheep tibia treated using a unilateral

Page 222: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

198

external fixator. Comparison of static and dynamic fixation. Injury 1999;30 Suppl 1:A44-51. 87. Hantes ME, Mavrodontidis AN, Zalavras CG, Karantanas AH, Karachalios T, Malizos KN. Low-intensity transosseous ultrasound accelerates osteotomy healing in a sheep fracture model. J Bone Joint Surg Am 2004;86-A(10):2275-2282. 88. Epari DR, Lienau J, Schell H, Witt F, Duda GN. Pressure, oxygen tension and temperature in the periosteal callus during bone healing-An in vivo study in sheep. Bone 2008. 89. Claes L, Augat P, Schorlemmer S, Konrads C, Ignatius A, Ehrnthaller C. Temporary distraction and compression of a diaphyseal osteotomy accelerates bone healing. J Orthop Res 2008;26(6):772-777. 90. Kuttenberger JJ, Stubinger S, Waibel A, Werner M, Klasing M, Ivanenko M, et al. Computer-guided CO2-laser osteotomy of the sheep tibia: technical prerequisites and first results. Photomedicine and laser surgery 2008;26(2):129-136. 91. Gao TJ, Lindholm TS, Kommonen B, Ragni P, Paronzini A, Lindholm TC, et al. The use of a coral composite implant containing bone morphogenetic protein to repair a segmental tibial defect in sheep. 1997;21(3):194-200. 92. Wefer J, Wefer A, Schratt HE, Thermann H, Wippermann BW. [Healing of autologous cancellous bone transplants and hydroxylapatite ceramics in tibial segment defects. Value of ultrasonic follow up]. Unfallchirurg 2000;103(6):452-461. 93. Bloemers FW, Blokhuis TJ, Patka P, Bakker FC, Wippermann BW, Haarman HJ. Autologous bone versus calcium-phosphate ceramics in treatment of experimental bone defects. Journal of biomedical materials research 2003;66(2):526-531. 94. Meinel L, Zoidis E, Zapf J, Hassa P, Hottiger MO, Auer JA, et al. Localized insulin-like growth factor I delivery to enhance new bone formation. Bone 2003;33(4):660-672. 95. Mastrogiacomo M, Corsi A, Francioso E, Di Comite M, Monetti F, Scaglione S, et al. Reconstruction of extensive long bone defects in sheep using resorbable bioceramics based on silicon stabilized tricalcium phosphate. Tissue Eng 2006;12(5):1261-1273. 96. Maissen O, Eckhardt C, Gogolewski S, Glatt M, Arvinte T, Steiner A, et al. Mechanical and radiological assessment of the influence of rhTGFbeta-3 on bone regeneration in a segmental defect in the ovine tibia: pilot study. J Orthop Res 2006;24(8):1670-1678.

Page 223: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

199

97. Sarkar MR, Augat P, Shefelbine SJ, Schorlemmer S, Huber-Lang M, Claes L, et al. Bone formation in a long bone defect model using a platelet-rich plasma-loaded collagen scaffold. Biomaterials 2006;27(9):1817-1823. 98. Tyllianakis M, Deligianni D, Panagopoulos A, Pappas M, Sourgiadaki E, Mavrilas D, et al. Biomechanical comparison of callus over a locked intramedullary nail in various segmental bone defects in a sheep model. Med Sci Monit 2007;13(5):BR125-130. 99. Teixeira CR, Rahal SC, Volpi RS, Taga R, Cestari TM, Granjeiro JM, et al. Tibial segmental bone defect treated with bone plate and cage filled with either xenogeneic composite or autologous cortical bone graft. An experimental study in sheep. Vet Comp Orthop Traumatol 2007;20(4):269-276. 100. Schneiders W, Reinstorf A, Biewener A, Serra A, Grass R, Kinscher M, et al. In vivo effects of modification of hydroxyapatite/collagen composites with and without chondroitin sulphate on bone remodeling in the sheep tibia. J Orthop Res 2008. 101. Rozen N, Bick T, Bajayo A, Shamian B, Schrift-Tzadok M, Gabet Y, et al. Transplanted blood-derived endothelial progenitor cells (EPC) enhance bridging of sheep tibia critical size defects. Bone 2009. 102. Niemeyer P, Schonberger TS, Hahn J, Kasten P, Fellenberg J, Suedkamp N, et al. Xenogenic transplantation of human mesenchymal stem cells in a critical size defect of the sheep tibia for bone regeneration. Tissue Eng Part A 2009. 103. Claes L, Eckert-Hubner K, Augat P. The fracture gap size influences the local vascularization and tissue differentiation in callus healing. Langenbecks Arch Surg 2003;388(5):316-322. 104. Duda GN, Sollmann M, Sporrer S, Hoffmann JE, Kassi JP, Khodadadyan C, et al. Interfragmentary motion in tibial osteotomies stabilized with ring fixators. Clin Orthop Relat Res 2002(396):163-172. 105. Kaspar K, Matziolis G, Strube P, Senturk U, Dormann S, Bail HJ, et al. A new animal model for bone atrophic nonunion: Fixation by external fixator. J Orthop Res 2008. 106. Hutmacher DW, Schantz JT, Lam CX, Tan KC, Lim TC. State of the art and future directions of scaffold-based bone engineering from a biomaterials perspective. J Tissue Eng Regen Med 2007;1(4):245-260. 107. Kettunen J, Makela A, Miettinen H, Nevalainen T, Heikkila M, Tormala P, et al. Fixation of femoral shaft osteotomy with an intramedullary composite rod: an experimental study on dogs with a two-year follow-up. J Biomater Sci Polym Ed 1999;10(1):33-45.

Page 224: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

200

108. Sinicropi SM, Su BW, Raia FJ, Parisien M, Strauch RJ, Rosenwasser MP. The effects of implant composition on extensor tenosynovitis in a canine distal radius fracture model. J Hand Surg [Am] 2005;30(2):300-307. 109. An YH, Friedman RJ, Powers DL, Draughn RA, Latour RA, Jr. Fixation of osteotomies using bioabsorbable screws in the canine femur. Clin Orthop Relat Res 1998(355):300-311. 110. Aro HT, Chao EY. Bone-healing patterns affected by loading, fracture fragment stability, fracture type, and fracture site compression. Clin Orthop Relat Res 1993(293):8-17. 111. Peter CP, Cook WO, Nunamaker DM, Provost MT, Seedor JG, Rodan GA. Effect of alendronate on fracture healing and bone remodeling in dogs. J Orthop Res 1996;14(1):74-79. 112. Wolf S, Janousek A, Pfeil J, Veith W, Haas F, Duda G, et al. The effects of external mechanical stimulation on the healing of diaphyseal osteotomies fixed by flexible external fixation. Clin Biomech (Bristol, Avon) 1998;13(4-5):359-364. 113. Schell H, Thompson MS, Bail HJ, Hoffmann JE, Schill A, Duda GN, et al. Mechanical induction of critically delayed bone healing in sheep: Radiological and biomechanical results. J Biomech 2008. 114. Curtis MJ, Brown PR, Dick JD, Jinnah RH. Contaminated fractures of the tibia: a comparison of treatment modalities in an animal model. J Orthop Res 1995;13(2):286-295. 115. den Boer FC, Bramer JA, Blokhuis TJ, Van Soest EJ, Jenner JM, Patka P, et al. Effect of recombinant human osteogenic protein-1 on the healing of a freshly closed diaphyseal fracture. Bone 2002;31(1):158-164. 116. Krappinger D, Schubert H, Wenzel V, Rieger M, Stadlbauer KH, Blauth M, et al. A pelvic fracture model for the assessment of treatment options in a laboratory environment. Injury 2007;38(10):1151-1157. 117. Kleinman PL, Zurakowski D, Strauss KJ, Cleveland RH, Perez-Rosello JM, Nichols DP, et al. Detection of simulated inflicted metaphyseal fractures in a fetal pig model: image optimization and dose reduction with computed radiography. Radiology 2008;247(2):381-390. 118. Hill PF, Watkins PE. The prevention of experimental osteomyelitis in a model of gunshot fracture in the pig. Eur J Orthop Surg Traumatol 2001;11:237-241. 119. Bail HJ, Kolbeck S, Krummrey G, Schmidmaier G, Haas NP, Raschke MJ. Systemic application of growth hormone for enhancement of secondary and intramembranous fracture healing. Hormone research 2002;58 Suppl 3:39-42.

Page 225: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

201

120. Allan DG, Russell GG, Moreau MJ, Raso VJ, Budney D. Vertebral end-plate failure in porcine and bovine models of spinal fracture instrumentation. J Orthop Res 1990;8(1):154-156. 121. Russell G, Lavoie G, Evenson R, Moreau M, Budney D, Raso VJ. A reproducible porcine vertebral fracture for biomechanical testing of spinal fixation devices. Clin Orthop Relat Res 1992(284):267-272. 122. Hillier ML, Bell LS. Differentiating human bone from animal bone: a review of histological methods. J Forensic Sci 2007;52(2):249-263. 123. Turner AS. The sheep as a model for osteoporosis in humans. Vet J 2002;163(3):232-239. 124. Dyce K.M. SWO, Wensing C.J.G. Textbook of veterinary Anatomy. 3rd ed. Philadelphia: Saunders 2002. 125. Getty R. Sissons and Grossman's The Anatomy of the Domestic Animals. 5th ed. Philadelphia: W B Saunders, 1975. 126. Reichert JC, Saifzadeh S, Wullschleger ME, Epari DR, Schutz MA, Duda GN, et al. The challenge of establishing preclinical models for segmental bone defect research. Biomaterials 2009;30(12):2149-2163. 127. Gronthos S, Mrozik K, Shi S, Bartold PM. Ovine periodontal ligament stem cells: isolation, characterization, and differentiation potential. Calcified tissue international 2006;79(5):310-317. 128. McCarty RC, Gronthos S, Zannettino AC, Foster BK, Xian CJ. Characterisation and developmental potential of ovine bone marrow derived mesenchymal stem cells. Journal of cellular physiology 2008;219(2):324-333. 129. Sheehan DC, Hrapchak BB. Theory and practice of histotechnology. 2 ed. St. Louis: Mosby, C. V., 1980. 130. Gronthos S, Mankani M, Brahim J, Robey PG, Shi S. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proceedings of the National Academy of Sciences of the United States of America 2000;97(25):13625-13630. 131. Gronthos S, Franklin DM, Leddy HA, Robey PG, Storms RW, Gimble JM. Surface protein characterization of human adipose tissue-derived stromal cells. Journal of cellular physiology 2001;189(1):54-63. 132. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science (New York, NY 1999;284(5411):143-147.

Page 226: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

202

133. Shin H, Zygourakis K, Farach-Carson MC, Yaszemski MJ, Mikos AG. Modulation of differentiation and mineralization of marrow stromal cells cultured on biomimetic hydrogels modified with Arg-Gly-Asp containing peptides. J Biomed Mater Res A 2004;69(3):535-543. 134. Porter JR, Henson A, Popat KC. Biodegradable poly(epsilon-caprolactone) nanowires for bone tissue engineering applications. Biomaterials 2008. 135. Martin I, Wendt D, Heberer M. The role of bioreactors in tissue engineering. Trends in biotechnology 2004;22(2):80-86. 136. Castro-Malaspina H, Gay RE, Resnick G, Kapoor N, Meyers P, Chiarieri D, et al. Characterization of human bone marrow fibroblast colony-forming cells (CFU-F) and their progeny. Blood 1980;56(2):289-301. 137. Friedenstein AJ. Stromal mechanisms of bone marrow: cloning in vitro and retransplantation in vivo. Haematology and blood transfusion 1980;25:19-29. 138. Owen M. Marrow stromal stem cells. Journal of cell science 1988;10:63-76. 139. Gronthos S, Zannettino AC, Hay SJ, Shi S, Graves SE, Kortesidis A, et al. Molecular and cellular characterisation of highly purified stromal stem cells derived from human bone marrow. Journal of cell science 2003;116(Pt 9):1827-1835. 140. Simmons PJ, Torok-Storb B. Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1. Blood 1991;78(1):55-62. 141. Kadiyala S, Young RG, Thiede MA, Bruder SP. Culture expanded canine mesenchymal stem cells possess osteochondrogenic potential in vivo and in vitro. Cell transplantation 1997;6(2):125-134. 142. Martin DR, Cox NR, Hathcock TL, Niemeyer GP, Baker HJ. Isolation and characterization of multipotential mesenchymal stem cells from feline bone marrow. Experimental hematology 2002;30(8):879-886. 143. Yoshimura H, Muneta T, Nimura A, Yokoyama A, Koga H, Sekiya I. Comparison of rat mesenchymal stem cells derived from bone marrow, synovium, periosteum, adipose tissue, and muscle. Cell and tissue research 2007;327(3):449-462. 144. Gronthos S, Brahim J, Li W, Fisher LW, Cherman N, Boyde A, et al. Stem cell properties of human dental pulp stem cells. Journal of dental research 2002;81(8):531-535.

Page 227: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

203

145. Hayflick L. The Limited in Vitro Lifetime of Human Diploid Cell Strains. Experimental cell research 1965;37:614-636. 146. Rheinwald JG, Green H. Serial cultivation of strains of human epidermal keratinocytes: the formation of keratinizing colonies from single cells. Cell 1975;6(3):331-343. 147. Thornton SC, Mueller SN, Levine EM. Human endothelial cells: use of heparin in cloning and long-term serial cultivation. Science (New York, NY 1983;222(4624):623-625. 148. Bruder SP, Jaiswal N, Haynesworth SE. Growth kinetics, self-renewal, and the osteogenic potential of purified human mesenchymal stem cells during extensive subcultivation and following cryopreservation. Journal of cellular biochemistry 1997;64(2):278-294. 149. Digirolamo CM, Stokes D, Colter D, Phinney DG, Class R, Prockop DJ. Propagation and senescence of human marrow stromal cells in culture: a simple colony-forming assay identifies samples with the greatest potential to propagate and differentiate. British journal of haematology 1999;107(2):275-281. 150. Stanford CM, Jacobson PA, Eanes ED, Lembke LA, Midura RJ. Rapidly forming apatitic mineral in an osteoblastic cell line (UMR 106-01 BSP). The Journal of biological chemistry 1995;270(16):9420-9428. 151. Gregory CA, Gunn WG, Peister A, Prockop DJ. An Alizarin red-based assay of mineralization by adherent cells in culture: comparison with cetylpyridinium chloride extraction. Analytical biochemistry 2004;329(1):77-84. 152. Niyibizi C, Eyre DR. Structural characteristics of cross-linking sites in type V collagen of bone. Chain specificities and heterotypic links to type I collagen. European journal of biochemistry / FEBS 1994;224(3):943-950. 153. Viguet-Carrin S, Garnero P, Delmas PD. The role of collagen in bone strength. Osteoporos Int 2006;17(3):319-336. 154. Price PA, Otsuka AA, Poser JW, Kristaponis J, Raman N. Characterization of a gamma-carboxyglutamic acid-containing protein from bone. Proceedings of the National Academy of Sciences of the United States of America 1976;73(5):1447-1451. 155. Boskey AL, Gadaleta S, Gundberg C, Doty SB, Ducy P, Karsenty G. Fourier transform infrared microspectroscopic analysis of bones of osteocalcin-deficient mice provides insight into the function of osteocalcin. Bone 1998;23(3):187-196. 156. Choi ST, Kim JH, Kang EJ, Lee SW, Park MC, Park YB, et al. Osteopontin might be involved in bone remodelling rather than in

Page 228: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

204

inflammation in ankylosing spondylitis. Rheumatology (Oxford, England) 2008;47(12):1775-1779. 157. Wagner HD, Weiner S. On the relationship between the microstructure of bone and its mechanical stiffness. J Biomech 1992;25(11):1311-1320. 158. Datta N, Pham QP, Sharma U, Sikavitsas VI, Jansen JA, Mikos AG. In vitro generated extracellular matrix and fluid shear stress synergistically enhance 3D osteoblastic differentiation. Proceedings of the National Academy of Sciences of the United States of America 2006;103(8):2488-2493. 159. Knothe Tate ML, Falls TD, McBride SH, Atit R, Knothe UR. Mechanical modulation of osteochondroprogenitor cell fate. The international journal of biochemistry & cell biology 2008;40(12):2720-2738. 160. Zhou Y, Chen F, Ho ST, Woodruff MA, Lim TM, Hutmacher DW. Combined marrow stromal cell-sheet techniques and high-strength biodegradable composite scaffolds for engineered functional bone grafts. Biomaterials 2007;28(5):814-824. 161. Abdallah BM, Kassem M. The use of mesenchymal (skeletal) stem cells for treatment of degenerative diseases: current status and future perspectives. Journal of cellular physiology 2009;218(1):9-12. 162. Griffith LG, Swartz MA. Capturing complex 3D tissue physiology in vitro. Nature reviews 2006;7(3):211-224. 163. Hutmacher DW, Cool S. Concepts of scaffold-based tissue engineering--the rationale to use solid free-form fabrication techniques. Journal of cellular and molecular medicine 2007;11(4):654-669. 164. Arthur A, Zannettino A, Gronthos S. The therapeutic applications of multipotential mesenchymal/stromal stem cells in skeletal tissue repair. Journal of cellular physiology 2009;218(2):237-245. 165. Biggs MJ, Richards RG, Gadegaard N, Wilkinson CD, Oreffo RO, Dalby MJ. The use of nanoscale topography to modulate the dynamics of adhesion formation in primary osteoblasts and ERK/MAPK signalling in STRO-1+ enriched skeletal stem cells. Biomaterials 2009;30(28):5094-5103. 166. Santos MI, Unger RE, Sousa RA, Reis RL, Kirkpatrick CJ. Crosstalk between osteoblasts and endothelial cells co-cultured on a polycaprolactone-starch scaffold and the in vitro development of vascularization. Biomaterials 2009;30(26):4407-4415. 167. Ripamonti U, Van Den Heever B, Sampath TK, Tucker MM, Rueger DC, Reddi AH. Complete regeneration of bone in the baboon by recombinant

Page 229: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

205

human osteogenic protein-1 (hOP-1, bone morphogenetic protein-7). Growth factors (Chur, Switzerland) 1996;13(3-4):273-289,color plates III-VIII,pre bk. 168. Ramoshebi LN, Ripamonti U. Osteogenic protein-1, a bone morphogenetic protein, induces angiogenesis in the chick chorioallantoic membrane and synergizes with basic fibroblast growth factor and transforming growth factor-beta1. The Anatomical record 2000;259(1):97-107. 169. Schumann D, Ekaputra AK, Lam CX, Hutmacher DW. Biomaterials/scaffolds. Design of bioactive, multiphasic PCL/collagen type I and type II-PCL-TCP/collagen composite scaffolds for functional tissue engineering of osteochondral repair tissue by using electrospinning and FDM techniques. Methods in molecular medicine 2007;140:101-124. 170. Miura M, Miura Y, Padilla-Nash HM, Molinolo AA, Fu B, Patel V, et al. Accumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation. Stem cells (Dayton, Ohio) 2006;24(4):1095-1103. 171. Noth U, Rackwitz L, Heymer A, Weber M, Baumann B, Steinert A, et al. Chondrogenic differentiation of human mesenchymal stem cells in collagen type I hydrogels. J Biomed Mater Res A 2007;83(3):626-635. 172. Denker AE, Haas AR, Nicoll SB, Tuan RS. Chondrogenic differentiation of murine C3H10T1/2 multipotential mesenchymal cells: I. Stimulation by bone morphogenetic protein-2 in high-density micromass cultures. Differentiation; research in biological diversity 1999;64(2):67-76. 173. Haas AR, Tuan RS. Chondrogenic differentiation of murine C3H10T1/2 multipotential mesenchymal cells: II. Stimulation by bone morphogenetic protein-2 requires modulation of N-cadherin expression and function. Differentiation; research in biological diversity 1999;64(2):77-89. 174. Tortelli F, Tasso R, Loiacono F, Cancedda R. The development of tissue-engineered bone of different origin through endochondral and intramembranous ossification following the implantation of mesenchymal stem cells and osteoblasts in a murine model. Biomaterials;31(2):242-249. 175. Erlebacher A, Derynck R. Increased expression of TGF-beta 2 in osteoblasts results in an osteoporosis-like phenotype. The Journal of cell biology 1996;132(1-2):195-210. 176. Tatapudy S, Bruening S, Gleason G, Toth M. Validation and use of a computer-assisted counting procedure to quantify BrdU-labeled proliferating cells in the early postnatal mouse hippocampus. Journal of neuroscience methods 2008;172(2):173-177.

Page 230: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

206

177. Roschger P, Fratzl P, Eschberger J, Klaushofer K. Validation of quantitative backscattered electron imaging for the measurement of mineral density distribution in human bone biopsies. Bone 1998;23(4):319-326. 178. Fratzl-Zelman N, Roschger P, Gourrier A, Weber M, Misof BM, Loveridge N, et al. Combination of nanoindentation and quantitative backscattered electron imaging revealed altered bone material properties associated with femoral neck fragility. Calcified tissue international 2009;85(4):335-343. 179. Chung ES, Saban DR, Chauhan SK, Dana R. Regulation of blood vessel versus lymphatic vessel growth in the cornea. Investigative ophthalmology & visual science 2009;50(4):1613-1618. 180. Wlodarski KH, Wlodarski PK, Galus R. Senescence of osteogenic cells. Review. Ortopedia, traumatologia, rehabilitacja 2007;9(1):63-67. 181. Hauschka PV, Lian JB, Cole DE, Gundberg CM. Osteocalcin and matrix Gla protein: vitamin K-dependent proteins in bone. Physiological reviews 1989;69(3):990-1047. 182. Hauschka PV, Wians FH, Jr. Osteocalcin-hydroxyapatite interaction in the extracellular organic matrix of bone. The Anatomical record 1989;224(2):180-188. 183. Yeh LC, Adamo ML, Olson MS, Lee JC. Osteogenic protein-1 and insulin-like growth factor I synergistically stimulate rat osteoblastic cell differentiation and proliferation. Endocrinology 1997;138(10):4181-4190. 184. Yeh LC, Tsai AD, Lee JC. Osteogenic protein-1 (OP-1, BMP-7) induces osteoblastic cell differentiation of the pluripotent mesenchymal cell line C2C12. Journal of cellular biochemistry 2002;87(3):292-304. 185. Pluhar GE, Turner AS, Pierce AR, Toth CA, Wheeler DL. A comparison of two biomaterial carriers for osteogenic protein-1 (BMP-7) in an ovine critical defect model. J Bone Joint Surg Br 2006;88(7):960-966. 186. Ristiniemi J, Flinkkila T, Hyvonen P, Lakovaara M, Pakarinen H, Jalovaara P. RhBMP-7 accelerates the healing in distal tibial fractures treated by external fixation. J Bone Joint Surg Br 2007;89(2):265-272. 187. Paul S, Lee JC, Yeh LC. A comparative study on BMP-induced osteoclastogenesis and osteoblastogenesis in primary cultures of adult rat bone marrow cells. Growth factors (Chur, Switzerland) 2009;27(2):121-131. 188. Cowan CM, Aghaloo T, Chou YF, Walder B, Zhang X, Soo C, et al. MicroCT evaluation of three-dimensional mineralization in response to BMP-2 doses in vitro and in critical sized rat calvarial defects. Tissue Eng 2007;13(3):501-512.

Page 231: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

207

189. Liu SB, Hu PZ, Hou Y, Li P, Cao W, Tian Q. Recombinant human bone morphogenetic protein-2 promotes the proliferation of mesenchymal stem cells in vivo and in vitro. Chinese medical journal 2009;122(7):839-843. 190. Young S, Patel ZS, Kretlow JD, Murphy MB, Mountziaris PM, Baggett LS, et al. Dose effect of dual delivery of vascular endothelial growth factor and bone morphogenetic protein-2 on bone regeneration in a rat critical-size defect model. Tissue Eng Part A 2009;15(9):2347-2362. 191. Ljusberg J, Ek-Rylander B, Andersson G. Tartrate-resistant purple acid phosphatase is synthesized as a latent proenzyme and activated by cysteine proteinases. The Biochemical journal 1999;343 Pt 1:63-69. 192. Ljusberg J, Wang Y, Lang P, Norgard M, Dodds R, Hultenby K, et al. Proteolytic excision of a repressive loop domain in tartrate-resistant acid phosphatase by cathepsin K in osteoclasts. The Journal of biological chemistry 2005;280(31):28370-28381. 193. Minkin C. Bone acid phosphatase: tartrate-resistant acid phosphatase as a marker of osteoclast function. Calcified tissue international 1982;34(3):285-290. 194. Hayman AR, Jones SJ, Boyde A, Foster D, Colledge WH, Carlton MB, et al. Mice lacking tartrate-resistant acid phosphatase (Acp 5) have disrupted endochondral ossification and mild osteopetrosis. Development (Cambridge, England) 1996;122(10):3151-3162. 195. Angel NZ, Walsh N, Forwood MR, Ostrowski MC, Cassady AI, Hume DA. Transgenic mice overexpressing tartrate-resistant acid phosphatase exhibit an increased rate of bone turnover. J Bone Miner Res 2000;15(1):103-110. 196. Kaneko H, Arakawa T, Mano H, Kaneda T, Ogasawara A, Nakagawa M, et al. Direct stimulation of osteoclastic bone resorption by bone morphogenetic protein (BMP)-2 and expression of BMP receptors in mature osteoclasts. Bone 2000;27(4):479-486. 197. Hentunen TA, Lakkakorpi PT, Tuukkanen J, Lehenkari PP, Sampath TK, Vaananen HK. Effects of recombinant human osteogenic protein-1 on the differentiation of osteoclast-like cells and bone resorption. Biochemical and biophysical research communications 1995;209(2):433-443. 198. Deckers MM, van Bezooijen RL, van der Horst G, Hoogendam J, van Der Bent C, Papapoulos SE, et al. Bone morphogenetic proteins stimulate angiogenesis through osteoblast-derived vascular endothelial growth factor A. Endocrinology 2002;143(4):1545-1553. 199. Valdimarsdottir G, Goumans MJ, Rosendahl A, Brugman M, Itoh S, Lebrin F, et al. Stimulation of Id1 expression by bone morphogenetic protein

Page 232: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

208

is sufficient and necessary for bone morphogenetic protein-induced activation of endothelial cells. Circulation 2002;106(17):2263-2270. 200. ten Dijke P, Fu J, Schaap P, Roelen BA. Signal transduction of bone morphogenetic proteins in osteoblast differentiation. J Bone Joint Surg Am 2003;85-A Suppl 3:34-38. 201. Chang SC, Tai CL, Chung HY, Lin TM, Jeng LB. Bone marrow mesenchymal stem cells form ectopic woven bone in vivo through endochondral bone formation. Artificial organs 2009;33(4):301-308. 202. Bastow ER, Byers S, Golub SB, Clarkin CE, Pitsillides AA, Fosang AJ. Hyaluronan synthesis and degradation in cartilage and bone. Cell Mol Life Sci 2008;65(3):395-413. 203. Hollister SJ. Scaffold engineering: a bridge to where? Biofabrication 2009;1(1):1-14. 204. Ratcliffe A. Industrial approaches in tissue engineering. In: Mao J, Vunjak-Novakovic G, Mikos AG, Atala A, editors. Translational Approaches in Tissue Engineering and Regenerative Medicine. Boston: Artech House Publishers, 2008. p. 463-472. 205. Konrad G, Sudkamp N. [Extra-articular proximal tibial fracture]. Der Chirurg; Zeitschrift fur alle Gebiete der operativen Medizen 2007;78(2):161-171; quiz 172-163. 206. Klein MP, Rahn BA, Frigg R, Kessler S, Perren SM. Reaming versus non-reaming in medullary nailing: interference with cortical circulation of the canine tibia. Arch Orthop Trauma Surg 1990;109(6):314-316. 207. Mueller CA, Rahn BA. Intramedullary pressure increase and increase in cortical temperature during reaming of the femoral medullary cavity: the effect of draining the medullary contents before reaming. The Journal of trauma 2003;55(3):495-503; discussion 503. 208. Wenda K, Ritter G, Degreif J, Rudigier J. [Pathogenesis of pulmonary complications following intramedullary nailing osteosyntheses]. Unfallchirurg 1988;91(9):432-435. 209. Pape HC, Hildebrand F, Krettek C, Green J, Giannoudis PV. Experimental background--review of animal studies. Injury 2006;37 Suppl 4:S25-38. 210. Baumgart F, Kohler G, Ochsner PE. The physics of heat generation during reaming of the medullary cavity. Injury 1998;29 Suppl 2:B11-25. 211. Giannoudis PV, Snowden S, Matthews SJ, Smye SW, Smith RM. Temperature rise during reamed tibial nailing. Clin Orthop Relat Res 2002(395):255-261.

Page 233: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

209

212. Ochsner PE, Baumgart F, Kohler G. Heat-induced segmental necrosis after reaming of one humeral and two tibial fractures with a narrow medullary canal. Injury 1998;29 Suppl 2:B1-10. 213. Seekamp A, Lehmann U, Pizanis A, Pohlemann T. [New aspects for minimally invasive interventions in orthopedic trauma surgery]. Der Chirurg; Zeitschrift fur alle Gebiete der operativen Medizen 2003;74(4):301-309. 214. Ekeland A, Thoresen BO, Alho A, Stromsoe K, Folleras G, Haukebo A. Interlocking intramedullary nailing in the treatment of tibial fractures. A report of 45 cases. Clin Orthop Relat Res 1988(231):205-215. 215. Hansen M, Mehler D, Rommens PM. Das Verhalten winkelstabiler Implantatsysteme an der proximalen Tibia. Verlag Hans Huber, 2004. 216. Gautier E, Ganz R. [The biological plate osteosynthesis]. Zentralblatt fur Chirurgie 1994;119(8):564-572. 217. Krettek C, Schandelmaier P, Tscherne H. [New developments in stabilization of dia- and metaphyseal fractures of long tubular bones]. Der Orthopade 1997;26(5):408-421. 218. Gautier E, Perren SM, Cordey J. Strain distribution in plated and unplated sheep tibia an in vivo experiment. Injury 2000;31 Suppl 3:C37-44. 219. Hertel R, Eijer H, Meisser A, Hauke C, Perren SM. Biomechanical and biological considerations relating to the clinical use of the Point Contact-Fixator--evaluation of the device handling test in the treatment of diaphyseal fractures of the radius and/or ulna. Injury 2001;32 Suppl 2:B10-14. 220. Beardi J, Hessmann M, Hansen M, Rommens PM. Operative treatment of tibial shaft fractures: a comparison of different methods of primary stabilisation. Arch Orthop Trauma Surg 2008;128(7):709-715. 221. Weiss RJ, Montgomery SM, Ehlin A, Al Dabbagh Z, Stark A, Jansson KA. Decreasing incidence of tibial shaft fractures between 1998 and 2004: information based on 10,627 Swedish inpatients. Acta Orthop 2008;79(4):526-533. 222. Connolly JF, Guse R, Tiedeman J, Dehne R. Autologous marrow injection as a substitute for operative grafting of tibial nonunions. Clin Orthop Relat Res 1991(266):259-270. 223. Alberts KA, Loohagen G, Einarsdottir H. Open tibial fractures: faster union after unreamed nailing than external fixation. Injury 1999;30(8):519-523.

Page 234: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

210

224. Darder-Garcia A, Darder-Prats A, Gomar-Sancho F. Nonreamed flexible locked intramedullary nailing in tibial open fractures. Clin Orthop Relat Res 1998(350):97-104. 225. Singer RW, Kellam JF. Open tibial diaphyseal fractures. Results of unreamed locked intramedullary nailing. Clin Orthop Relat Res 1995(315):114-118. 226. Whittle AP, Russell TA, Taylor JC, Lavelle DG. Treatment of open fractures of the tibial shaft with the use of interlocking nailing without reaming. J Bone Joint Surg Am 1992;74(8):1162-1171. 227. Swiontkowski MF, Senf D, Taylor S. Plate desgn has an effect on cortical bone perfusion. Trans 37th Orthop Res Soc, 1991. p. 387. 228. Korvick DL, Monville JD, Pijanowski GJ, Phillips JW. The effects of screw removal on bone strain in an idealized plated bone model. Vet Surg 1988;17(3):111-116. 229. Auer JA, Goodship A, Arnoczky S, Pearce S, Price J, Claes L, et al. Refining animal models in fracture research: seeking consensus in optimising both animal welfare and scientific validity for appropriate biomedical use. BMC Musculoskelet Disord 2007;8:72. 230. Kneser U, Polykandriotis E, Ohnolz J, Heidner K, Grabinger L, Euler S, et al. Engineering of vascularized transplantable bone tissues: induction of axial vascularization in an osteoconductive matrix using an arteriovenous loop. Tissue Eng 2006;12(7):1721-1731. 231. Mikos AG, Herring SW, Ochareon P, Elisseeff J, Lu HH, Kandel R, et al. Engineering complex tissues. Tissue Eng 2006;12(12):3307-3339. 232. Hing KA, Wilson LF, Buckland T. Comparative performance of three ceramic bone graft substitutes. Spine J 2007;7(4):475-490. 233. Schliephake H, Neukam FW, Hutmacher D, Wustenfeld H. Experimental transplantation of hydroxylapatite-bone composite grafts. J Oral Maxillofac Surg 1995;53(1):46-51; discussion 52. 234. Becker S, Maissen O, Ponomarev I, Stoll T, Rahn B, Wilke I. Osteopromotion by a beta-tricalcium phosphate/bone marrow hybrid implant for use in spine surgery. Spine 2006;31(1):11-17. 235. Leiggener CS, Curtis R, Muller AA, Pfluger D, Gogolewski S, Rahn BA. Influence of copolymer composition of polylactide implants on cranial bone regeneration. Biomaterials 2006;27(2):202-207. 236. Hutmacher DW. Scaffolds in tissue engineering bone and cartilage. Biomaterials 2000;21(24):2529-2543.

Page 235: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

211

237. Hutmacher DW, Garcia AJ. Scaffold-based bone engineering by using genetically modified cells. Gene 2005;347(1):1-10. 238. Abbah SA, Lam CX, Hutmacher DW, Goh JC, Wong HK. Biological performance of a polycaprolactone-based scaffold used as fusion cage device in a large animal model of spinal reconstructive surgery. Biomaterials 2009;30(28):5086-5093. 239. Blair HC, Zaidi M, Schlesinger PH. Mechanisms balancing skeletal matrix synthesis and degradation. The Biochemical journal 2002;364(Pt 2):329-341. 240. Blumenthal NM, Koh-Kunst G, Alves ME, Miranda D, Sorensen RG, Wozney JM, et al. Effect of surgical implantation of recombinant human bone morphogenetic protein-2 in a bioabsorbable collagen sponge or calcium phosphate putty carrier in intrabony periodontal defects in the baboon. Journal of periodontology 2002;73(12):1494-1506. 241. Cheng H, Jiang W, Phillips FM, Haydon RC, Peng Y, Zhou L, et al. Osteogenic activity of the fourteen types of human bone morphogenetic proteins (BMPs). J Bone Joint Surg Am 2003;85-A(8):1544-1552. 242. Wu XB, Li Y, Schneider A, Yu W, Rajendren G, Iqbal J, et al. Impaired osteoblastic differentiation, reduced bone formation, and severe osteoporosis in noggin-overexpressing mice. The Journal of clinical investigation 2003;112(6):924-934. 243. Mikic B, van der Meulen MC, Kingsley DM, Carter DR. Long bone geometry and strength in adult BMP-5 deficient mice. Bone 1995;16(4):445-454. 244. Sykaras N, Opperman LA. Bone morphogenetic proteins (BMPs): how do they function and what can they offer the clinician? Journal of oral science 2003;45(2):57-73. 245. Janssens K, ten Dijke P, Janssens S, Van Hul W. Transforming growth factor-beta1 to the bone. Endocrine reviews 2005;26(6):743-774. 246. Linkhart TA, Mohan S, Baylink DJ. Growth factors for bone growth and repair: IGF, TGF beta and BMP. Bone 1996;19(1 Suppl):1S-12S. 247. Sawyer AA, Song SJ, Susanto E, Chuan P, Lam CX, Woodruff MA, et al. The stimulation of healing within a rat calvarial defect by mPCL-TCP/collagen scaffolds loaded with rhBMP-2. Biomaterials 2009;30(13):2479-2488. 248. Ugural AC, Fenster SK. Advanced Strength and Applied Elasticity. 3 ed. Englewood Cliffs, NJ: Prentice-Hall Inc., 1995.

Page 236: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

212

249. Movat HZ. Demonstration of all connective tissue elements in a single section; pentachrome stains. A M A 1955;60(3):289-295. 250. Carter DR, Hayes WC. Bone compressive strength: the influence of density and strain rate. Science (New York, NY 1976;194(4270):1174-1176. 251. Yetkinler DN, McClellan RT, Reindel ES, Carter D, Poser RD. Biomechanical comparison of conventional open reduction and internal fixation versus calcium phosphate cement fixation of a central depressed tibial plateau fracture. J Orthop Trauma 2001;15(3):197-206. 252. Sanger C, Soto A, Mussa F, Sanzo M, Sardo L, Donati PA, et al. Maximizing results in craniofacial surgery with bioresorbable fixation devices. The Journal of craniofacial surgery 2007;18(4):926-930. 253. Hench LL, Polak JM. Third-generation biomedical materials. Science (New York, NY 2002;295(5557):1014-1017. 254. Hollister SJ. Porous scaffold design for tissue engineering. Nature materials 2005;4(7):518-524. 255. Lohmann H, Grass G, Rangger C, Mathiak G. Economic impact of cancellous bone grafting in trauma surgery. Arch Orthop Trauma Surg 2007;127(5):345-348. 256. Asahina I, Sampath TK, Nishimura I, Hauschka PV. Human osteogenic protein-1 induces both chondroblastic and osteoblastic differentiation of osteoprogenitor cells derived from newborn rat calvaria. The Journal of cell biology 1993;123(4):921-933. 257. Friedlaender GE, Perry CR, Cole JD, Cook SD, Cierny G, Muschler GF, et al. Osteogenic protein-1 (bone morphogenetic protein-7) in the treatment of tibial nonunions. J Bone Joint Surg Am 2001;83-A Suppl 1(Pt 2):S151-158. 258. Uludag H, Gao T, Porter TJ, Friess W, Wozney JM. Delivery systems for BMPs: factors contributing to protein retention at an application site. J Bone Joint Surg Am 2001;83-A Suppl 1(Pt 2):S128-135. 259. Erickson BP, Pierce AR, Simpson AK, Nash J, Grauer JN. 125I-labeled OP-1 is locally retained in a rabbit lumbar fusion model. Clin Orthop Relat Res 2008;466(1):210-215. 260. Uludag H, D'Augusta D, Golden J, Li J, Timony G, Riedel R, et al. Implantation of recombinant human bone morphogenetic proteins with biomaterial carriers: A correlation between protein pharmacokinetics and osteoinduction in the rat ectopic model. J Biomed Mater Res 2000;50(2):227-238.

Page 237: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

213

261. Winn SR, Uludag H, Hollinger JO. Carrier systems for bone morphogenetic proteins. Clin Orthop Relat Res 1999(367 Suppl):S95-106. 262. Obert L, Deschaseaux F, Garbuio P. Critical analysis and efficacy of BMPs in long bones non-union. Injury 2005;36 Suppl 3:S38-42. 263. Takigami H, Kumagai K, Latson L, Togawa D, Bauer T, Powell K, et al. Bone formation following OP-1 implantation is improved by addition of autogenous bone marrow cells in a canine femur defect model. J Orthop Res 2007;25(10):1333-1342. 264. Bostrom M, Lane JM, Tomin E, Browne M, Berberian W, Turek T, et al. Use of bone morphogenetic protein-2 in the rabbit ulnar nonunion model. Clin Orthop Relat Res 1996(327):272-282. 265. Cook SD, Wolfe MW, Salkeld SL, Rueger DC. Effect of recombinant human osteogenic protein-1 on healing of segmental defects in non-human primates. J Bone Joint Surg Am 1995;77(5):734-750. 266. Geesink RG, Hoefnagels NH, Bulstra SK. Osteogenic activity of OP-1 bone morphogenetic protein (BMP-7) in a human fibular defect. J Bone Joint Surg Br 1999;81(4):710-718. 267. Haidar ZS, Hamdy RC, Tabrizian M. Delivery of recombinant bone morphogenetic proteins for bone regeneration and repair. Part A: Current challenges in BMP delivery. Biotechnology letters 2009;31(12):1817-1824. 268. Einhorn TA. Clinical applications of recombinant human BMPs: early experience and future development. J Bone Joint Surg Am 2003;85-A Suppl 3:82-88. 269. Dimitriou R, Tsiridis E, Giannoudis PV. Current concepts of molecular aspects of bone healing. Injury 2005;36(12):1392-1404. 270. Gerstenfeld LC, Alkhiary YM, Krall EA, Nicholls FH, Stapleton SN, Fitch JL, et al. Three-dimensional reconstruction of fracture callus morphogenesis. J Histochem Cytochem 2006;54(11):1215-1228. 271. Gerstenfeld LC, Cullinane DM, Barnes GL, Graves DT, Einhorn TA. Fracture healing as a post-natal developmental process: molecular, spatial, and temporal aspects of its regulation. Journal of cellular biochemistry 2003;88(5):873-884. 272. Le AX, Miclau T, Hu D, Helms JA. Molecular aspects of healing in stabilized and non-stabilized fractures. J Orthop Res 2001;19(1):78-84. 273. Cho TJ, Gerstenfeld LC, Einhorn TA. Differential temporal expression of members of the transforming growth factor beta superfamily during murine fracture healing. J Bone Miner Res 2002;17(3):513-520.

Page 238: Tissue Engineering Bone - Reconstruction of Critical Sized ...eprints.qut.edu.au/48080/1/Johannes_Reichert_Thesis.pdf · Tissue Engineering Bone - Reconstruction of Critical Sized

214

274. Yeh LC, Unda R, Lee JC. Osteogenic protein-1 differentially regulates the mRNA expression of bone morphogenetic proteins and their receptors in primary cultures of osteoblasts. Journal of cellular physiology 2000;185(1):87-97. 275. White AP, Weinstein MA, Patel T, Horowitz MC, Friedlaender GE. The 2002 Marshall Urist Young Investigator Award Paper. Lumbar arthrodesis gene expression: a comparison of autograft with osteogenic protein-1. Clin Orthop Relat Res 2004(429):330-337. 276. Onishi T, Ishidou Y, Nagamine T, Yone K, Imamura T, Kato M, et al. Distinct and overlapping patterns of localization of bone morphogenetic protein (BMP) family members and a BMP type II receptor during fracture healing in rats. Bone 1998;22(6):605-612. 277. Sampath TK, Maliakal JC, Hauschka PV, Jones WK, Sasak H, Tucker RF, et al. Recombinant human osteogenic protein-1 (hOP-1) induces new bone formation in vivo with a specific activity comparable with natural bovine osteogenic protein and stimulates osteoblast proliferation and differentiation in vitro. The Journal of biological chemistry 1992;267(28):20352-20362.